1
|
Gao RR, Han C, Sui GY, Chen YB, Zhou L, Hu HZ, Wang YC, Liu Y, Li W. Huangqi and Danshen improve the chronic nephrotoxicity of cyclosporin A by regulating lipid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156582. [PMID: 40056636 DOI: 10.1016/j.phymed.2025.156582] [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: 10/23/2024] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND The clinical application of cyclosporine A (CsA) is limited due to nephrotoxicity. Lipid metabolism disorders play important roles in renal injury, but their role in CsA nephrotoxicity is not yet clear. Huangqi (Astragalus mongholicus Bunge) and Danshen (Salvia miltiorrhiza Bunge) (HD) play roles in ameliorating the nephrotoxicity of CsA, but their mechanisms still need to be fully clarified. OBJECTIVE This study innovatively aimed to analyse the coexpression of renal proteins and serum metabolites for the identification of key pathways and targets. This study provides novel insight into the mechanism by which HD ameliorates CsA-induced nephrotoxicity. METHODS We utilized HD to intervene in both in vivo and in vitro nephrotoxicity models induced by CsA. For the in vivo experiments, we constructed a coexpression network of renal proteins and serum metabolites, which was used to screen for key pathways. To validate these findings, we knocked down key proteins in vivo. For the in vitro studies, we employed MTT, Transwell, flow cytometry, and immunofluorescence assays to monitor the epithelial-mesenchymal transition (EMT) of HK-2 cells. Additionally, we used electron microscopy and Seahorse assays to examine the effects of HD on mitochondrial structure and function. Furthermore, we overexpressed Ppara to further confirm the mechanism by which HD improves renal function. RESULTS HD can improve renal pathological damage and function; regulate blood lipids, inflammation and oxidative stress indicators; and reduce apoptosis in renal tissues. Joint protein and metabolomics analyses revealed that two lipid metabolism-related pathways (the PPAR signalling pathway and linoleic acid metabolism pathway) were coenriched, involving six differential proteins (Cyp2e1, Cyp4a10, Gk, Lpl, Ppara, and Pck1) and two differentially abundant metabolites (alpha-Dimorphecolic acid and 12,13-EpOME). Western blot was used to verify differentially expressed proteins. HD improved mitochondrial damage and lipid accumulation, as demonstrated by transmission electron microscopy (TEM) analysis and Oil Red O staining. Knockdown of the key protein Ppara affected the expression of ACOX1 and exacerbated RF. In vitro verification demonstrated that HD significantly inhibited CsA-induced EMT in HK-2 cells and improved mitochondrial structure and function. Ppara overexpression promoted HD-mediated regulation of mitochondrial function, reduced apoptosis, and improved HK-2 RF. CONCLUSION HD can ameliorate CsA nephrotoxicity through renal protein-serum metabolism coexpression, the PPAR signalling pathway, and linoleic acid metabolism. HD-induced upregulation of Ppara to regulate lipid metabolism, improve mitochondrial function and reduce apoptosis are important mechanisms. The Ppara/ACOX1/TGF-β1 axis may play an important role in this process. These findings offer potential targets for the future development of therapeutic strategies and novel drugs.
Collapse
Affiliation(s)
- Ran-Ran Gao
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, PR China
| | - Cong Han
- Nephropathy Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, PR China.
| | - Gui-Yuan Sui
- Nephropathy Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, PR China
| | - Yi-Bing Chen
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, PR China
| | - Le Zhou
- Nephropathy Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, PR China
| | - Hong-Zhen Hu
- Nephropathy Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, PR China
| | - Yi-Chuan Wang
- Nephropathy Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, PR China
| | - Yao Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, PR China
| | - Wei Li
- Nephropathy Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, PR China.
| |
Collapse
|
2
|
Yu W, Haoyu Y, Ling Z, Xing H, Pengfei X, Anzhu W, Lili Z, Linhua Z. Targeting lipid metabolic reprogramming to alleviate diabetic kidney disease: molecular insights and therapeutic strategies. Front Immunol 2025; 16:1549484. [PMID: 40352935 PMCID: PMC12061959 DOI: 10.3389/fimmu.2025.1549484] [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: 12/21/2024] [Accepted: 03/14/2025] [Indexed: 05/14/2025] Open
Abstract
Diabetic kidney disease (DKD) is one of the major complications of diabetes, and its pathological progression is closely associated with lipid metabolic reprogramming. Under diabetic conditions, renal cells undergo significant lipid metabolic abnormalities, including increased lipid uptake, impaired fatty acid oxidation, disrupted cholesterol efflux, and enhanced lipid catabolism, as adaptive responses to metabolic stress. These changes result in the accumulation of lipids such as free fatty acids, diacylglycerol, and ceramides, leading to lipotoxicity that triggers inflammation and fibrosis. Hypoxia in the DKD microenvironment suppresses fatty acid oxidation and promotes lipid synthesis through the HIF-1α pathway, while chronic inflammation exacerbates lipid metabolic disturbances via inflammatory cytokines, inflammasomes, and macrophage polarization. Targeting lipid metabolism represents a promising therapeutic strategy for alleviating DKD; however, further clinical translational studies are warranted to validate the efficacy and safety of these approaches.
