1
|
Zhang C, Ren W, Lu X, Feng L, Li J, Zhu B. The compound XueShuanTong promotes podocyte mitochondrial autophagy via the AMPK/mTOR pathway to alleviate diabetic nephropathy injury. Mitochondrion 2025; 83:102024. [PMID: 40043830 DOI: 10.1016/j.mito.2025.102024] [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: 08/13/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025]
Abstract
The study aimed to elucidate the molecular mechanisms underlying the protective effects of Compound Xueshuantong (CXst) in the context of diabetic nephropathy (DN), a major cause of kidney failure driven by podocyte injury and metabolic dysfunction. Given the critical role of the AMPK/mTOR signaling pathway in regulating cellular energy balance, autophagy, and mitochondrial health, we focused on its involvement in podocyte function and how it might be influenced by CXst. Through a series of experiments, we found that CXst treatment led to the upregulation of key proteins involved in autophagy, such as LC3 and p62, as well as proteins critical for mitochondrial function, like PGC-1α. These molecular changes helped to counteract the damaging effects of high glucose levels on podocytes, which are central to maintaining the filtration function of the kidneys. Additionally, CXst's ability to modulate the AMPK/mTOR pathway was shown to be a pivotal factor in its protective effects, as inhibition of AMPK significantly reduced these benefits. This comprehensive study provides strong evidence that CXst exerts its protective effects against DN by modulating the AMPK/mTOR pathway, thus preserving podocyte integrity and function. These findings suggest that CXst could be a promising candidate for the development of new therapeutic strategies for the treatment of DN, offering hope for better management of this challenging condition.
Collapse
Affiliation(s)
- Chuangbiao Zhang
- Department of Endocrinology, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province 510630, China
| | - Weiwei Ren
- Department of Endocrinology, Guangzhou Baiyun District Maternal And Child Health Hospital, Guangzhou, Guangdong Province 51000, China
| | - Xiaohua Lu
- Department of Endocrinology, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province 510630, China
| | - Lie Feng
- Department of Endocrinology, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province 510630, China
| | - Jiaying Li
- Department of Endocrinology, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province 510630, China.
| | - Beibei Zhu
- Endoscopy Center, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province 510630, China.
| |
Collapse
|
2
|
Nie P, Qin W, Nie WC, Li B. Progress in the application of mesenchymal stem cells to attenuate apoptosis in diabetic kidney disease. World J Diabetes 2025; 16:105711. [DOI: 10.4239/wjd.v16.i6.105711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/24/2025] [Accepted: 04/25/2025] [Indexed: 06/13/2025] Open
Abstract
Diabetic kidney disease (DKD) has a high incidence and mortality rate and lacks effective preventive and therapeutic methods. Apoptosis is one of the main reasons for the occurrence and development of DKD. Mesenchymal stem cells (MSCs) have shown great promise in tissue regeneration for DKD treatment and have protective effects against DKD, including decreased blood glucose and urinary protein levels and improved renal function. MSCs can directly differentiate into kidney cells or act via paracrine mechanisms to reduce apoptosis in DKD by modulating signaling pathways. MSC-derived extracellular vesicles (MSC-EVs) mitigate apoptosis and DKD-related symptoms by transferring miRNAs to target cells or organs. However, studies on the regulatory mechanisms of MSCs and MSC-EVs in apoptosis in DKD are insufficient. This review comprehensively examines the mechanisms of apoptosis in DKD and research progress regarding the roles of MSCs and MSC-EVs in the disease process.
Collapse
Affiliation(s)
- Ping Nie
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, China
| | - Wei Qin
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, China
| | - Wei-Chen Nie
- Basic Clinical Specialization in Integrative Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, China
| | - Bing Li
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, China
| |
Collapse
|
3
|
Liang Y, Zhang Q, Qian JR, Li SS, Liu QF. Inflammation-Induced Klotho Deficiency: A Possible Key Driver of Chronic Kidney Disease Progression. Int J Gen Med 2025; 18:2507-2520. [PMID: 40376197 PMCID: PMC12080484 DOI: 10.2147/ijgm.s513497] [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: 12/20/2024] [Accepted: 05/05/2025] [Indexed: 05/18/2025] Open
Abstract
Chronic kidney disease (CKD) is influenced by inflammation, a critical factor in its progression. However, the underlying mechanism through which inflammation contributes to CKD is still obscure. The Klotho protein, which is predominantly found in the kidneys, is known for its protective functions, including anti-inflammatory, anti-aging, antioxidant, and anti-fibrotic effects. A myriad of studies have suggested that inflammation in CKD leads to a decrease in Klotho expression, diminishing Klotho protection capabilities and exacerbating kidney damage, thereby promoting CKD progression. These findings suggest that Klotho deficiency could be a crucial link between inflammation and CKD progression. However, the mechanism regarding their relationship is still unclear. The reduction in Klotho due to inflammation may be attributed to epigenetic mechanisms, such as DNA methylation, histone deacetylation, transcription factor, microRNA (miRNA) regulation and long non-coding RNA (lncRNA) regulation or non-epigenetic factors, such as endoplasmic reticulum (ER) stress and ER-associated degradation (ERAD), which affect Klotho protein metabolism. Through these pathways, inflammation triggers a decrease in Klotho expression, further driving CKD progression. Notably, Klotho also exerts a strong anti-inflammatory effect by inhibiting key inflammatory factors and pathways, suggesting that there is intricate crosstalk between inflammatory factors and Klotho in CKD. This review highlights how inflammation suppresses the expression of Klotho and further contributes to the development and exacerbation of CKD. By focusing on the interplay between inflammation and Klotho, the present review provides novel potential therapeutic strategies such as correcting epigenetic and non-epigenetic abnormalities for treating CKD by targeting this specific axis.
Collapse
Affiliation(s)
- Yan Liang
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, Jiangsu, 215300, People’s Republic of China
| | - Qi Zhang
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, Jiangsu, 215300, People’s Republic of China
| | - Jing-Rong Qian
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, People’s Republic of China
| | - Sha-Sha Li
- Clinical Research and Laboratory Centre, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, People’s Republic of China
| | - Qi-Feng Liu
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, Jiangsu, 215300, People’s Republic of China
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, People’s Republic of China
| |
Collapse
|
4
|
Kang Y, Jin Q, Zhou M, Zheng H, Li D, Zhou J, Lv J, Wang Y. Association between serum α-klotho levels and the incidence of diabetic kidney disease and mortality in type 2 diabetes: evidence from a Chinese cohort and the NHANES database. Diabetol Metab Syndr 2025; 17:148. [PMID: 40312464 PMCID: PMC12046805 DOI: 10.1186/s13098-025-01711-x] [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: 10/08/2024] [Accepted: 04/22/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND The α-klotho is crucial in diabetes and its related complications. This study seeks to explore the link between α-klotho levels and the risk of diabetic kidney disease (DKD) as well as all-cause and cardiovascular mortality among individuals with type 2 diabetes mellitus (T2DM). METHODS The investigation involved 126 Chinese T2DM patients and 4,451 individuals from the National Health and Nutrition Examination Survey (NHANES) database. To evaluate the relationship between α-klotho levels and DKD risk, multivariate logistic regression was utilized. Additionally, restricted cubic spline (RCS) regression analysis was conducted to examine the nonlinear relationship between α-klotho levels and DKD incidence. RCS analysis was employed to explore the correlation between α-klotho and both all-cause and cardiovascular mortality. RESULTS In the Chinese cohort, α-klotho levels were notably elevated in T2DM group compared to DKD group. The NHANES data revealed a significant inverse relationship between α-klotho levels and DKD risk. Nonlinear analysis further illustrated a substantial nonlinear connection between α-klotho levels and DKD risk. Serum α-klotho levels below 880.78 pg/mL were linked to increased DKD risk in T2DM patients. When compared to the T2DM group, the DKD group had markedly higher all-cause and cardiovascular mortality rates, with the α-klotho low group (e.g., Q1) exhibiting lower survival compared to other groups. Cox regression findings indicated that elevated α-klotho levels could mitigate all-cause mortality in T2DM patients. The relationship between α-klotho levels and all-cause mortality was also nonlinear, with the minimal risk found at α-klotho levels between 776.95 pg/mL and 812.69 pg/mL, varying by gender. CONCLUSION There exists a notable association between α-klotho levels and DKD risk, along with mortality in T2DM patients, with varying effects based on gender. These results highlight the potential importance of α-klotho as both a biomarker and a therapeutic target.
Collapse
Affiliation(s)
- Yi Kang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Renal Research Institution, Beijing University of Chinese Medicine, Beijing, China
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| | - Qian Jin
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| | - Mengqi Zhou
- Department of Traditional Chinese Medicine, Beijing Puren Hospital, Beijing, China
| | - Huijuan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Renal Research Institution, Beijing University of Chinese Medicine, Beijing, China
| | - Danwen Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Renal Research Institution, Beijing University of Chinese Medicine, Beijing, China
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| | - Jingwei Zhou
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Renal Research Institution, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Lv
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
- Renal Research Institution, Beijing University of Chinese Medicine, Beijing, China.
| | - Yaoxian Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
- Renal Research Institution, Beijing University of Chinese Medicine, Beijing, China.
- Henan University of Traditional Chinese Medicine, Henan, China.
| |
Collapse
|
5
|
Alonazi AS, Aloraini RM, Albulayhi LM, Alshehri LM, Bin Dayel AF, Alamin MA, Aldamri NT, Alshammari TK, Alkhelb DA, Sarawi WS, Alghibiwi HK, Alrasheed NM, Elnagar DM, Alrasheed NM. Macrophage Depletion Alleviates Immunosenescence in Diabetic Kidney by Modulating GDF-15 and Klotho. Int J Mol Sci 2025; 26:3990. [PMID: 40362229 PMCID: PMC12071727 DOI: 10.3390/ijms26093990] [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: 03/27/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Cellular senescence is a hallmark of aging and contributes to age-related diseases, including diabetic nephropathy (DN). Additionally, macrophage-mediated inflammation has been linked with DKD. Therefore, we investigated the effect of macrophage depletion on kidney cell senescence in DN, focusing on the relationship between the GDF-15 and Klotho signaling pathways. Wistar albino rats (n = 24) were divided into four groups: healthy control, liposomal clodronate (LC)-treated healthy, diabetic, and LC-treated diabetic groups. Rats in the LC-treated healthy, diabetic, and LC-treated diabetic groups were intravenously administered LC once a week for 4 weeks. Rat models of type 2 diabetes were successfully established via the administration of streptozotocin and a high-fat diet, as evidenced by increased blood glucose levels, kidney weight to body weight (KW/BW) ratio, serum albumin, creatinine, and urea levels, kidney damage, and oxidative stress. However, LC-mediated macrophage depletion reduced the KW/BW ratio, improved serum and oxidative parameters, decreased inflammatory markers (IL-6 and TNF-α), and ameliorated oxidative stress. Additionally, LC treatment promoted macrophage polarization towards the anti-inflammatory phenotype, downregulated GDF-15 expression, upregulated Klotho expression, and ameliorated kidney damage. In conclusion, macrophage depletion combats kidney senescence by modulating Klotho and GDF-15, indicating their potential as novel targets in DN treatment.