Collapse
Affiliation(s)
- Wei Yu
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Haoyu
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhou Ling
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Hang Xing
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Xie Pengfei
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Wang Anzhu
- Chinese-Japanese Friendship Hospital, Beijing, China
| | - Zhang Lili
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhao Linhua
- Department of Endocrinology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
3
|
Kolligundla LP, Sullivan KM, Mukhi D, Andrade-Silva M, Liu H, Guan Y, Gu X, Wu J, Doke T, Hirohama D, Guarnieri P, Hill J, Pullen SS, Kuo J, Inamoto M, Susztak K. Glutathione-specific gamma-glutamylcyclotransferase 1 ( CHAC1) increases kidney disease risk by modulating ferroptosis. Sci Transl Med 2025; 17:eadn3079. [PMID: 40267214 DOI: 10.1126/scitranslmed.adn3079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 08/20/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Genome-wide association studies (GWASs) have identified more than 1000 loci where genetic variants correlate with kidney function. However, the specific genes, cell types, and mechanisms influenced by these genetic variants remain largely uncharted. Here, we identified glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) on chromosome 15 as affected by GWAS variants by analyzing human kidney gene expression and methylation information. Both CHAC1 RNA and protein were expressed in the loop of Henle region in mouse and human kidneys, and CHAC1 expression was higher in patients carrying disease risk variants. Using CRISPR technology, we created mice with a single functional copy of the Chac1 gene (Chac1+/-) that displayed no baseline phenotypic alterations in kidney structure or function. These mice demonstrated resilience to kidney disease in multiple models, including folic acid-induced nephropathy, adenine-induced chronic kidney disease, and uninephrectomy-streptozotocin-induced diabetic nephropathy. We further showed that CHAC1 plays a critical role in degrading the cellular antioxidant glutathione. Tubule cells isolated from Chac1+/- mice showed increased glutathione, decreased lipid peroxidation, improved cell viability, and protection against ferroptosis. Expression of ferroptosis-associated genes was also lower in mice with only one copy of Chac1. Higher CHAC1 protein also correlated with ferroptosis-related protein abundance in kidney biopsies from patients with kidney disease. This study positions CHAC1 as an important mediator of kidney disease that influences glutathione concentrations and ferroptosis, suggesting potential avenues to explore for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Lakshmi P Kolligundla
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Katie M Sullivan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Pediatrics, Medical College of Wisconsin Pediatric Nephrology, Milwaukee, WI 53226, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Magaiver Andrade-Silva
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Hongbo Liu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Yuting Guan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Xiangchen Gu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Junnan Wu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Daigoro Hirohama
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Paolo Guarnieri
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jon Hill
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jay Kuo
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | | | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| |
Collapse
|
4
|
Hall AM. Protein handling in kidney tubules. Nat Rev Nephrol 2025; 21:241-252. [PMID: 39762367 DOI: 10.1038/s41581-024-00914-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 03/23/2025]
Abstract
The kidney proximal tubule reabsorbs and degrades filtered plasma proteins to reclaim valuable nutrients and maintain body homeostasis. Defects in this process result in proteinuria, one of the most frequently used biomarkers of kidney disease. Filtered proteins enter proximal tubules via receptor-mediated endocytosis and are processed within a highly developed apical endo-lysosomal system (ELS). Proteinuria is a strong risk factor for chronic kidney disease progression and genetic disorders of the ELS cause hereditary kidney diseases, so deepening understanding of how the proximal tubule handles proteins is crucial for translational nephrology. Moreover, the ELS is both an entry point for nephrotoxins that induce tubular damage and a target for novel therapies to prevent it. Cutting-edge research techniques, such as functional intravital imaging and computational modelling, are shedding light on spatial and integrative aspects of renal tubular protein processing in vivo, how these are altered under pathological conditions and the consequences for other tubular functions. These insights have potentially important implications for understanding the origins of systemic complications arising in proteinuric states, and might lead to the development of new ways of monitoring and treating kidney diseases.
Collapse
Affiliation(s)
- Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.
- Zurich Kidney Center, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Wei J, Chen XY, Wang ZJ, Li XY, Zhang MM, Sun T, Zhang XR, Wang DG, Hou C, Meng XM. Galloflavin mitigates acute kidney injury by suppressing LDHA-dependent macrophage glycolysis. Int Immunopharmacol 2025; 150:114265. [PMID: 39955920 DOI: 10.1016/j.intimp.2025.114265] [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: 01/05/2025] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
Macrophage-mediated inflammation is closely linked to the pathogenesis of acute kidney injury (AKI) and the shift of macrophages to a pro-inflammatory phenotype being reliant on glycolytic metabolism. Galloflavin, a polyphenol derived from tea, functions as a lactate dehydrogenase A (LDHA) inhibitor, effectively obstructing glycolytic metabolic pathways. However, the specific immunometabolic regulatory functions of galloflavin in macrophages remain unclear. Here, we observed that galloflavin drives alleviation of glycolytic metabolism levels in lipopolysaccharide (LPS)-induced macrophages (RAW264.7 cells and human peripheral blood mononuclear cells-derived macrophages) through downregulation of LDHA expression, thereby inhibiting macrophage conversion to a pro-inflammatory phenotype and reducing the release of inflammatory cytokines. However, the overexpression of LDHA counteracts the effects of galloflavin in macrophages. In addition, in vivo experiments observed a protective effect of galloflavin against cecal ligation and puncture (CLP) and cisplatin-induced renal injury. The ability of galloflavin to inhibit glycolysis in renal macrophages, thereby regulating their phenotypic transition during AKI was further validated through the isolation of renal primary macrophages. This intervention ultimately ameliorated the inflammatory response and decelerated the progression of AKI. Collectively, galloflavin confers protection against AKI by suppressing glycolysis in macrophages through a LDHA-dependent mechanism, thereby positioning it as a potential therapeutic option for AKI in the future.
Collapse
Affiliation(s)
- Jie Wei
- Department of Nephrology, the Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Xin-Yu Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhi-Juan Wang
- Department of Nephrology, the Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Xiang-Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Meng-Meng Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Tao Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xin-Ru Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - De-Guang Wang
- Department of Nephrology, the Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China.