Collapse
Affiliation(s)
- Asma S. Alonazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Rana M. Aloraini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Lama M. Albulayhi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Layal M. Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Anfal F. Bin Dayel
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Maha A. Alamin
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Nouf T. Aldamri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Tahani K. Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Dalal A. Alkhelb
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Wedad S. Sarawi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Hanan K. Alghibiwi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Nawal M. Alrasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| | - Doaa M. Elnagar
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
- Department of Zoology, Faculty of Woman, Ain Shams University, Cairo 11566, Egypt
| | - Nouf M. Alrasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (R.M.A.); (L.M.A.); (L.M.A.); (A.F.B.D.); (M.A.A.); (N.T.A.); (T.K.A.); (D.A.A.); (W.S.S.); (H.K.A.); (N.M.A.)
| |
Collapse
|
6
|
Wang X, Abu Bakar MH, Liqun S, Kassim MA, Shariff KA, Karunakaran T. Targeting metabolic diseases with celastrol: A comprehensive review of anti-inflammatory mechanisms and therapeutic potential. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119560. [PMID: 40015541 DOI: 10.1016/j.jep.2025.119560] [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: 12/10/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii is a traditional Chinese medicine used to treat rheumatic diseases, with properties such as clearing heat, detoxifying, dispelling wind, and relieving pain. In recent years, its active compound, celastrol, garnered significant attention for its potential therapeutic effects on metabolic diseases. Celastrol exhibits bioactivities such as regulating metabolic functions and anti-inflammatory effects, positioning it as a promising candidate for the treatment of obesity, diabetes, atherosclerosis (AS), and non-alcoholic fatty liver disease (NAFLD). AIM OF THE REVIEW This review aims to explore the pharmacological mechanisms of celastrol in metabolic diseases, focusing on its anti-inflammatory mechanisms and metabolic regulation effects, providing theoretical support for further investigation of its therapeutic potential in metabolic diseases. METHODS Literature was retrieved from PubMed, Web of Science, Scopus, Cochrane, and Google Scholar. This review primarily focuses on anti-inflammatory mechanisms of celastrol, its metabolic regulation, and toxicity studies, by systematically analyzing its effects in obesity, diabetes, AS, and NAFLD, providing scientific evidence for its potential clinical applications. RESULTS Celastrol regulates multiple signaling pathways, particularly inhibiting NF-κB and activating AMPK, reducing the production of pro-inflammatory cytokines and improving insulin sensitivity, enhancing its therapeutic potential in metabolic diseases. Additionally, celastrol regulates adipogenesis and energy metabolism by influencing key transcription factors such as PPARγ and SREBP-1c. Numerous studies highlight its role in alleviating oxidative stress and improving mitochondrial function, further enhancing its metabolic benefits. CONCLUSION In summary, celastrol holds great promise as a multi-target therapeutic agent for metabolic diseases, offering anti-inflammatory, metabolic regulatory, and antioxidative benefits. Despite these, challenges remain for the clinical application of celastrol due to its poor bioavailability and potential toxicity. Advanced formulation strategies and targeted delivery systems are urgently needed to overcome challenges related to bioavailability and clinical translation.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia; Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| | - Song Liqun
- Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohd Asyraf Kassim
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia
| | - Khairul Anuar Shariff
- School of Materials & Mineral Resources Engineering, Universiti Sains Malaysia, Nibong Tebal, 14300, Penang, Malaysia
| | | |
Collapse
|
7
|
Zhou X, Yang N, Xu W, Li X, Spiliopoulou A, Theodoratou E. Associations of genetic factors with vascular diabetes complications: an umbrella review. J Glob Health 2025; 15:04081. [PMID: 40116328 PMCID: PMC11927039 DOI: 10.7189/jogh.15.04081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025] Open
Abstract
Background To comprehensively assess evidence from published systematic review and meta-analyses (SRMAs) on the genetics of vascular diabetes complications. Methods A systematic literature search conducted in Medline and Embase identified 63 non-overlapping SRMAs. We re-conducted meta-analyses to compare diabetes with and without complications using multiple genetic models; evaluated associations using Venice criteria and Bayesian false-discovery probability (BFDP); and graded as highly credible, credible, and not credible. We also contrasted highly credible and credible associations to recent genome-wide association studies (GWASs). Results Highly credible evidence was discovered for single nucleotide polymorphisms (SNPs) rs1024611 at MCP-1 gene and SNP rs3025039 at VEGF gene with diabetic retinopathy (DR) in type 2 diabetes; SNP rs2268388 at ACACB gene, insertion/deletion (Ins/Del) variant at ACE gene, SNP rs1801133 at MTHFR gene, and SNP rs7903146 at TCF7L2 gene with diabetic kidney disease (DKD) in type 2 diabetes; and SNP rs4880 at SOD2 gene with diabetic peripheral neuropathy (DPN) in type 1 diabetes. Combining type 1 and 2 diabetes, highly credible evidence was discovered for insertion/deletion variant at ACE gene, SNP rs759853 at AKR1B1 gene, SNP rs1044498 at ENPP1 gene and DKD, and SNP rs1617640 at EPO gene for the combined endpoint of DR and DKD. None of these associations was directly replicated in the latest GWASs for DR and DKD, however, another SNP, rs55853916 at TCF7L2 gene had been detected as a GWAS hit for DKD. Conclusions This umbrella review rigorously assessed evidence on the genetics of vascular diabetes complications, complemented findings in recent GWASs and yielded insight into the optimal selection of genetic models for the design of GWASs on vascular diabetes complications. Mechanistic or bioinformatic studies are warranted to further assess the role of these genes in the pathology of vascular diabetes complications and their potential as drug targets. Registration PROSPERO: CRD42022384423.
Collapse
Affiliation(s)
- Xuan Zhou
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Nan Yang
- Department of Big Data in Health Science School of Public Health, and Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Xu
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, and Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Athina Spiliopoulou
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Evropi Theodoratou
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
8
|
Gao G, Su X, Liu S, Wang P, Chen JJ, Liu T, Xu J, Zhang Z, Zhang X, Xie Z. Cornuside as a promising therapeutic agent for diabetic kidney disease: Targeting regulation of Ca 2+ disorder-mediated renal tubular epithelial cells apoptosis. Int Immunopharmacol 2025; 149:114190. [PMID: 39904045 DOI: 10.1016/j.intimp.2025.114190] [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: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Renal tubular epithelial cells (RTECs) apoptosis is the key factor in the development of diabetic kidney disease (DKD). Endoplasmic reticulum stress (ERS) leading to mitochondrial Ca2+ overload is one of the causes of apoptosis in RTECs. Corni Fructus (CF) is an herbal medicine, developed and applied as a functional food, and it is commonly used to treat DKD. Cornuside (Cor) is one of the main chemical components in CF. This research seeks to investigate the function of Cor in DKD and delve into its possible mechanisms. Cor significantly improved renal function and ameliorated renal pathological changes of db/db mice. Bioinformatics analyses suggested that the modulation of endoplasmic reticulum-induced intrinsic apoptosis pathway was a primary mechanism by which Cor ameliorated DKD. TUNEL assays and flow cytometry assays indicated that Cor effectively inhibited RTECs apoptosis in db/db mice and AGE-induced HK-2 cells. Further experimental studies showed that Cor mitigated ERS by inhibiting the activation of PERK/ATF4/CHOP signal pathway and down-regulation of VDAC1 protein expression, thus alleviating mitochondrial Ca2+ overload. More importantly, Cor directly targeted NEDD4 to facilitate VDAC1 degradation. Notably, the silencing of NEDD4 nearly abolished Cor's inhibitory effects on mitochondrial Ca2+ overload and apoptosis. In conclusion, Cor modulated Ca2+ homeostasis by alleviating ERS and targeting NEDD4, thus mitigating apoptosis of RTECs in DKD. These findings indicate that Cor has the potential for the treatment and drug development of DKD.
Collapse
Affiliation(s)
- Gai Gao
- School of Pharmacy, Minzu University of China, Beijing, 100081, China; Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Henan Province, Henan University of Chinese Medicine, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, 450046, China
| | - Xuan Su
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Henan Province, Henan University of Chinese Medicine, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, 450046, China
| | - Shuyan Liu
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Henan Province, Henan University of Chinese Medicine, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, 450046, China
| | - Pan Wang
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Henan Province, Henan University of Chinese Medicine, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, 450046, China
| | - Jenny Jie Chen
- International Academic Affairs Department, Management and Science University, University Drive, Off Persiaran Olahraga, Section 13, 40100, Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Tongxiang Liu
- School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Jiangyan Xu
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Henan Province, Henan University of Chinese Medicine, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, 450046, China
| | - Zhenqiang Zhang
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Henan Province, Henan University of Chinese Medicine, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, 450046, China.
| | - Xiaowei Zhang
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Henan Province, Henan University of Chinese Medicine, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, 450046, China.
| | - Zhishen Xie
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Henan Province, Henan University of Chinese Medicine, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, 450046, China.
| |
Collapse
|
9
|
Li W, Xu G, Li M. Diabetic kidney disease: m6A modification as a marker of disease progression and subtype classification. Front Med (Lausanne) 2025; 12:1494162. [PMID: 40103797 PMCID: PMC11914134 DOI: 10.3389/fmed.2025.1494162] [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: 09/10/2024] [Accepted: 02/14/2025] [Indexed: 03/20/2025] Open
Abstract
This paper aims to investigate m6A modification during DKD progression. We evaluated m6A regulators expression in peripheral blood mononuclear cells, whole kidney tissue, glomerular, and tubulointerstitial samples. CIBERSORT and single-sample gene set enrichment analysis analyzed glomerular immune characteristics. Logistic-LASSO regression were used to develop the m6A regulators model that can identify early DKD. Consensus clustering algorithms were used to classify DKD in glomerular samples into m6A modified subtypes based on the expression of m6A regulators. Gene set variation analysis algorithm was used to evaluate the functional pathway enrichment of m6A modified subtypes. Weighted gene co-expression network analysis and protein-protein interaction networks identified m6A modified subtype marker genes. The Nephroseq V5 tool was used to evaluate the correlation between m6A modified subtypes marker genes and renal function. DKD patients' m6A regulators expression differed from the control group in various tissue types. DKD stages have various immune characteristics. The m6A regulators model with YTHDC1, METTL3, and ALKBH5 better identified early DKD. DKD was divided into two subtypes based on the expression of 26 m6A regulators. Subtype 1 was enriched in myogenesis, collagen components, and cytokine receptor interaction, while subtype 2 was enriched in protein secretion, proliferation, apoptosis, and various signaling pathways (e.g., TGFβ signaling pathway, PI3K/AKT/mTOR pathway, and etc.). Finally, AXIN1 and GOLGA4 were identified as possible biomarkers associated with glomerular filtration rate. From the viewpoint of m6A modification, the immune characteristics and molecular mechanisms of DKD at various stages are different, and targeted treatment would improve efficacy.
Collapse
Affiliation(s)
- Wenzhe Li
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Gaosi Xu
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Manna Li
- Department of Nephrology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
10
|
Liu J, Li Z, Zhang Z, Shen Z. Uncovering the mechanism of Huangkui capsule in the treatment of diabetic kidney disease based on network pharmacology and experimental validation. Sci Rep 2025; 15:6503. [PMID: 39987179 PMCID: PMC11846948 DOI: 10.1038/s41598-025-91264-w] [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: 04/22/2024] [Accepted: 02/19/2025] [Indexed: 02/24/2025] Open
Abstract
Diabetic kidney disease (DKD) is a main complication of diabetes mellitus. experimental in vitro validation and Network pharmacology were used in this study to explore the potential mechanism of Huangkui capsules (HKC) in treating DKD. First, we used CCK8 to analyze the optimal drug concentration of HKC. Next, we used flow cytometry, ELISA, Scratch test, and immunofluorescence to examine the apoptosis, oxidative stress, inflammatory factors, and fibrotic factors (FN and α-SMA) expression in HK-2 cells. Thereafter, in order to determine the potential molecular mechanisms underlying the therapeutic effect of HKC in DKD. Compounds contained in HKC were explored by UPLC-Q-TOF-MS/MS. SwissTargetPrediction was utilized for predicting potential gene targets of these compounds. OMIM, DisGeNet and GeneCards databases were employed to identify DKD-related genes. Meanwhile, the association of compounds with DKD genes was examined by protein-protein interaction, GO and KEGG analysis. Finally, molecular docking and molecular dynamics simulation were adopted for further validation. The results showed that HKC had 40 active ingredients, 1051 possible gene targets, and 133 DKD-HKC intersection genes. IL6, TNF, GAPDH, AKT1, PPARG, and TP53 were candidate hub genes by which HKC exerted its anti-DKD function based on molecular docking, molecular dynamics simulation and experimental results. To conclude, this study sheds more lights on the possible pharmacological activities of HKC in DKD and a foundation for further clinical application.
Collapse
Affiliation(s)
- Junhong Liu
- Department of Gastroenterology, The Fuyang Affiliated Hospital of Anhui Medical University, Fuyang, 236000, Anhui, China
| | - Ziwei Li
- Department of Clinical Nutrition, The Fuyang Affiliated Hospital of Anhui Medical University, Fuyang, 236000, Anhui, China
| | - ZongYao Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan, 232007, China
| | - Zhongyuan Shen
- Department of Radiology, The Fuyang Affiliated Hospital of Anhui Medical University, Fuyang, 236000, Anhui, China
| |
Collapse
|
11
|
Vishwanath R, Biswas A, Modi U, Gupta S, Bhatia D, Solanki R. Programmable short peptides for modulating stem cell fate in tissue engineering and regenerative medicine. J Mater Chem B 2025; 13:2573-2591. [PMID: 39871657 DOI: 10.1039/d4tb02102a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Recent advancements in tissue engineering and regenerative medicine have introduced promising strategies to address tissue and organ deficiencies. This review highlights the critical role of short peptides, particularly their ability to self-assemble into matrices that mimic the extracellular matrix (ECM). These low molecular weight peptides exhibit target-specific activities, modulate gene expression, and influence cell differentiation pathways. They are stable, programmable, non-cytotoxic, biocompatible, biodegradable, capable of crossing the cell membrane and easy to synthesize. This review underscores the importance of peptide structure and concentration in directing stem cell differentiation and explores their diverse biomedical applications. Peptides such as Aβ1-40, Aβ1-42, RADA16, A13 and KEDW are discussed for their roles in modulating stem cell differentiation into neuronal, glial, myocardial, osteogenic, hepatocyte and pancreatic lineages. Furthermore, this review delves into the underlying signaling mechanisms, the chemistry and design of short peptides and their potential for engineering biocompatible materials that mimic stem cell microenvironments. Short peptide-based biomaterials and scaffolds represent a promising avenue in stem cell therapy, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Rohan Vishwanath
- School of Life Science, Central University of Gujarat, Gandhinagar-382030, India
| | - Abhijit Biswas
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Unnati Modi
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Sharad Gupta
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
12
|
Ke J, Fan Y, Zhang S. Effects of PARP1 inhibitor PJ-34 on TGFα, IL-6, and IL-1β levels in diabetic nephropathy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:304-315. [PMID: 40073245 DOI: 10.1093/jimmun/vkae024] [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: 07/10/2024] [Accepted: 11/13/2024] [Indexed: 03/14/2025]
Abstract
Diabetic nephropathy is a severe chronic complication characterized by cytotoxicity, inflammation, and fibrosis, ultimately leading to renal failure. This study systematically investigated the effects of the PARP1 inhibitor PJ-34 on high glucose-induced cytotoxicity, inflammation, and fibrosis in HK-2 cells, as well as its improvement on neuropathic pain response and transforming growth factor β (TGFβ) expression in a type 1 diabetes mellitus diabetic nephropathy mouse model. Through cellular and animal experiments, we observed that PJ-34 significantly enhanced the proliferative capacity of cells damaged by high glucose, reduced apoptosis, and decreased the release of proinflammatory factors TGFα, interleukin-6, and interleukin-1β. In the type 1 diabetes mellitus nephropathy mouse model, the administration of PJ-34 substantially improved parameters of neuropathic pain, alleviated renal tissue damage, reduced indicators of renal functional impairment-inhibited renal fibrosis, and reduced the key protein expression in the epithelial-mesenchymal transition process, acting through the regulation of the TGFβ/Smads signaling pathway. This study elucidated the mechanism of action of the PARP1 inhibitor PJ-34 as a potential therapeutic agent for diabetic nephropathy, offering a novel strategy for its treatment.
Collapse
Affiliation(s)
- Jing Ke
- Department of Endocrinology, Central Hospital of Ezhou, Ezhou, China
| | - Yanan Fan
- Department of Thyroid and Breast Surgery, Central Hospital of Ezhou, Ezhou, China
| | - Shaochun Zhang
- Orthopedics Department, Central Hospital of Ezhou, Ezhou, China
| |
Collapse
|
13
|
Zhu LN, Xiang DL, Zuo JC, Wang GY, Xiao N. Potential of Klotho as a Biomarker for Overwork: A Study of Frontline Medical Workers. J Occup Environ Med 2025; 67:79-82. [PMID: 39805121 PMCID: PMC11801441 DOI: 10.1097/jom.0000000000003263] [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: 01/16/2025]
Abstract
OBJECTIVE This study evaluates the utility of serum s-αKlotho levels as a quantifiable biomarker for overwork. METHODS Frontline medical workers aged 20-55 years from Yiling People's Hospital of Yichang were recruited. Criteria included nonsmokers, non-heavy drinkers, no chronic medication use, and no acute illnesses recently. Participants worked over 10 hours per day, 60 hours weekly, and had at least 3 years of experience. A control group was matched except for work conditions. Data were collected through surveys, and serum levels were measured. RESULTS Significant differences in serum Klotho were found between overwork and control groups. The overwork group had higher median s-αKlotho levels (49.99 pg/mL) compared to controls (27.88 pg/mL). CONCLUSION Overworked medical workers exhibited elevated serum s-αKlotho, suggesting s-αKlotho as a potential biomarker for overwork. Future research should use multicenter designs with larger samples to validate findings.
Collapse
|
14
|
Zhao H, Zhang Z, Liu H, Ma M, Sun P, Zhao Y, Liu X. Multi-omics perspective: mechanisms of gastrointestinal injury repair. BURNS & TRAUMA 2025; 13:tkae057. [PMID: 39845194 PMCID: PMC11752642 DOI: 10.1093/burnst/tkae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 01/24/2025]
Abstract
In this review, we examine the significance of multi-omics technologies in understanding the plethora of intricate processes that activate gastrointestinal (GI) injury repair. Multi-omics, which includes genomics, transcriptomics, proteomics, and metabolomics, allows intricate mapping of cellular responses and molecular pathways involved in GI repair. We highlight the potential of multi-omics to discover previously unknown therapeutic targets or elucidate the molecular basis of the pathogenesis of GI. Furthermore, we explore the possibilities of integrating omics data to improve prediction models, and summarize the state-of-the-art technological developments and persisting obstacles that hinder the translation of multi-omics into clinical practice. Finally, innovative multi-omics approaches that can improve patient outcomes and advance therapeutic strategies in GI medicine are discussed.
Collapse
Affiliation(s)
- Haibin Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Zhigang Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Hongyu Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Mingxiu Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Peng Sun
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Yang Zhao
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Xun Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| |
Collapse
|
15
|
Fan X, Li J, Gao Y, Li L, Zhang H, Bi Z. The mechanism of enterogenous toxin methylmalonic acid aggravating calcium-phosphorus metabolic disorder in uremic rats by regulating the Wnt/β-catenin pathway. Mol Med 2025; 31:19. [PMID: 39844078 PMCID: PMC11756144 DOI: 10.1186/s10020-025-01067-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/03/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Uremia (UR) is caused by increased UR-related toxins in the bloodstream. We explored the mechanism of enterogenous toxin methylmalonic acid (MMA) in calcium-phosphorus metabolic disorder in UR rats via the Wnt/β-catenin pathway. METHODS The UR rat model was established by 5/6 nephrectomy. The fecal bacteria of UR rats were transplanted into Sham rats. Sham rats were injected with exogenous MMA or Salinomycin (SAL). Pathological changes in renal/colon tissues were analyzed. MMA concentration, levels of renal function indicators, serum inflammatory factors, Ca2+/P3+, and parathyroid hormone, intestinal flora structure, fecal metabolic profile, intestinal permeability, and glomerular filtration rate (GFR) were assessed. Additionally, rat glomerular podocytes were cultured, with cell viability and apoptosis measured. RESULTS Intestinal flora richness and diversity in UR rats were decreased, along with unbalanced flora structure. Among the screened 133 secondary differential metabolites, the MMA concentration rose, showing the most significant difference. UR rat fecal transplantation caused elevated MMA concentration in the serum and renal tissues of Sham rats. The intestinal flora metabolite MMA or exogenous MMA promoted intestinal barrier impairment, increased intestinal permeability, induced glomerular podocyte loss, and reduced GFR, causing calcium-phosphorus metabolic disorder. The intestinal flora metabolite MMA or exogenous MMA induced inflammatory responses and facilitated glomerular podocyte apoptosis by activating the Wnt/β-catenin pathway, which could be counteracted by repressing the Wnt/β-catenin pathway. CONCLUSIONS Enterogenous toxin MMA impelled intestinal barrier impairment in UR rats, enhanced intestinal permeability, and activated the Wnt/β-catenin pathway to induce glomerular podocyte loss and reduce GFR, thus aggravating calcium-phosphorus metabolic disorder.