| | - Chao Hou
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
6
|
He C, Peng W, Li S, Xu C, Chen X, Qin Y. ECHS1 as a Lipid Metabolism Biomarker for Pediatric Focal Segmental Glomerulosclerosis. PLoS One 2025; 20:e0319049. [PMID: 40063869 PMCID: PMC11893130 DOI: 10.1371/journal.pone.0319049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/26/2025] [Indexed: 05/13/2025] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a common cause of nephrotic syndrome and often leads to end-stage renal disease. However, the underlying pathophysiological mechanisms that contribute to disease progression require further investigation to establish appropriate therapeutic targets and biomarkers. This study aimed to clarify the molecular mechanisms underlying FSGS by focusing on differentially expressed genes (DEGs) and lipid metabolism-related genes (LMRGs). We utilized the GSE69814, GSE129973, and GSE121233 datasets, which comprise glomerular transcriptomes from patients with FSGS, minimal change disease (MCD), and unaffected kidney tissues. We identified 2,459 DEGs from the GSE69814 dataset and 982 DEGs from the GSE129973 dataset. These DEGs intersected 1,450 LMRGs, resulting in 56 differentially expressed LMRGs (DELMRGs). Enrichment analysis revealed that these DELMRGs were primarily involved in fatty acid metabolic processes; localized in microbodies, peroxisomes, and mitochondrial matrices; and exhibited oxidoreductase activity. Protein-protein interaction networks were constructed using Cytoscape, and five hub DELMRGs (enoyl-CoA hydratase, short chain 1 [ECHS1], EHHADH, IDH1, SUCLG1, and ALDH3A2) were identified using multiple algorithms. We assessed the diagnostic performance using receiver operating characteristic curves and expression levels from the GSE121233 dataset, and found that ECHS1 and ALDH3A2 showed strong diagnostic potential. Immunohistochemical verification of clinical specimens from children confirmed significant expression of ECHS1 in FSGS compared with that in normal and MCD tissues. This study highlights ECHS1 as a potential biomarker for pediatric FSGS, suggesting a potential role in early diagnosis or personalized treatment, offering insights into its pathogenesis and paving the way for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Chao He
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University
- The First Affiliated Hospital, Department of Pediatrics, Hengyang Medical School, University of South China
| | - Wei Peng
- Department of Pediatrics, People’s Hospital of Ningxiang City
| | - Sheng Li
- The First Affiliated Hospital, Department of Pediatrics, Hengyang Medical School, University of South China
| | - Can Xu
- The First Affiliated Hospital, Department of Cardiology, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China
| | - Xiuping Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University
| | - Yuanhan Qin
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University
| |
Collapse
|
7
|
Sun P, Chen Q, Chen X, Zhou J, Long T, Ma Y, Zhou M, Hu Z, Tian J, Zhu F, Yang Z, Xie L, Wu Q, Nie J. Renal tubular S100A7a impairs fatty acid oxidation and exacerbates renal fibrosis via both intracellular and extracellular pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167656. [PMID: 39778778 DOI: 10.1016/j.bbadis.2025.167656] [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: 09/19/2024] [Revised: 12/16/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025]
Abstract
A couple of S100 family proteins (S100s) have been reported to exert pro-inflammatory functions in the progression of renal fibrosis. Unlike some S100s which are expressed by both epithelial and stromal inflammatory cells, S100A7 is restricted expressed in epithelium. Persistent S100A7 expression occurs in some invasive carcinomas and is associated with poor prognostic factors. Whereas, whether it is implicated in renal tubular epithelial cell injury and kidney disease remains unexplored. In this study, we demonstrate that S100A7 is highly upregulated in tubular cells of both mouse renal fibrotic lesions and kidney biopsies from patients with chronic kidney disease (CKD). The level of renal S100A7 was associated with both the decline of renal function and the progression of renal fibrosis in CKD patients. Overexpressing S100A7a impaired fatty acid oxidation (FAO) and promoted lipid peroxidation in proximal tubular cells (PTCs). Mechanistically, S100A7a interacts with β-catenin, thereby preventing its ubiquitination and degradation by the β-TrCP-SCF complex, and in turn activated β-catenin signaling, downregulated the expression of PGC-1α. Additionally, S100A7a exacerbated lipid peroxidation via RAGE-p-ERK-NOX2 pathway. Specific deletion of S100a7a in tubular cells enhanced FAO and reduced lipid peroxidation, resulting in improved renal function and alleviation of renal fibrosis induced by unilateral ureteral obstruction and unilateral ischemia-reperfusion injury. Collectively, we delineate a previously unrecognized function of S100A7a in the progression of renal fibrosis.
Collapse
Affiliation(s)
- Pengxiao Sun
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qingzhou Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaomei Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiaxin Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tantan Long
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuanyuan Ma
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Miaomiao Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zheng Hu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianwei Tian
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fengxin Zhu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhenhua Yang
- Department of Nephrology, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China
| | - Liling Xie
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiaoyuan Wu
- Department of Nephrology, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China.
| | - Jing Nie
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Biobank of Peking University First Hospital, Peking University First Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University Health Science Center, Peking University, Beijing 100034, China.
| |
Collapse
|
8
|
Guo X, Zhang Z, Li C, Li X, Cao Y, Wang Y, Li J, Wang Y, Wang K, Liu Y, Xie C, Zhong Y. Lipidomics reveals potential biomarkers and pathophysiological insights in the progression of diabetic kidney disease. Metabol Open 2025; 25:100354. [PMID: 40125416 PMCID: PMC11930151 DOI: 10.1016/j.metop.2025.100354] [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: 01/14/2025] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/25/2025] Open
Abstract
Background Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease, affecting over 30 % of diabetes mellitus (DM) patients. Early detection of DKD in DM patients can enable timely preventive therapies, and potentially delay disease progression. Since the kidney relies on fatty acid oxidation for energy, dysregulated lipid metabolism has been implicated in proximal tubular cell damage and DKD pathogenesis. This study aimed to identify lipid alterations during DKD development and potential biomarkers differentiating DKD from DM. Methods lipidomics analysis was performed on serum collected from 55 patients with DM, 21 with early DKD stage and 32 with advanced DKD, and 22 healthy subjects. Associations between lipids and DKD risk were evaluated by logistic regression. Results Lipid profiling revealed elevated levels of certain lysophosphatidylethanolamines (LPEs), phosphatidylethanolamines (PEs), ceramides (Cers), and diacylglycerols (DAGs) in the DM-DKD transition, while most LPEs, lysophosphatidylcholines (LPCs), along with several monoacylglycerol (MAG) and triacylglycerols (TAGs), increased further from DKD-E to DKD-A. Logistic regression indicated positive associations between LPCs, LPEs, PEs, and DAGs with DKD risk, with most LPEs correlating significantly with urinary albumin-to-creatinine ratio (UACR) and inversely with estimated glomerular filtration rate (eGFR). A machine-learning-derived biomarker panel, Lipid9, consisting of LPC(18:2), LPC(20:5), LPE (16:0), LPE (18:0), LPE (18:1), LPE (24:0), PE (34:1), PE (34:2), and PE (36:2), accurately distinguished DKD (AUC: 0.78, 95 % CI 0.68-0.86) from DM. Incorporating two clinical indexes, serum creatinine and blood urea nitrogen, the Lipid9-SCB model further improved DKD detection (AUC: 0.83, 95 % CI 0.75-0.90) from DM, and was notably more sensitive for identifying DKD-E (AUC: 0.79, 95 % CI 0.67-0.91). Conclusion This study deciphers the lipid signature in DKD progression, and suggests the Lipid9-SCB panel as a promising tool for early DKD detection in DM patients.