Collapse
Affiliation(s)
- Xing Fan
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
| | - Jing Li
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
| | - Yan Gao
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China.
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China.
| | - Lin Li
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China.
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China.
| | - Haisong Zhang
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
| | - Zhaoyu Bi
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
| |
Collapse
|
16
|
Zhang Y, Wang F, Zhang C, Yao F, Zhang B, Zhang Y, Sun X. FGF21 ameliorates diabetic nephropathy through CDK1-dependently regulating the cell cycle. Front Pharmacol 2025; 15:1500458. [PMID: 39830349 PMCID: PMC11739279 DOI: 10.3389/fphar.2024.1500458] [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: 09/23/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
Background Diabetic nephropathy (DN) is a prevalent global renal illness and one of the main causes of end-stage renal disease (ESRD). FGF21 has been shown to ameliorate diabetic nephropathy, and in addition FGF-21-treated mice impeded mitogenicity, whereas it is unclear whether FGF21 can influence DN progression by regulating the cell cycle in diabetic nephropathy. Methods In order to create a diabetic model, STZ injections were given to C57BL/6J mice for this investigation. Then, FGF21 was administered, and renal tissue examination and pathological observation were combined with an assessment of glomerular injury, inflammation, oxidative stress, and the fibrinogen system in mice following the administration of the intervention. Furthermore, we used db/db mice and FGF21 direct therapy for 8 weeks to investigate changes in fasting glucose and creatinine expression as well as pathological changes in glomeruli glycogen deposition, fibrosis, and nephrin expression. To investigate the mechanism of action of FGF21 in the treatment of glycolytic kidney, transcriptome sequencing of renal tissues and KEGG pathway enrichment analysis of differential genes were performed. Results The study's findings demonstrated that FGF21 intervention increased clotting time, decreased oxidative stress and inflammation, and avoided thrombosis in addition to considerably improving glomerular filtration damage. After 8 weeks of FGF21 treatment, glomerular glycogen deposition, fibrosis, and renin expression decreased in db/db mice. Moreover, there was a notable reduction of creatinine and fasting blood glucose levels. Additionally, the CDK1 gene, a key player in controlling the cell cycle, was discovered through examination of the transcriptome sequencing data. It was also shown that FGF21 dramatically reduces the expression of CDK1, which may help diabetic nephropathy by averting mitotic catastrophe and changing the renal cell cycle. Conclusion In short, FGF21 improved the development of diabetic nephropathy in diabetic nephropathy-affected animals by reducing glomerular filtration damage, inflammation, and oxidative stress, inhibiting the formation of thrombus, and controlling the cell cycle through CDK1.
Collapse
Affiliation(s)
- Yudie Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Fan Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Chongyang Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Fan Yao
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Yongping Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
17
|
Trivedi MV, Jadhav HR, Gaikwad AB. Novel therapeutic targets for cardiorenal syndrome. Drug Discov Today 2025; 30:104285. [PMID: 39761847 DOI: 10.1016/j.drudis.2024.104285] [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: 11/02/2024] [Revised: 12/06/2024] [Accepted: 12/31/2024] [Indexed: 01/14/2025]
Abstract
Cardiorenal syndrome (CRS) is an interdependent dysfunction of the heart and kidneys, where failure in one organ precipitates failure in the other. The pathophysiology involves sustained renin-angiotensin-aldosterone-system (RAAS) activation, mitochondrial dysfunction, inflammation, fibrosis, oxidative stress and tissue remodeling, culminating in organ dysfunction. Existing therapies targeting the RAAS, diuretics and other agents have limitations, including diuretic resistance and compensatory sodium reabsorption. Therefore, there is a pressing need for novel druggable targets involved in CRS pathogenesis. This review addresses the challenges of existing treatments and emphasizes the importance of discovering new therapeutic targets. It highlights emerging targets such as Klotho, sex-determining region Y box 9 (SOX9), receptor-interacting protein kinase 3 (RIPK3), β-amino-isobutyric acid (BAIBA), thrombospondin-1 (TSP-1), among others, with their potential roles in CRS.
Collapse
Affiliation(s)
- Mansi Vinodkumar Trivedi
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Hemant R Jadhav
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
18
|
Li Y, Chen Y, Zhang H, Chen W, Pan Y. Liraglutide Ameliorates Renal Endothelial Dysfunction in Diabetic Rats Through the Inhibition of the Dll4/Notch2 Pathway. Diabetes Metab Syndr Obes 2024; 17:4091-4104. [PMID: 39492960 PMCID: PMC11531755 DOI: 10.2147/dmso.s492252] [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: 08/20/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Purpose The glucagon-like peptide-1 receptor agonist (GLP-1RA) is a pharmacological agent utilized for the treatment of diabetes, known for its significant reno protective effects. This study aims to investigate the impact of liraglutide, a representative GLP-1RA medication, on early endothelial dysfunction in diabetic rats and elucidate its underlying mechanisms. Methods The present study employed a high-fat, high-sugar diet in combination with a single intraperitoneal injection of streptozotocin (STZ) to establish an experimental rat model of diabetes. Subsequently, the therapeutic efficacy of liraglutide on renal injury in this model was evaluated using various doses. Results Compared to the DKD rats, the rats treated with Liraglutide exhibited significant reductions in levels of blood glucose (Glu), serum creatinine (Scr), and blood urea nitrogen (BUN) (P < 0.05). Furthermore, there was a dose-dependent decrease in urinary protein levels, including 24-hour urinary protein excretion rate and microalbuminuria (m-ALB), with higher doses demonstrating more pronounced therapeutic effects (P <0.05). In addition, treatment with Liraglutide effectively improved glomerular and interstitial damage, and suppressed the expression of CD31, CD34, and VE-cadherin associated with endothelial cell injury (P < 0.05). Furthermore, Liraglutide administration significantly increased nitric oxide (NO) production (P < 0.05). Moreover, Liraglutide treatment resulted in decreased expression of vascular endothelial growth factor (VEGF), Delta-like ligand-4(Dll4), and Notch2 protein in the Notch2 signaling pathway (P < 0.05). Conclusion The findings indicate that Liraglutide has a substantial effect on decreasing urinary protein excretion and improving vascular microinflammation, thus alleviating endothelial dysfunction in diabetic nephropathy. This observed mechanism can be attributed to the inhibition of the Dll4/Notch2 signaling pathway.
Collapse
Affiliation(s)
- Yining Li
- Department of Nephrology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, People’s Republic of China
| | - Yulin Chen
- Department of Nephrology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, People’s Republic of China
| | - Hui Zhang
- Department of Nephrology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, People’s Republic of China
| | - Weidong Chen
- Department of Nephrology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, People’s Republic of China
| | - Yan Pan
- Department of Nephrology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, People’s Republic of China
| |
Collapse
|
19
|
Chen X, Wei Y, Li Z, Zhou C, Fan Y. Distinct role of Klotho in long bone and craniofacial bone: skeletal development, repair and regeneration. PeerJ 2024; 12:e18269. [PMID: 39465174 PMCID: PMC11505971 DOI: 10.7717/peerj.18269] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/17/2024] [Indexed: 10/29/2024] Open
Abstract
Bone defects are highly prevalent diseases caused by trauma, tumors, inflammation, congenital malformations and endocrine abnormalities. Ideally effective and side effect free approach to dealing with bone defects remains a clinical conundrum. Klotho is an important protein, which plays an essential role in regulating aging and mineral ion homeostasis. More recently, research revealed the function of Klotho in regulating skeleton development and regeneration. Klotho has been identified in mesenchymal stem cells, osteoblasts, osteocytes and osteoclasts in different skeleton regions. The specific function and regulatory mechanisms of Klotho in long bone and craniofacial bone vary due to their different embryonic development, ossification and cell types, which remain unclear and without conclusion. Moreover, studies have confirmed that Klotho is a multifunctional protein that can inhibit inflammation, resist cancer and regulate the endocrine system, which may further accentuate the potential of Klotho to be the ideal molecule in inducing bone restoration clinically. Besides, as an endogenous protein, Klotho has a promising potential for clinical therapy without side effects. In the current review, we summarized the specific function of Klotho in long bone and craniofacial skeleton from phenotype to cellular alternation and signaling pathway. Moreover, we illustrated the possible future clinical application for Klotho. Further research on Klotho might help to solve the existing clinical difficulties in bone healing and increase the life quality of patients with bone injury and the elderly.
Collapse
Affiliation(s)
- Xinyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yali Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zucen Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Xiang W, Li L, Qin M, Li L, Yu H, Wang F, Ni S, Shen A, Lu H, Ni H, Wang Y. Diminished nuclear-localized β-adrenoceptor signalling activates YAP to promote kidney fibrosis in diabetic nephropathy. Br J Pharmacol 2024. [PMID: 39359016 DOI: 10.1111/bph.17347] [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: 12/31/2023] [Revised: 05/27/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND AND PURPOSE Diabetic nephropathy (DN) is a leading cause of chronic kidney disease (CKD), which is characterized by mesangial matrix expansion that involves dysfunctional mesangial cells (MCs). However, the underlying mechanisms remain unclear. This study aims to delineate the spatiotemporal contribution of adrenergic signalling in diabetic kidney fibrosis to reveal potential therapeutic targets. EXPERIMENTAL APPROACH A model of diabetic nephropathy was induced by in db/db mice. Gene expression in kidneys was profiled by RNA-seq analyses, western blot and immunostaining. Subcellular-localized fluorescence resonance energy transfer (FRET) biosensors determined adrenergic signalling microdomains in MCs. Effects of oral rolipram, a phosphodiesterase 4 (PDE4) inhibitor, on the model were measured. KEY RESULTS Our model exhibited impaired kidney function with elevated expression of adrenergic and fibrotic genes, including Adrb1, PDEs, Acta2 and Tgfβ. RNA-seq analysis revealed that MCs with dysregulated YAP pathway were crucial to the extracellular matrix secretion in kidneys from diabetic nephropathy patients. In cultured MCs, TGF-β promoted profibrotic gene transcription, which was regulated by nuclear-localized β-adrenoceptor signalling. Mechanistically, TGF-β treatment diminished nuclear-specific cAMP signalling in MCs and reduced PKA-dependent phosphorylation of YAP, leading to its activation. In parallel, db/db mouse kidneys showed increased expressions of PDE4B and PDE4D. Treatment with oral rolipram alleviated kidney fibrosis in db/db mice. CONCLUSION AND IMPLICATIONS Diabetic nephropathy impaired nuclear-localized β1-adrenoceptor-cAMP signalling microdomain through upregulating PDE4 expression, promoting fibrosis in MCs via PKA dephosphorylation-dependent YAP activation. Our results suggest PDE4 inhibition as a promising strategy for alleviating kidney fibrosis in diabetic nephropathy.
Collapse
Affiliation(s)
- Wenjing Xiang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lei Li
- School of Public Health, Xi'an Jiao Tong University, Xi'an, China
| | - Manman Qin
- Mass Spectrometry Laboratory for BioSample analysis, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Lei Li
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hualong Yu
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Fangyuan Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Siyuan Ni
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Ao Shen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease School of Pharmaceutical Sciences & The Fifth Affiliated Hospital Guangzhou Medical University, Guangzhou, China
| | - Haocheng Lu
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Haibo Ni
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, Shenzhen, China
| |
Collapse
|
21
|
Grigore TV, Zuidscherwoude M, Olauson H, Hoenderop JG. Lessons from Klotho mouse models to understand mineral homeostasis. Acta Physiol (Oxf) 2024; 240:e14220. [PMID: 39176993 DOI: 10.1111/apha.14220] [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: 05/31/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
AIM Klotho, a key component of the endocrine fibroblast growth factor receptor-fibroblast growth factor axis, is a multi-functional protein that impacts renal electrolyte handling. The physiological significance of Klotho will be highlighted in the regulation of calcium, phosphate, and potassium metabolism. METHODS In this review, we compare several murine models with different renal targeted deletions of Klotho and the insights into the molecular and physiological function that these models offer. RESULTS In vivo, Klotho deficiency is associated with severely impaired mineral metabolism, with consequences on growth, longevity and disease development. Additionally, we explore the perspectives of Klotho in renal pathology and vascular events, as well as potential Klotho treatment options. CONCLUSION This comprehensive review emphasizes the use of Klotho to shed light on deciphering the renal molecular in vivo mechanisms in electrolyte handling, as well as novel therapeutic interventions.
Collapse
Affiliation(s)
- Teodora V Grigore
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Malou Zuidscherwoude
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hannes Olauson
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joost G Hoenderop
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
22
|
Prud’homme GJ, Wang Q. Anti-Inflammatory Role of the Klotho Protein and Relevance to Aging. Cells 2024; 13:1413. [PMID: 39272986 PMCID: PMC11394293 DOI: 10.3390/cells13171413] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The α-Klotho protein (hereafter Klotho) is an obligate coreceptor for fibroblast growth factor 23 (FGF23). It is produced in the kidneys, brain and other sites. Klotho insufficiency causes hyperphosphatemia and other anomalies. Importantly, it is associated with chronic pathologies (often age-related) that have an inflammatory component. This includes atherosclerosis, diabetes and Alzheimer's disease. Its mode of action in these diseases is not well understood, but it inhibits or regulates multiple major pathways. Klotho has a membrane form and a soluble form (s-Klotho). Cytosolic Klotho is postulated but not well characterized. s-Klotho has endocrine properties that are incompletely elucidated. It binds to the FGF receptor 1c (FGFR1c) that is widely expressed (including endothelial cells). It also attaches to soluble FGF23, and FGF23/Klotho binds to FGFRs. Thus, s-Klotho might be a roaming FGF23 coreceptor, but it has other functions. Notably, Klotho (cell-bound or soluble) counteracts inflammation and appears to mitigate related aging (inflammaging). It inhibits NF-κB and the NLRP3 inflammasome. This inflammasome requires priming by NF-κB and produces active IL-1β, membrane pores and cell death (pyroptosis). In accord, Klotho countered inflammation and cell injury induced by toxins, damage-associated molecular patterns (DAMPs), cytokines, and reactive oxygen species (ROS). s-Klotho also blocks the TGF-β receptor and Wnt ligands, which lessens fibrotic disease. Low Klotho is associated with loss of muscle mass (sarcopenia), as occurs in aging and chronic diseases. s-Klotho counters the inhibitory effects of myostatin and TGF-β on muscle, reduces inflammation, and improves muscle repair following injury. The inhibition of TGF-β and other factors may also be protective in diabetic retinopathy and age-related macular degeneration (AMD). This review examines Klotho functions especially as related to inflammation and potential applications.
Collapse
Affiliation(s)
- Gérald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 220 Walmer Rd, Toronto, ON M5R 3R7, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200030, China
- Shanghai Innogen Pharmaceutical Co., Ltd., Shanghai 201318, China
| |
Collapse
|
23
|
Asadi R, Shadpour P, Nakhaei A. Non-dialyzable uremic toxins and renal tubular cell damage in CKD patients: a systems biology approach. Eur J Med Res 2024; 29:412. [PMID: 39123228 PMCID: PMC11311939 DOI: 10.1186/s40001-024-01951-z] [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: 08/02/2023] [Accepted: 06/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Chronic kidney disease presents global health challenges, with hemodialysis as a common treatment. However, non-dialyzable uremic toxins demand further investigation for new therapeutic approaches. Renal tubular cells require scrutiny due to their vulnerability to uremic toxins. METHODS In this study, a systems biology approach utilized transcriptomics data from healthy renal tubular cells exposed to healthy and post-dialysis uremic plasma. RESULTS Differential gene expression analysis identified 983 up-regulated genes, including 70 essential proteins in the protein-protein interaction network. Modularity-based clustering revealed six clusters of essential proteins associated with 11 pathological pathways activated in response to non-dialyzable uremic toxins. CONCLUSIONS Notably, WNT1/11, AGT, FGF4/17/22, LMX1B, GATA4, and CXCL12 emerged as promising targets for further exploration in renal tubular pathology related to non-dialyzable uremic toxins. Understanding the molecular players and pathways linked to renal tubular dysfunction opens avenues for novel therapeutic interventions and improved clinical management of chronic kidney disease and its complications.
Collapse
Affiliation(s)
- Roya Asadi
- Industrial Engineering Department, Faculty of Technical and Engineering, University of Science and Culture (USC), Tehran, Iran
| | - Pejman Shadpour
- Hospital Management Research Center (HMRC), Hasheminejad Kidney Center (HKC), Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Akram Nakhaei
- Computer Engineering Department, Mazandaran University of Science and Technology (MUST), Babol, Iran.
| |
Collapse
|
24
|
Jasper AA, Shah KH, Karim H, Gujral S, Miljkovic I, Rosano C, Barchowsky A, Sahu A. Regenerative rehabilitation measures to restore tissue function after arsenic exposure. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2024; 30:100529. [PMID: 40191583 PMCID: PMC11970924 DOI: 10.1016/j.cobme.2024.100529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Environmental exposure of arsenic impairs the cardiometabolic profile, skeletal muscle health, and neurological function. Such declining tissue health is observed as early as in one's childhood, where the exposure is prevalent, thereby accelerating the effect of time's arrow. Despite the known deleterious effects of arsenic exposure, there is a paucity of specific treatment plans for restoring tissue function in exposed individuals. In this review, we propose to harness the untapped potential of existing regenerative rehabilitation programs, such as stem cell therapeutics with rehabilitation, acellular therapeutics, and artificial intelligence/robotics technologies, to address this critical gap in environmental toxicology. With regenerative rehabilitation techniques showing promise in other injury paradigms, fostering collaboration between these scientific realms offers an effective means of mitigating the detrimental effects of arsenic on tissue function.
Collapse
Affiliation(s)
- Adam A Jasper
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - Kush H Shah
- The Lake Erie College of Osteopathic Medicine (LECOM), Erie, PA, USA
| | - Helmet Karim
- Department of Psychiatry, University of Pittsburgh, USA
- Department of Bioengineering, University of Pittsburgh, USA
| | - Swathi Gujral
- Department of Psychiatry, University of Pittsburgh, USA
| | - Iva Miljkovic
- Department of Epidemiology, University of Pittsburgh, USA
| | | | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| |
Collapse
|
25
|
Hosseininasab SS, Dhiaa SM, Shahrtash SA, Lak M, Faghihkhorasani A, Mahdi F. The interaction between klotho protein and epigenetic alteration in diabetes and treatment options. J Diabetes Metab Disord 2024; 23:333-341. [PMID: 38932867 PMCID: PMC11196449 DOI: 10.1007/s40200-024-01387-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/11/2024] [Indexed: 06/28/2024]
Abstract
Introduction Klotho is a membrane protein predominantly expressed in the kidneys, and its discovery was serendipitously made through gene-targeting experiments conducted on mice. Klotho has a favorable role in the regulation of multiple cellular processes, such as aging, oxidative stress, inflammation, and apoptosis. This regulation occurs through the targeting of diverse signaling molecules, cell membrane receptors, and ion channels, achieved by physical contacts or enzymatic activities of Klotho. This review examines the role of Klotho in the epigenetic regulation of molecules associated with diabetes. Methods Authors conducted a thorough literature search using the PubMed®, Web of Science™, and Scopus®. Relevant articles up to September 2023, published in the English language were considered. We reviewed research databases searching for studies that included keywords klotho, epigenetic, and diabetes. Results 14 related papers about epigenetic modification of proteins involved in diabetes pathogenesis were selected to be included in this narrative review. In the studies, the kidney was the most investigated organ regarding this correlation. Also, phosphorylation and methylation were the common epigenetic modifications of proteins by Klotho. Conclusion Klotho has a significant role in the maturation of adipocytes and the regulation of systemic glucose metabolism, exhibiting a strong association with the pathogenesis of diabetes. Both epigenetic alterations and the modulation of protein phosphorylation by Klotho play significant roles in the regulation of Klotho expression and the modulation of other molecules implicated in the etiology of diabetes.