Collapse
Affiliation(s)
- Xiaozhen Guo
- Department of Nephrology A, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
| | - Zixuan Zhang
- Department of Nephrology A, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
| | - Cuina Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
| | - Xueling Li
- Department of Nephrology A, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yutang Cao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Yangyang Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Jiaqi Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yibin Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Kanglong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
| | - Yameng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yifei Zhong
- Department of Nephrology A, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| |
Collapse
|
9
|
Chen Y, Torta F, Koh HWL, Benke PI, Gurung RL, Liu JJ, Ang K, Shao YM, Chan GC, Choo JCJ, Ching J, Kovalik JP, Kalhan T, Dorajoo R, Khor CC, Li Y, Tang WE, Seah DEJ, Sabanayagam C, Sobota RM, Venkataraman K, Coffman T, Wenk MR, Sim X, Lim SC, Tai ES. Metabolomics profiling in multi-ancestral individuals with type 2 diabetes in Singapore identified metabolites associated with renal function decline. Diabetologia 2025; 68:557-575. [PMID: 39621102 DOI: 10.1007/s00125-024-06324-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/19/2024] [Indexed: 02/19/2025]
Abstract
AIMS/HYPOTHESIS This study aims to explore the association between plasma metabolites and chronic kidney disease progression in individuals with type 2 diabetes. METHODS We performed a comprehensive metabolomic analysis in a prospective cohort study of 5144 multi-ancestral individuals with type 2 diabetes in Singapore, using eGFR slope as the primary outcome of kidney function decline. In addition, we performed genome-wide association studies on metabolites to assess how these metabolites could be genetically influenced by metabolite quantitative trait loci and performed colocalisation analysis to identify genes affecting both metabolites and kidney function. RESULTS Elevated levels of 61 lipids with long unsaturated fatty acid chains such as phosphatidylethanolamines, triacylglycerols, diacylglycerols, ceramides and deoxysphingolipids were prospectively associated with more rapid kidney function decline. In addition, elevated levels of seven amino acids and three lipids in the plasma were associated with a slower decline in eGFR. We also identified 15 metabolite quantitative trait loci associated with these metabolites, within which variants near TM6SF2, APOE and CPS1 could affect both metabolite levels and kidney functions. CONCLUSIONS/INTERPRETATION Our study identified plasma metabolites associated with prospective renal function decline, offering insights into the underlying mechanism by which the metabolite abnormalities due to fatty acid oversupply might reflect impaired β-oxidation and associate with future chronic kidney disease progression in individuals with diabetes.
Collapse
Affiliation(s)
- Yuqing Chen
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Republic of Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Republic of Singapore
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Hiromi W L Koh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Peter I Benke
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Republic of Singapore
| | - Resham L Gurung
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Keven Ang
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Yi-Ming Shao
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Gek Cher Chan
- Division of Nephrology, Department of Medicine, National University Hospital, Singapore, Republic of Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Republic of Singapore
| | - Jason Chon-Jun Choo
- Department of Renal Medicine, Singapore General Hospital, Singapore, Republic of Singapore
| | - Jianhong Ching
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Republic of Singapore
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Republic of Singapore
| | - Jean-Paul Kovalik
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Tosha Kalhan
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Republic of Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Chiea Chuen Khor
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
- Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - Wern Ee Tang
- National Healthcare Group Polyclinics, Singapore, Republic of Singapore
| | - Darren E J Seah
- National Healthcare Group Polyclinics, Singapore, Republic of Singapore
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Republic of Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Radoslaw M Sobota
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Kavita Venkataraman
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Republic of Singapore
| | - Thomas Coffman
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Republic of Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Republic of Singapore.
| | - Su-Chi Lim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Republic of Singapore.
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore.
- Diabetes Center, Khoo Teck Puat Hospital, Singapore, Republic of Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Republic of Singapore.
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Republic of Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Republic of Singapore.
| |
Collapse
|
10
|
Cheng Z, Zhan H, Yuan H, Wang N, Lan Y, Qu W, Lan X, Liao Z, Wang G, Chen M. Boeravinone C ameliorates lipid accumulation and inflammation in diabetic kidney disease by activating PPARα signaling. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119398. [PMID: 39880066 DOI: 10.1016/j.jep.2025.119398] [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: 10/16/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The roots of Oxybaphus himalaicus Edgew. is a traditional Tibetan herbal medicine with kidney reinforcing and tonifying effects, which is commonly applied to treat nephritis. Boeravinone C has been identified as one of the primary constituents of O. himalaicus. However, the potential renal protective effects of boeravinone C remains unclear. AIM OF THE STUDY This research aimed to investigate the protective effects of boeravinone C on diabetic kidney disease and the underlying mechanisms. MATERIALS AND METHODS Streptozotocin (100 mg/kg) was intraperitoneally injected to induce DKD in mice. High glucose (50 mM)-induced HK-2 cells were utilized to investigate the mechanisms of boeravinone C against tubular injuries in vitro. Anti-DKD activity was assessed by measuring reactive oxygen species (ROS) levels, analyzing apoptosis through flow cytometry, and evaluating inflammation, apoptosis, and FAO-related proteins via Western blotting. Additionally, serum biochemical assays, as well as histopathological and immunohistochemical analyses of kidney tissues, were performed to explore the pharmacological effects of boeravinone C. RESULTS In vivo, boeravinone C administered significantly reduced the creatinine (CRE), blood urea nitrogen (BUN), triglycerides (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) levels in serum of DKD mice. In vitro, boeravinone C significantly restored the apoptosis induced by HG in HK-2 cells, which is further validated by an upregulation of the apoptosis-inhibiting protein Bcl-2, along with a decreased expression of the apoptosis-promoting proteins Bax and caspase-3. Mechanistically, boeravinone C reversed HG-induced downregulation of peroxisome proliferator-activated receptor α (PPARα) expression. As a transcription factor, elevated expression of PPARα led to upregulation of CPT1A and ACOX1, which then enhanced fatty acid oxidation (FAO) to reduce lipid accumulation in HK-2 cells. Furthermore, boeravinone C-mediated high expression of PPARα sequestered p65 subunit of NF-κB in the cytoplasm, leading to reduced expression of proinflammatory cytokines such as iNOS, TNF-α and IL-6. To verify that the therapeutic effects of boeravinone C in diabetic kidney disease (DKD) are mediated via PPARα activation, we developed a PPARα knockdown HK-2 cell line. Our findings revealed that PPARα downregulation modified biological effects of boeravinone C, especially regarding fatty acid metabolism and the inflammatory response, with significant repercussions on apoptosis. CONCLUSION This study demonstrates that the major component boeravinone C from O. himalaicus promotes the fatty acid oxidation and suppresses inflammatory response by upregulating PPARα expression, thereby reducing apoptosis in HG-induced renal tubule cells. Consequently, boeravinone C restores tubular function in DKD mice. Collectively, this study provides a pharmacological basis for utilizing of O. himalaicus in treating DKD.