Collapse
Affiliation(s)
| | | | | | - Mehrnoosh Lak
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Mahdi
- Department of Internal Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
26
|
Chen X, Xiao J, Tao D, Liang Y, Chen S, Shen L, Li S, Zheng Z, Zeng Y, Luo C, Peng F, Long H. Metadherin orchestrates PKA and PKM2 to activate β-catenin signaling in podocytes during proteinuric chronic kidney disease. Transl Res 2024; 266:68-83. [PMID: 37995969 DOI: 10.1016/j.trsl.2023.11.006] [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: 07/15/2023] [Revised: 08/23/2023] [Accepted: 11/21/2023] [Indexed: 11/25/2023]
Abstract
Podocyte damage is the major cause of glomerular injury and proteinuria in multiple chronic kidney diseases. Metadherin (MTDH) is involved in podocyte apoptosis and promotes renal tubular injury in mouse models of diabetic nephropathy and renal fibrosis; however, its role in podocyte injury and proteinuria needs further exploration. Here, we show that MTDH was induced in the glomerular podocytes of patients with proteinuric chronic kidney disease and correlated with proteinuria. Podocyte-specific knockout of MTDH in mice reversed proteinuria, attenuated podocyte injury, and prevented glomerulosclerosis after advanced oxidation protein products challenge or adriamycin injury. Furthermore, specific knockout of MTDH in podocytes repressed β-catenin phosphorylation at the Ser675 site and inhibited its downstream target gene transcription. Mechanistically, on the one hand, MTDH increased cAMP and then activated protein kinase A (PKA) to induce β-catenin phosphorylation at the Ser675 site, facilitating the nuclear translocation of MTDH and β-catenin; on the other hand, MTDH induced the deaggregation of pyruvate kinase M2 (PKM2) tetramers and promoted PKM2 monomers to enter the nucleus. This cascade of events leads to the formation of the MTDH/PKM2/β-catenin/CBP/TCF4 transcription complex, thus triggering TCF4-dependent gene transcription. Inhibition of PKA activity by H-89 or blockade of PKM2 deaggregation by TEPP-46 abolished this cascade of events and disrupted transcription complex formation. These results suggest that MTDH induces podocyte injury and proteinuria by assembling the β-catenin-mediated transcription complex by regulating PKA and PKM2 function.
Collapse
Affiliation(s)
- Xiaowen Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Jing Xiao
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Danping Tao
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Department of Gerontology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yunyi Liang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Sijia Chen
- Department of Nephrology and Rheumatology, The First Hospital of Changsha, Changsha, China
| | - Lingyu Shen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zerong Zheng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yao Zeng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
27
|
Luo Y, Zhang L, Su N, Liu L, Zhao T. YME1L-mediated mitophagy protects renal tubular cells against cellular senescence under diabetic conditions. Biol Res 2024; 57:10. [PMID: 38494498 PMCID: PMC10946153 DOI: 10.1186/s40659-024-00487-0] [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: 08/19/2023] [Accepted: 02/27/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND The senescence of renal tubular epithelial cells (RTECs) is crucial in the progression of diabetic kidney disease (DKD). Accumulating evidence suggests a close association between insufficient mitophagy and RTEC senescence. Yeast mitochondrial escape 1-like 1 (YME1L), an inner mitochondrial membrane metalloprotease, maintains mitochondrial integrity. Its functions in DKD remain unclear. Here, we investigated whether YME1L can prevent the progression of DKD by regulating mitophagy and cellular senescence. METHODS We analyzed YME1L expression in renal tubules of DKD patients and mice, explored transcriptomic changes associated with YME1L overexpression in RTECs, and assessed its impact on RTEC senescence and renal dysfunction using an HFD/STZ-induced DKD mouse model. Tubule-specific overexpression of YME1L was achieved through the use of recombinant adeno-associated virus 2/9 (rAAV 2/9). We conducted both in vivo and in vitro experiments to evaluate the effects of YME1L overexpression on mitophagy and mitochondrial function. Furthermore, we performed LC-MS/MS analysis to identify potential protein interactions involving YME1L and elucidate the underlying mechanisms. RESULTS Our findings revealed a significant decrease in YME1L expression in the renal tubules of DKD patients and mice. However, tubule-specific overexpression of YME1L significantly alleviated RTEC senescence and renal dysfunction in the HFD/STZ-induced DKD mouse model. Moreover, YME1L overexpression exhibited positive effects on enhancing mitophagy and improving mitochondrial function both in vivo and in vitro. Mechanistically, our LC-MS/MS analysis uncovered a crucial mitophagy receptor, BCL2-like 13 (BCL2L13), as an interacting partner of YME1L. Furthermore, YME1L was found to promote the phosphorylation of BCL2L13, highlighting its role in regulating mitophagy. CONCLUSIONS This study provides compelling evidence that YME1L plays a critical role in protecting RTECs from cellular senescence and impeding the progression of DKD. Overexpression of YME1L demonstrated significant therapeutic potential by ameliorating both RTEC senescence and renal dysfunction in the DKD mice. Moreover, our findings indicate that YME1L enhances mitophagy and improves mitochondrial function, potentially through its interaction with BCL2L13 and subsequent phosphorylation. These novel insights into the protective mechanisms of YME1L offer a promising strategy for developing therapies targeting DKD.
Collapse
Affiliation(s)
- Yuanyuan Luo
- Department of Endocrinology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Department of Endocrinology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Lingxiao Zhang
- Department of Endocrinology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Ning Su
- Department of Hematology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Lerong Liu
- Department of Endocrinology, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Tongfeng Zhao
- Department of Endocrinology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Department of Endocrinology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 51000, China.
| |
Collapse
|
28
|
Wang H, Liu H, Cheng H, Xue X, Ge Y, Wang X, Yuan J. Klotho Stabilizes the Podocyte Actin Cytoskeleton in Idiopathic Membranous Nephropathy through Regulating the TRPC6/CatL Pathway. Am J Nephrol 2024; 55:345-360. [PMID: 38330925 PMCID: PMC11152006 DOI: 10.1159/000537732] [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: 10/13/2023] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
INTRODUCTION The aim of this study was to explore the renoprotective effects of Klotho on podocyte injury mediated by complement activation and autoantibodies in idiopathic membranous nephropathy (IMN). METHODS Rat passive Heymann nephritis (PHN) was induced as an IMN model. Urine protein levels, serum biochemistry, kidney histology, and podocyte marker levels were assessed. In vitro, sublytic podocyte injury was induced by C5b-9. The expression of Klotho, transient receptor potential channel 6 (TRPC6), and cathepsin L (CatL); its substrate synaptopodin; and the intracellular Ca2+ concentration were detected via immunofluorescence. RhoA/ROCK pathway activity was measured by an activity quantitative detection kit, and the protein expression of phosphorylated-LIMK1 (p-LIMK1) and p-cofilin in podocytes was detected via Western blotting. Klotho knockdown and overexpression were performed to evaluate its role in regulating the TRPC6/CatL pathway. RESULTS PHN rats exhibited proteinuria, podocyte foot process effacement, decreased Klotho and Synaptopodin levels, and increased TRPC6 and CatL expression. The RhoA/ROCK pathway was activated by the increased phosphorylation of LIMK1 and cofilin. Similar changes were observed in C5b-9-injured podocytes. Klotho knockdown exacerbated podocyte injury, while Klotho overexpression partially ameliorated podocyte injury. CONCLUSION Klotho may protect against podocyte injury in IMN patients by inhibiting the TRPC6/CatL pathway. Klotho is a potential target for reducing proteinuria in IMN patients.
Collapse
Affiliation(s)
- Hongyun Wang
- Hubei University of Chinese Medicine, Wuhan, China
| | - Hongyan Liu
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hong Cheng
- Department of Nephrology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| | - Xue Xue
- Hubei University of Chinese Medicine, Wuhan, China
| | - Yamei Ge
- Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaoqin Wang
- Department of Nephrology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| | - Jun Yuan
- Hubei University of Chinese Medicine, Wuhan, China
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
29
|
Hadpech S, Thongboonkerd V. Epithelial-mesenchymal plasticity in kidney fibrosis. Genesis 2024; 62:e23529. [PMID: 37345818 DOI: 10.1002/dvg.23529] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is an important biological process contributing to kidney fibrosis and chronic kidney disease. This process is characterized by decreased epithelial phenotypes/markers and increased mesenchymal phenotypes/markers. Tubular epithelial cells (TECs) are commonly susceptible to EMT by various stimuli, for example, transforming growth factor-β (TGF-β), cellular communication network factor 2, angiotensin-II, fibroblast growth factor-2, oncostatin M, matrix metalloproteinase-2, tissue plasminogen activator (t-PA), plasmin, interleukin-1β, and reactive oxygen species. Similarly, glomerular podocytes can undergo EMT via these stimuli and by high glucose condition in diabetic kidney disease. EMT of TECs and podocytes leads to tubulointerstitial fibrosis and glomerulosclerosis, respectively. Signaling pathways involved in EMT-mediated kidney fibrosis are diverse and complex. TGF-β1/Smad and Wnt/β-catenin pathways are the major venues triggering EMT in TECs and podocytes. These two pathways thus serve as the major therapeutic targets against EMT-mediated kidney fibrosis. To date, a number of EMT inhibitors have been identified and characterized. As expected, the majority of these EMT inhibitors affect TGF-β1/Smad and Wnt/β-catenin pathways. In addition to kidney fibrosis, these EMT-targeted antifibrotic inhibitors are expected to be effective for treatment against fibrosis in other organs/tissues.
Collapse
Affiliation(s)
- Sudarat Hadpech
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
30
|
Xu J, Lin E, Hong X, Li L, Gu J, Zhao J, Liu Y. Klotho-derived peptide KP1 ameliorates SARS-CoV-2-associated acute kidney injury. Front Pharmacol 2024; 14:1333389. [PMID: 38239193 PMCID: PMC10795167 DOI: 10.3389/fphar.2023.1333389] [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: 11/07/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction: The severe cases of COVID-19, a disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), often present with acute kidney injury (AKI). Although old age and preexisting medical conditions have been identified as principal risk factors for COVID-19-associated AKI, the molecular basis behind such a connection remains unknown. In this study, we investigated the pathogenic role of Klotho deficiency in COVID-19-associated AKI and explored the therapeutic potential of Klotho-derived peptide 1 (KP1). Methods: We assessed the susceptibility of Klotho deficient Kl/Kl mice to developing AKI after expression of SARS-CoV-2 N protein. The role of KP1 in ameliorating tubular injury was investigated by using cultured proximal tubular cells (HK-2) in vitro and mouse model of ischemia-reperfusion injury (IRI) in vivo. Results: Renal Klotho expression was markedly downregulated in various chronic kidney disease (CKD) models and in aged mice. Compared to wild-type counterparts, mutant KL/KL mice were susceptible to overexpression of SARS-CoV-2 N protein and developed kidney lesions resembling AKI. In vitro, expression of N protein alone induced HK-2 cells to express markers of tubular injury, cellular senescence, apoptosis and epithelial-mesenchymal transition, whereas both KP1 and Klotho abolished these lesions. Furthermore, KP1 mitigated kidney dysfunction, alleviated tubular injury and inhibited apoptosis in AKI model induced by IRI and N protein. Conclusion: These findings suggest that Klotho deficiency is a key determinant of developing COVID-19-associated AKI. As such, KP1, a small peptide recapitulating Klotho function, could be an effective therapeutic for alleviating AKI in COVID-19 patients.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Enqing Lin
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Xue Hong
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Li Li
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Jun Gu
- State Key Laboratory of Protein and Plant Gene Research, College of Life Science, Peking University, Beijing, China
| | - Jinghong Zhao
- Division of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
31
|
Abboud M, Merenbakh-Lamin K, Volkov H, Ben-Neriah S, Ligumsky H, Bronfeld S, Keren-Khadmy N, Giladi M, Shomron N, Wolf I, Rubinek T. Revealing the tumor suppressive sequence within KL1 domain of the hormone Klotho. Oncogene 2024; 43:354-362. [PMID: 38040805 DOI: 10.1038/s41388-023-02904-2] [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: 08/16/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
Klotho, a 1012 amino acid transmembrane protein, is a potent tumor suppressor in different cancer types. Klotho is composed of two internal repeats KL1 and KL2, and the tumor suppressor activity is primarily attributed to the KL1 domain. Despite its significant role in regulating various cancer-related pathways, the precise mechanism underlying its tumor suppressor activity remains unresolved. In this study, we aimed to identify the sequence responsible for the tumor suppressor function of Klotho and gain insights into its mechanism of action. To accomplish this, we generated expression vectors of truncated KL1 at the C and N-terminal regions and evaluated their ability to inhibit the colony formation of several cancer cell lines. Our findings demonstrated that truncated KL1 1-340 (KL340) effectively inhibited colony formation similar to KL1, while truncated KL1 1-320 (KL320) lost this activity. Furthermore, this correlated with the inhibitory effect of KL1 and KL340 on the Wnt/β-catenin pathway, whereas KL320 had no effect. Transcriptomic analysis of MCF-7 cells expressing the constructs revealed enriched pathways associated with tumor suppressor activity in KL1 and KL340. Interestingly, the α-fold predictor tool highlighted distinct differences in the α and β sheets of the TIM barrel fold of the truncated Klotho constructs, adding to our understanding of their structural variations. In summary, this study identified the 340 N-terminal amino acids as the sequence that possesses Klotho's tumor suppressor activity and reveals a critical role in the 320-340 sequence for this function. It also provides a foundation for the development of Klotho-based therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Marana Abboud
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | | | - Hadas Volkov
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Edmond J. Safra Center for Bioinformatics at Tel-Aviv University, Tel Aviv, Israel
| | - Shira Ben-Neriah
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Hagai Ligumsky
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Sarai Bronfeld
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noa Keren-Khadmy
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Moshe Giladi
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Internal Medicine Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Noam Shomron
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Edmond J. Safra Center for Bioinformatics at Tel-Aviv University, Tel Aviv, Israel
| | - Ido Wolf
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tami Rubinek
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
32
|
Dusso A, Bauerle KT, Zhang RM, Bernal-Mizrachi C. Vitamin D and renal disease. FELDMAN AND PIKE'S VITAMIN D 2024:587-618. [DOI: 10.1016/b978-0-323-91338-6.00029-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
33
|
Roghani M, Golchoobian R, Mohammadian M, Shanehbandpour-Tabari F, Salehi Z, Gilaki-Bisheh S. Time-Dependent Molecular Changes Following MDMA-Induced Nephrotoxicity. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2024; 23:e145483. [PMID: 39830664 PMCID: PMC11742579 DOI: 10.5812/ijpr-145483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/04/2024] [Accepted: 04/13/2024] [Indexed: 01/22/2025]
Abstract
The increasing recreational use of ecstasy (MDMA) poses significant risks to human health, including reports of fatal renal failure due to its adverse renal effects. While MDMA-induced renal toxicity might result from systemic effects, there is also substantial evidence of direct harm to renal tissues by MDMA or its metabolites. The precise mechanisms underlying renal toxicity remain unclear. This study explored the impact of a single intraperitoneal dose of MDMA (20 mg/kg) on rat kidneys. Serum BUN and creatinine levels were evaluated to assess renal function, while TNF-α and TGF-β protein concentrations were measured using ELISA. mRNA levels of Bax, Bcl-xl, and Bcl-2 were quantified using quantitative RT-PCR. Additionally, apoptosis and histopathological changes in renal tissue were examined. Results showed a transient increase in serum BUN and creatinine in MDMA-treated rats. There were decreases in TNF-α and TGF-β levels in the renal tissue. Both pro-apoptotic Bax and anti-apoptotic Bcl-xl gene expressions were significantly reduced, whereas Bcl-2 expression and apoptosis did not show significant changes. No structural alterations were observed in the renal tissues. Overall, this study suggests that the renal adverse effects of MDMA may be mediated through the disruption of cytokine pathways, with notable reductions in TGF-β possibly linked to decreased TNF-α levels.
Collapse
Affiliation(s)
- Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| | - Ravieh Golchoobian
- Department of Physiology, Babol University of Medical Sciences, Babol, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Mohammadian
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farzane Shanehbandpour-Tabari
- Department of Physiology, Babol University of Medical Sciences, Babol, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Saba Gilaki-Bisheh
- Department of Pathology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Liu Y, Lu X, Chen M, Wei Z, Peng G, Yang J, Tang C, Yu P. Advances in screening, synthesis, modification, and biomedical applications of peptides and peptide aptamers. Biofactors 2024; 50:33-57. [PMID: 37646383 DOI: 10.1002/biof.2001] [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: 05/29/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023]
Abstract
Peptides and peptide aptamers have emerged as promising molecules for a wide range of biomedical applications due to their unique properties and versatile functionalities. The screening strategies for identifying peptides and peptide aptamers with desired properties are discussed, including high-throughput screening, display screening technology, and in silico design approaches. The synthesis methods for the efficient production of peptides and peptide aptamers, such as solid-phase peptide synthesis and biosynthesis technology, are described, along with their advantages and limitations. Moreover, various modification techniques are explored to enhance the stability, specificity, and pharmacokinetic properties of peptides and peptide aptamers. This includes chemical modifications, enzymatic modifications, biomodifications, genetic engineering modifications, and physical modifications. Furthermore, the review highlights the diverse biomedical applications of peptides and peptide aptamers, including targeted drug delivery, diagnostics, and therapeutic. This review provides valuable insights into the advancements in screening, synthesis, modification, and biomedical applications of peptides and peptide aptamers. A comprehensive understanding of these aspects will aid researchers in the development of novel peptide-based therapeutics and diagnostic tools for various biomedical challenges.
Collapse
Affiliation(s)
- Yijie Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xiaoling Lu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Meilun Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zheng Wei
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Guangnan Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Jie Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Chunhua Tang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Peng Yu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
35
|
Zhang C, Liu L, Li W, Li M, Zhang X, Zhang C, Yang H, Xie J, Pan W, Guo X, She P, Zhong L, Li T. Upregulation of FAM83F by c-Myc promotes cervical cancer growth and aerobic glycolysis via Wnt/β-catenin signaling activation. Cell Death Dis 2023; 14:837. [PMID: 38104106 PMCID: PMC10725447 DOI: 10.1038/s41419-023-06377-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Cervical cancer (CC) seriously affects women's health. Therefore, elucidation of the exact mechanisms and identification of novel therapeutic targets are urgently needed. In this study, we identified FAM83F, which was highly expressed in CC cells and tissues, as a potential target. Our clinical data revealed that FAM83F protein expression was markedly elevated in CC tissues and was positively correlated with poor prognosis. Moreover, we observed that FAM83F knockdown significantly inhibited cell proliferation, induced apoptosis, and suppressed glycolysis in CC cells, while its overexpression displayed opposite effects. Mechanistically, FAM83F regulated CC cell growth and glycolysis by the modulation of Wnt/β-catenin pathway. The enhancing effects of FAM83F overexpression on CC cell proliferation and glycolysis could be impaired by the Wnt/β-catenin inhibitor XAV939. Moreover, we found that c-Myc bound to the FAM83F promoter and activated the transcription of FAM83F. Notably, knockdown of FAM83F impaired the enhancement of cell proliferation and glycolysis induced by ectopic c-Myc. Consistent with in vitro findings, results from a xenograft mouse model confirmed the promoting role of FAM83F. In summary, our study demonstrated that FAM83F promoted CC growth and glycolysis through regulating the Wnt/β-catenin pathway, suggesting that FAM83F may be a potential molecular target for CC treatment. Schematic summary of c-Myc-activated FAM83F transcription to promote cervical cancer growth and glycolysis by targeting the Wnt/β-catenin signal pathway.
Collapse
Affiliation(s)
- Changlin Zhang
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, China
| | - Lixiang Liu
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Weizhao Li
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Mengxiong Li
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xunzhi Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Chi Zhang
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Huan Yang
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jiayuan Xie
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Wei Pan
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xue Guo
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Peng She
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Li Zhong
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Tian Li
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, China.
| |
Collapse
|
36
|
Wang T, Li C, Wang X, Liu F. MAGI2 ameliorates podocyte apoptosis of diabetic kidney disease through communication with TGF-β-Smad3/nephrin pathway. FASEB J 2023; 37:e23305. [PMID: 37950637 DOI: 10.1096/fj.202301058r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/27/2023] [Indexed: 11/13/2023]
Abstract
Podocytes, the key component of the glomerular filtration barrier (GFB), are gradually lost during the progression of diabetic kidney disease (DKD), severely compromising kidney functionality. The molecular mechanisms regulating the survival of podocytes in DKD are incompletely understood. Here, we show that membrane-associated guanylate kinase inverted 2 (MAGI2) is specifically expressed in renal podocytes, and promotes podocyte survival in DKD. We found that MAGI2 expression was downregulated in podocytes cultured with high-glucose in vitro, and in kidneys of db/db mice as well as DKD patients. Conversely, we found enforced expression of MAGI2 via AAV transduction protected podocytes from apoptosis, with concomitant improvement of renal functions. Mechanistically, we found that MAGI2 deficiency induced by high glucose levels activates TGF-β signaling to decrease the expression of anti-apoptotic proteins. These results indicate that MAGI2 protects podocytes from cell death, and can be harnessed therapeutically to improve renal function in diabetic kidney disease.