Collapse
Affiliation(s)
- Zhuoqing Cheng
- Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Honghong Zhan
- Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Han Yuan
- Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Nan Wang
- Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Yi Lan
- Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Weijian Qu
- Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Xiaozhong Lan
- TAAHC-SWU Medicinal Plant R&D Center, Xizang Agriculture and Animal Husbandry College, Nyingchi, Tibet, 860000, PR China
| | - Zhihua Liao
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City & Southwest University, School of Life Sciences, Southwest University, Chongqing, 400715, PR China
| | - Guowei Wang
- Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Min Chen
- Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
11
|
Tan SHC, Loh WJ, Lim SC. Precision medicine in diabetes care. Curr Opin Endocrinol Diabetes Obes 2025; 32:12-19. [PMID: 39564663 DOI: 10.1097/med.0000000000000894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
PURPOSE OF REVIEW This review highlights emerging evidence supporting the premise of precision diabetes care including but not limited to monogenic diabetes and discuss potential opportunities, challenges, and limitations for clinical adoption. RECENT FINDINGS Driven by a single gene mutation, monogenic diabetes remains the best use-case for precision diabetes care. However, the increasing prevalence of diabetes among adolescents and young adults in an obesogenic environment makes triaging potential patients for genetic screening clinically challenging. High-dimensional molecular biomarkers (i.e., multiomics) can improve the risk prediction for incident type 2 diabetes (T2D), over and above a well established prediction model based on clinical variables alone. Machine learning approaches using clinical variable-based clustering methods have generated novel and reproducible T2D subgroups with distinct phenotypic and omics characteristics that are associated with differential long-term outcomes. This stratification-strategy may inform clinical decisions. However, on-going discussion and research will be needed to understand the clinical utility of sub-phenotyping T2D for precision care. SUMMARY Precision diabetes care has extended from uncommon monogenic diabetes to T2D which will need more complex approaches like multiomics and machine-learning methods. The successful clinical translation will require cumulative evidence and close collaboration among the stake holders.
Collapse
Affiliation(s)
| | - Wann Jia Loh
- Department of Endocrinology, Changi General Hospital
- Duke-NUS Medical School, Singapore
- Medical School, University of Western Australia, Perth, Australia
| | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital
- Diabetes Centre, Admiralty Medical Centre
- Saw Swee Hock School of Public Heath
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
12
|
Liu Y, Zhang X, Yi R, Tian Q, Xu J, Yan X, Ma J, Wang S, Yang G. Exploring the nephrotoxicity and molecular mechanisms of Di-2-ethylhexyl phthalate: A comprehensive review. Chem Biol Interact 2025; 405:111310. [PMID: 39549904 DOI: 10.1016/j.cbi.2024.111310] [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: 09/04/2024] [Revised: 10/21/2024] [Accepted: 11/14/2024] [Indexed: 11/18/2024]
Abstract
Di-2-ethylhexyl phthalate (DEHP), a widely applied plasticizer in various products, can be absorbed into the human body through several channels and accumulate in the lungs, liver, testes, and kidneys, potentially impairing the function of these organs. Recently, the nephrotoxicity of DEHP has received heightened attention. Numerous epidemiologic findings have demonstrated that DEHP exposure may contribute to renal damage, leading to structural and functional abnormalities and exacerbating the progression of kidney disease. Recent research has discovered the mechanisms behind DEHP-induced nephrotoxicity may involve a variety of pathways, including apoptosis, autophagy, ferroptosis, oxidative stress, inflammation, DNA damage, and lipid metabolism disorders. This review discusses the impact of DEHP on kidney function and delves into the molecular mechanisms of nephrotoxicity mediated by DEHP in recent years. In addition, the review examines evidence for the antioxidant and anti-inflammatory capacities of lycopene, green tea polyphenols, and quercetin in ameliorating DEHP-induced renal injury is reviewed, providing a basis for further research.
Collapse
Affiliation(s)
- Yun Liu
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian, 116044, China
| | - Xu Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian, 116044, China
| | - Ruhan Yi
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian, 116044, China
| | - Qing Tian
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian, 116044, China
| | - Jiawei Xu
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian, 116044, China
| | - Xinyu Yan
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian, 116044, China
| | - Jiaxuan Ma
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian, 116044, China
| | - Shaopeng Wang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Guang Yang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian, 116044, China.
| |
Collapse
|
13
|
Su X, Bai M, Shang Y, Du Y, Xu S, Lin X, Xiao Y, Zhang Y, Chen H, Zhang A. Slc25a21 in cisplatin-induced acute kidney injury: a new target for renal tubular epithelial protection by regulating mitochondrial metabolic homeostasis. Cell Death Dis 2024; 15:891. [PMID: 39695098 DOI: 10.1038/s41419-024-07231-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024]
Abstract
Acute kidney injury (AKI) is a significant global health issue, which is often caused by cisplatin therapy and characterized by mitochondrial dysfunction. Restoring mitochondrial homeostasis in tubular cells could exert therapeutic effects. Here, we investigated Slc25a21, a mitochondrial carrier, as a potential target for AKI intervention. Renal Slc25a21 expression is negatively associated with kidney function in both AKI patients and cisplatin-induced murine models. Sustaining renal expression of Slc25a21 slowed down AKI progression by reducing cellular apoptosis, necroptosis, and the inflammatory response, likely through its regulation of 2-oxoadipate conversion. Slc25a21 is highly expressed in proximal tubular epithelial cells, and its down-regulation contributes to compromised mitochondrial biogenesis and integrity, as well as impaired oxidative phosphorylation. Mechanistically, reduced Slc25a21 in AKI disrupts mitochondrial 2-oxoadipate transport, affecting related metabolites influx and the tricarboxylic acid cycle. These findings demonstrate a previously unappreciated metabolic function of Slc25a21 in tubular cells, and suggest that targeting mitochondrial metabolic homeostasis by sustaining Slc25a21 expression could be a potential novel therapeutic strategy for AKI.
Collapse
Affiliation(s)
- Xin Su
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
| | - Mi Bai
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Yaqiong Shang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Yang Du
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Shuang Xu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Xiuli Lin
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Yunzhi Xiao
- Centre for Computational Biology and Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Huimei Chen
- Centre for Computational Biology and Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore.
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
14
|
Xu W, Zhu Y, Wang S, Liu J, Li H. From Adipose to Ailing Kidneys: The Role of Lipid Metabolism in Obesity-Related Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:1540. [PMID: 39765868 PMCID: PMC11727289 DOI: 10.3390/antiox13121540] [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: 10/31/2024] [Revised: 12/01/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
Obesity has emerged as a significant public health crisis, closely linked to the pathogenesis and progression of chronic kidney disease (CKD). This review explores the intricate relationship between obesity-induced lipid metabolism disorders and renal health. We discuss how excessive free fatty acids (FFAs) lead to lipid accumulation in renal tissues, resulting in cellular lipotoxicity, oxidative stress, and inflammation, ultimately contributing to renal injury. Key molecular mechanisms, including the roles of transcriptional regulators like PPARs and SREBP-1, are examined for their implications in lipid metabolism dysregulation. The review also highlights the impact of glomerular and tubular lipid overload on kidney pathology, emphasizing the roles of podocytes and tubular cells in maintaining kidney function. Various therapeutic strategies targeting lipid metabolism, including pharmacological agents such as statins and SGLT2 inhibitors, as well as lifestyle modifications, are discussed for their potential to mitigate CKD progression in obese individuals. Future research directions are suggested to better understand the mechanisms linking lipid metabolism to kidney disease and to develop personalized therapeutic approaches. Ultimately, addressing obesity-related lipid metabolism disorders may enhance kidney health and improve outcomes for individuals suffering from CKD.
Collapse
Affiliation(s)
- Wenchao Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuting Zhu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Siyuan Wang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
15
|
Srivastava SP, Zhou H, Shenoi R, Morris M, Lainez-Mas B, Goedeke L, Rajendran BK, Setia O, Aryal B, Kanasaki K, Koya D, Inoki K, Dardik A, Bell T, Fernández-Hernando C, Shulman GI, Goodwin JE. Renal Angptl4 is a key fibrogenic molecule in progressive diabetic kidney disease. SCIENCE ADVANCES 2024; 10:eadn6068. [PMID: 39630889 PMCID: PMC11616692 DOI: 10.1126/sciadv.adn6068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
Angiopoietin-like 4 (ANGPTL4), a key protein involved in lipoprotein metabolism, has diverse effects. There is an association between Angptl4 and diabetic kidney disease; however, this association has not been well investigated. We show that both podocyte- and tubule-specific ANGPTL4 are crucial fibrogenic molecules in diabetes. Diabetes accelerates the fibrogenic phenotype in control mice but not in ANGPTL4 mutant mice. The protective effect observed in ANGPTL4 mutant mice is correlated with a reduction in stimulator of interferon genes pathway activation, expression of pro-inflammatory cytokines, reduced epithelial-to-mesenchymal transition and endothelial-to-mesenchymal transition, lessened mitochondrial damage, and increased fatty acid oxidation. Mechanistically, we demonstrate that podocyte- or tubule-secreted Angptl4 interacts with Integrin β1 and influences the association between dipeptidyl-4 with Integrin β1. We demonstrate the utility of a targeted pharmacologic therapy that specifically inhibits Angptl4 gene expression in the kidneys and protects diabetic kidneys from proteinuria and fibrosis. Together, these data demonstrate that podocyte- and tubule-derived Angptl4 is fibrogenic in diabetic kidneys.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Han Zhou
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
| | - Rachel Shenoi
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Myshal Morris
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Begoña Lainez-Mas
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
| | - Leigh Goedeke
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ocean Setia
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT, USA
| | - Binod Aryal
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Keizo Kanasaki
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
- Department of Internal Medicine 1, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
- The Center for Integrated Kidney Research and Advance, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT, USA
| | | | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Gerald I. Shulman
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
16
|
Mu Z, Li B, Chen M, Liang C, Gu W, Su J. Endoplasmic reticulum stress induces renal fibrosis in high‑fat diet mice via the TGF‑β/SMAD pathway. Mol Med Rep 2024; 30:235. [PMID: 39422027 PMCID: PMC11544397 DOI: 10.3892/mmr.2024.13360] [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: 07/03/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The aim of the present study was to investigate the role and mechanism of endoplasmic reticulum stress (ERS) in kidney injury caused by high‑fat diet (HFD). An obese mouse model was established via HFD feeding and intervention was performed by intraperitoneal injection of the ERS inhibitor salubrinal (Sal). Changes in the body and kidney weight and serum biochemical indices of the mice were determined. Hematoxylin and eosin and Masson staining were used to observe the pathological changes of renal tissues. Reverse transcription‑quantitative PCR and western blotting were used to observe the expression of ERS‑related proteins and TGF‑β/SMAD pathway‑related proteins. Immunohistochemistry was employed to explore the distribution of these proteins. Compared with those in the control group, the weight gain, lipid metabolism disorders and deterioration of renal function in the model group were greater. Malondialdehyde was elevated and superoxide dismutase was decreased in renal tissues. The mRNA and protein levels of TGF‑β1, SMAD2/3, α‑smooth muscle actin, collagen I, glucose‑regulated protein 78 and C/EBP‑homologous protein were markedly elevated, whereas SMAD7 was markedly decreased. Sal markedly inhibited the aforementioned effects. This investigation revealed a link between ERS and renal injury caused by HFD. ERS in HFD‑fed mice triggers renal fibrosis through the TGF‑β/SMAD pathway.
Collapse
Affiliation(s)
- Zhidan Mu
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Bin Li
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Mingyang Chen
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Chen Liang
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Wei Gu
- Department of Infection Disease, First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Juan Su
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| |
Collapse
|
17
|
Reiter RJ, Sharma RN, Manucha W, Rosales-Corral S, Almieda Chuffa LGD, Loh D, Luchetti F, Balduini W, Govitrapong P. Dysfunctional mitochondria in age-related neurodegeneration: Utility of melatonin as an antioxidant treatment. Ageing Res Rev 2024; 101:102480. [PMID: 39236857 DOI: 10.1016/j.arr.2024.102480] [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: 06/21/2024] [Revised: 08/18/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Mitochondria functionally degrade as neurons age. Degenerative changes cause inefficient oxidative phosphorylation (OXPHOS) and elevated electron leakage from the electron transport chain (ETC) promoting increased intramitochondrial generation of damaging reactive oxygen and reactive nitrogen species (ROS and RNS). The associated progressive accumulation of molecular damage causes an increasingly rapid decline in mitochondrial physiology contributing to aging. Melatonin, a multifunctional free radical scavenger and indirect antioxidant, is synthesized in the mitochondrial matrix of neurons. Melatonin reduces electron leakage from the ETC and elevates ATP production; it also detoxifies ROS/RNS and via the SIRT3/FOXO pathway it upregulates activities of superoxide dismutase 2 and glutathione peroxidase. Melatonin also influences glucose processing by neurons. In neurogenerative diseases, neurons often adopt Warburg-type metabolism which excludes pyruvate from the mitochondria causing reduced intramitochondrial acetyl coenzyme A production. Acetyl coenzyme A supports the citric acid cycle and OXPHOS. Additionally, acetyl coenzyme A is a required co-substrate for arylalkylamine-N-acetyl transferase, which rate limits melatonin synthesis; therefore, melatonin production is diminished in cells that experience Warburg-type metabolism making mitochondria more vulnerable to oxidative stress. Moreover, endogenously produced melatonin diminishes during aging, further increasing oxidative damage to mitochondrial components. More normal mitochondrial physiology is preserved in aging neurons with melatonin supplementation.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA.
| | - Ramaswamy N Sharma
- Applied Biomedical Sciences, University of the Incarnate Word, School of Osteopathic Medicine, San Antonio, TX, USA.
| | - Walter Manucha
- Instituto de Medicina y Biologia Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Cientificas y Tecnologicas (CONICET), Mendoza 5500, Argentina.
| | - Sergio Rosales-Corral
- Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico.
| | - Luiz Gustavo de Almieda Chuffa
- Departamento de Biologia Estrutural e Funcional, Setor de Anatomia - Instituto de Biociências, IBB/UNESP, Campus Botucatu, Botucatu, São Paulo, Brazil.
| | - Doris Loh
- Independent Researcher, Marble Falls, TX, USA.
| | - Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - Walter Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Laksi, Bangkok, Thailand.
| |
Collapse
|
18
|
Gorman BL, Lukowski JK. Spatial Metabolomics and Lipidomics in Kidney Disease. Semin Nephrol 2024; 44:151582. [PMID: 40234137 DOI: 10.1016/j.semnephrol.2025.151582] [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] [Indexed: 04/17/2025]
Abstract
Kidney disease is a global health issue that affects over 850 million people, and early detection is key to preventing severe disease and complications. Kidney diseases are associated with complex and dysregulation of lipid metabolism. Spatial metabolomics through mass spectrometry imaging (MSI) enables spatial mapping of the lipids in tissue and includes a variety of techniques that can be used to image lipids. In the kidney, MSI studies often seek to resolve individual functional tissue units such as glomeruli and proximal tubules. Several different MSI techniques, such as matrix-assisted laser desorption/ionization (MALDI) and desorption electrospray ionization (DESI), have been used to characterize lipids and small molecules in chronic kidney disease, acute kidney injury, genetic kidney disease, and cancer. In this review we provide several examples of how spatial metabolomics data can provide critical information concerning the localization of changes in various disease states. Additionally, when combined with pathology, transcriptomics, or proteomics, the metabolomic changes can illuminate underlying mechanisms and provide new clinical insights into disease mechanisms.
Collapse
Affiliation(s)
| | - Jessica K Lukowski
- Mass Spectrometry Imaging Lead, Mass Spectrometry Technology Access Center at the McDonnell Genome Institute, Washington University in St. Louis School of Medicine, St. Louis, MO
| |
Collapse
|
19
|
Rojas-Canales DM, Wong SW, Tucker EJ, Fedele AO, McNicholas K, Mehdorn AS, Gleadle JM. The transcriptome of early compensatory kidney growth reveals cell and time specific responses. iScience 2024; 27:110608. [PMID: 39220259 PMCID: PMC11363579 DOI: 10.1016/j.isci.2024.110608] [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: 03/22/2024] [Revised: 04/24/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Following kidney removal, the remaining kidney enlarges and increases its function. The mechanism and signals driving this compensatory kidney hypertrophy and the enlargement of its constituent kidney cells remains elusive. RNA-seq studies in mice undergoing hypertrophy 24, 48, and 72 h following nephrectomy were undertaken to understand the early transcriptional changes. This revealed substantial enhancement of cholesterol biosynthesis pathways, increases in mitochondrial gene expression and cell cycle perturbations. Single nuclei RNA-seq delineated cell specific changes at 24 h post nephrectomy and showed that sterol binding protein 2 (SREBP2) activity increases in medullary thick ascending limb cells in keeping with promotion of cholesterol synthesis. Cultured renal tubular cells were examined for insulin-like growth factor-1 (IGF-1) stimulated hypertrophy and SREBP2 was found to be required for increase in cell size. This work describes the early cell specific growth pathways mediating cellular and kidney hypertrophy with an intriguing role for cholesterol synthesis.
Collapse
Affiliation(s)
- Darling M. Rojas-Canales
- Department of Renal Medicine, Southern Adelaide Local Health Network, Flinders Medical Centre, Bedford Park, SA, Australia
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Soon Wei Wong
- Department of Renal Medicine, Southern Adelaide Local Health Network, Flinders Medical Centre, Bedford Park, SA, Australia
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Elise J. Tucker
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Anthony O. Fedele
- Department of Renal Medicine, Southern Adelaide Local Health Network, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Kym McNicholas
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Anne-Sophie Mehdorn
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
- Department of General, Abdominal, Thoracic, Transplantation and Paediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Jonathan M. Gleadle
- Department of Renal Medicine, Southern Adelaide Local Health Network, Flinders Medical Centre, Bedford Park, SA, Australia
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
20
|
Zhang J, Gao Y, Li Y, Liu D, Sun W, Liu C, Zhao X. Transcriptome Analysis of the Effect of Nickel on Lipid Metabolism in Mouse Kidney. BIOLOGY 2024; 13:655. [PMID: 39336083 PMCID: PMC11429462 DOI: 10.3390/biology13090655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/17/2024] [Accepted: 08/18/2024] [Indexed: 09/30/2024]
Abstract
Although the human body needs nickel as a trace element, too much nickel exposure can be hazardous. The effects of nickel on cells include inducing oxidative stress, interfering with DNA damage repair, and altering epigenetic modifications. Glucose metabolism and lipid metabolism are closely related to oxidative stress; however, their role in nickel-induced damage needs further study. In Institute of Cancer Research (ICR) mice, our findings indicated that nickel stress increased the levels of blood lipid indicators (triglycerides, high-density lipoprotein, and cholesterol) by about 50%, blood glucose by more than two-fold, and glycated serum protein by nearly 20%. At the same time, nickel stress increased oxidative stress (malondialdehyde) and inflammation (Interleukin 6) by about 30% in the kidney. Based on next-generation sequencing technology, we detected and analyzed differentially expressed genes in the kidney caused by nickel stress. Bioinformatics analysis and experimental verification showed that nickel inhibited the expression of genes related to lipid metabolism and the AMPK and PPAR signaling pathways. The finding that nickel induces kidney injury and inhibits key genes involved in lipid metabolism and the AMPK and PPAR signaling pathways provides a theoretical basis for a deeper understanding of the mechanism of nickel-induced kidney injury.
Collapse
Affiliation(s)
- Jing Zhang
- School of Life Science and Technology, Inner Mongolia University of Science & Technology, Baotou 014020, China; (J.Z.); (Y.G.); (Y.L.); (D.L.); (W.S.)
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou 014010, China
| | - Yahong Gao
- School of Life Science and Technology, Inner Mongolia University of Science & Technology, Baotou 014020, China; (J.Z.); (Y.G.); (Y.L.); (D.L.); (W.S.)
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou 014010, China
| | - Yuewen Li
- School of Life Science and Technology, Inner Mongolia University of Science & Technology, Baotou 014020, China; (J.Z.); (Y.G.); (Y.L.); (D.L.); (W.S.)
| | - Dongdong Liu
- School of Life Science and Technology, Inner Mongolia University of Science & Technology, Baotou 014020, China; (J.Z.); (Y.G.); (Y.L.); (D.L.); (W.S.)
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou 014010, China
| | - Wenpeng Sun
- School of Life Science and Technology, Inner Mongolia University of Science & Technology, Baotou 014020, China; (J.Z.); (Y.G.); (Y.L.); (D.L.); (W.S.)
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou 014010, China
| | - Chuncheng Liu
- School of Life Science and Technology, Inner Mongolia University of Science & Technology, Baotou 014020, China; (J.Z.); (Y.G.); (Y.L.); (D.L.); (W.S.)
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou 014010, China
| | - Xiujuan Zhao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
21
|
Li H, Ren Q, Hu Y, Guo F, Huang R, Lin L, Tan Z, Ma L, Fu P. SKLB023 protects against inflammation and apoptosis in sepsis-associated acute kidney injury via the inhibition of toll-like receptor 4 signaling. Int Immunopharmacol 2024; 139:112668. [PMID: 39008938 DOI: 10.1016/j.intimp.2024.112668] [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: 05/20/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is one of common critical illnesses with high morbidity and mortality. At present, effective therapeutic drugs for SA-AKI are remain lacking. SKLB023 is a synthetic small-molecule compound which exerts potent anti-inflammatory effects in our previous studies. Here, this study aimed to characterize the protective effect of SKLB023 on SA-AKI and explore its underlying mechanism. The SA-AKI experimental models have been established by cecum ligation/puncture (CLP) and lipopolysaccharide (LPS) injection in male C57BL/6J mice. SKLB023 was administered by gavage (50 or 25 mg/kg in CLP model and 50 mg/kg in LPS model) daily 3 days in advance and 30 min earlier on the day of modeling. Our results confirmed SKLB023 treatment could improve the survival of SA-AKI mice and ameliorate renal pathological injury, inflammation, and apoptosis in the two types of septic AKI mice. Mechanically, SKLB023 deceased the expression of TLR4 in LPS-triggered renal tubular epithelial cells, and inhibited the activation of downstream pathways including NF-κB and MAPK pathways. Our study suggested that SKLB023 is expected to be a potential drug for the prevention and treatment of septic AKI.
Collapse
Affiliation(s)
- Hui Li
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| | - Qian Ren
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| | - Yao Hu
- Department of Medicine Renal Division, Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu 610041, PR China
| | - Fan Guo
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| | - Rongshuang Huang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| | - Lin Lin
- West-district Outpatient Department, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| | - Zhouke Tan
- Department of Nephrology, Organ Transplant Center, Guizhou Province Key Laboratory of Cell Engineering, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, PR China.
| | - Liang Ma
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China.
| | - Ping Fu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| |
Collapse
|