Collapse
Affiliation(s)
- Tingli Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of medicine, University of Electronic Science and Technology of China, Chengdu, China
- West China Hospital, Sichuan University, Chengdu, China
| | - Xiaofei Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Liu
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Chen Q, Xie C, Tang K, Luo M, Zhang Z, Jin Y, Liu Y, Zhou L, Kong Y. The E3 ligase Trim63 promotes podocyte injury and proteinuria by targeting PPARα to inhibit fatty acid oxidation. Free Radic Biol Med 2023; 209:40-54. [PMID: 37793501 DOI: 10.1016/j.freeradbiomed.2023.09.039] [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: 08/15/2023] [Revised: 09/21/2023] [Accepted: 09/30/2023] [Indexed: 10/06/2023]
Abstract
Podocyte injury is a hallmark of glomerular disease and one of the leading causes of chronic kidney disease (CKD). Peroxisome proliferator-activated receptor α (PPARα) plays a key role in podocyte fatty acid oxidation (FAO). However, the underlying regulatory mechanisms remain unresolved. Trim63 is an E3 ubiquitin ligase that has been shown to inhibit PPARα activity; however, its role in fatty acid metabolism in the kidney has not been elucidated to date. In this study, we investigated the effects of overexpression and knockdown of Trim63 in Adriamycin (ADR)-induced nephropathy and diabetic nephropathy models and a podocyte cell line. In both rodents and human patients with proteinuric CKD, Trim63 was upregulated, particularly in the podocytes of injured glomeruli. In the ADR-induced nephropathy model, ectopic Trim63 application aggravated FAO deficiency and mitochondrial dysfunction and triggered intense lipid deposition, podocyte injury, and proteinuria. Notably, Trim63 inhibition alleviated FAO deficiency and mitochondrial dysfunction, and markedly restored podocyte injury and renal fibrosis in ADR-induced and diabetic nephropathy (DN) models. Additionally, Trim63 was observed to mediate PPARα ubiquitination and degradation, leading to podocyte injury. We demonstrate the pathological role of Trim63, which was previously unrecognized in kidney tissue, in FAO deficiency and podocyte injury. Targeting Trim63 may represent a viable therapeutic strategy for podocyte injury and proteinuria.
Collapse
Affiliation(s)
- Qiyan Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Division of Nephrology, The First People's Hospital of Foshan, Foshan, China
| | - Chao Xie
- Division of Nephrology, The First People's Hospital of Foshan, Foshan, China
| | - Kaiyue Tang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mujin Luo
- Division of Nephrology, The First People's Hospital of Foshan, Foshan, China
| | - Zhe Zhang
- Division of Nephrology, The First People's Hospital of Foshan, Foshan, China
| | - Yabin Jin
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yaozhong Kong
- Division of Nephrology, The First People's Hospital of Foshan, Foshan, China.
| |
Collapse
|
38
|
Jiang W, Gan C, Zhou X, Yang Q, Chen D, Xiao H, Dai L, Chen Y, Wang M, Yang H, Li Q. Klotho inhibits renal ox-LDL deposition via IGF-1R/RAC1/OLR1 signaling to ameliorate podocyte injury in diabetic kidney disease. Cardiovasc Diabetol 2023; 22:293. [PMID: 37891556 PMCID: PMC10612302 DOI: 10.1186/s12933-023-02025-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
OBJECTIVE Diabetic kidney disease (DKD) is characterized by the abnormal deposition of oxidized low-density lipoprotein (ox-LDL), which contributes to podocyte damage. Klotho, an aging suppressor that plays a critical role in protecting podocytes in DKD, is mainly expressed in kidney tubular epithelium and secreted in the blood. However, it has not been established whether Klotho can alleviate podocyte injury by inhibiting renal ox-LDL deposition, and the potential molecular mechanisms require further investigation. METHODS We conducted a comprehensive analysis of serum and kidney biopsy samples obtained from patients diagnosed with DKD. Additionally, to explore the underlying mechanism of Klotho in the deposition of ox-LDL in the kidneys, we employed a mouse model of DKD with the Klotho genotype induced by streptozotocin (STZ). Furthermore, we conducted meticulous in vitro experiments on podocytes to gain further insights into the specific role of Klotho in the deposition of ox-LDL within the kidney. RESULTS Our groundbreaking study unveiled the remarkable ability of the soluble form of Klotho to effectively inhibit high glucose-induced ox-LDL deposition in podocytes affected by DKD. Subsequent investigations elucidated that Klotho achieved this inhibition by reducing the expression of the insulin/insulin-like growth factor 1 receptor (IGF-1R), consequently leading to a decrease in the expression of Ras-related C3 botulinum toxin substrate 1 (RAC1) and an enhancement of mitochondrial function. Ultimately, this series of events culminated in a significant reduction in the expression of the oxidized low-density lipoprotein receptor (OLR1), thereby resulting in a notable decrease in renal ox-LDL deposition in DKD. CONCLUSION Our findings suggested that Klotho had the potential to mitigate podocyte injury and reduced high glucose-induced ox-LDL deposition in glomerulus by modulating the IGF-1R/RAC1/OLR1 signaling. These results provided valuable insights that could inform the development of novel strategies for diagnosing and treating DKD.
Collapse
Affiliation(s)
- Wei Jiang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Chun Gan
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xindi Zhou
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qing Yang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dan Chen
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Han Xiao
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Lujun Dai
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Yaxi Chen
- Department of Infectious Diseases, Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Mo Wang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Haiping Yang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Qiu Li
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
39
|
Liang J, Liu Y. Animal Models of Kidney Disease: Challenges and Perspectives. KIDNEY360 2023; 4:1479-1493. [PMID: 37526653 PMCID: PMC10617803 DOI: 10.34067/kid.0000000000000227] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Kidney disease is highly prevalent and affects approximately 850 million people worldwide. It is also associated with high morbidity and mortality, and current therapies are incurable and often ineffective. Animal models are indispensable for understanding the pathophysiology of various kidney diseases and for preclinically testing novel remedies. In the last two decades, rodents continue to be the most used models for imitating human kidney diseases, largely because of the increasing availability of many unique genetically modified mice. Despite many limitations and pitfalls, animal models play an essential and irreplaceable role in gaining novel insights into the mechanisms, pathologies, and therapeutic targets of kidney disease. In this review, we highlight commonly used animal models of kidney diseases by focusing on experimental AKI, CKD, and diabetic kidney disease. We briefly summarize the pathological characteristics, advantages, and drawbacks of some widely used models. Emerging animal models such as mini pig, salamander, zebrafish, and drosophila, as well as human-derived kidney organoids and kidney-on-a-chip are also discussed. Undoubtedly, careful selection and utilization of appropriate animal models is of vital importance in deciphering the mechanisms underlying nephropathies and evaluating the efficacy of new treatment options. Such studies will provide a solid foundation for future diagnosis, prevention, and treatment of human kidney diseases.
Collapse
Affiliation(s)
- Jianqing Liang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
40
|
Ming WH, Luan ZL, Yao Y, Liu HC, Hu SY, Du CX, Zhang C, Zhao YH, Huang YZ, Sun XW, Qiao RF, Xu H, Guan YF, Zhang XY. Pregnane X receptor activation alleviates renal fibrosis in mice via interacting with p53 and inhibiting the Wnt7a/β-catenin signaling. Acta Pharmacol Sin 2023; 44:2075-2090. [PMID: 37344564 PMCID: PMC10545797 DOI: 10.1038/s41401-023-01113-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/18/2023] [Indexed: 06/23/2023]
Abstract
Renal fibrosis is a common pathological feature of chronic kidney disease (CKD) with various etiologies, which seriously affects the structure and function of the kidney. Pregnane X receptor (PXR) is a member of the nuclear receptor superfamily and plays a critical role in regulating the genes related to xenobiotic and endobiotic metabolism in mammals. Previous studies show that PXR is expressed in the kidney and has protective effect against acute kidney injury (AKI). In this study, we investigated the role of PXR in CKD. Adenine diet-induced CKD (AD) model was established in wild-type and PXR humanized (hPXR) mice, respectively, which were treated with pregnenolone-16α-carbonitrile (PCN, 50 mg/kg, twice a week for 4 weeks) or rifampicin (RIF, 10 mg·kg-1·d-1, for 4 weeks). We showed that both PCN and RIF, which activated mouse and human PXR, respectively, improved renal function and attenuated renal fibrosis in the two types of AD mice. In addition, PCN treatment also alleviated renal fibrosis in unilateral ureter obstruction (UUO) mice. On the contrary, PXR gene deficiency exacerbated renal dysfunction and fibrosis in both adenine- and UUO-induced CKD mice. We found that PCN treatment suppressed the expression of the profibrotic Wnt7a and β-catenin in AD mice and in cultured mouse renal tubular epithelial cells treated with TGFβ1 in vitro. We demonstrated that PXR was colocalized and interacted with p53 in the nuclei of tubular epithelial cells. Overexpression of p53 increased the expression of Wnt7a, β-catenin and its downstream gene fibronectin. We further revealed that p53 bound to the promoter of Wnt7a gene to increase its transcription and β-catenin activation, leading to increased expression of the downstream profibrotic genes, which was inhibited by PXR. Taken together, PXR activation alleviates renal fibrosis in mice via interacting with p53 and inhibiting the Wnt7a/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Wen-Hua Ming
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian, 116044, China
| | - Yao Yao
- Department of nephrology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226006, China
| | - Hang-Chi Liu
- Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Shu-Yuan Hu
- Department of nephrology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226006, China
| | - Chun-Xiu Du
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, 241100, China
| | - Cong Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yi-Hang Zhao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ying-Zhi Huang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Xiao-Wan Sun
- Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Rong-Fang Qiao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Hu Xu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian, 116044, China
| | - You-Fei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian, 116044, China.
| | - Xiao-Yan Zhang
- Health Science Center, East China Normal University, Shanghai, 200241, China.
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, 241100, China.
| |
Collapse
|
41
|
Tang Y, Wan F, Tang X, Lin Y, Zhang H, Cao J, Yang R. Celastrol attenuates diabetic nephropathy by upregulating SIRT1-mediated inhibition of EZH2related wnt/β-catenin signaling. Int Immunopharmacol 2023; 122:110584. [PMID: 37454630 DOI: 10.1016/j.intimp.2023.110584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/22/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
Proteinuria is an independent risk factor for the progression of diabetic nephropathy (DN) and an imbalance in podocyte function aggravates proteinuria. Celastrol is the primary active ingredient of T. wilfordii, effective in treating DN renal injury; however, the mechanisms underlying its effect are unclear. We explored how celastrol prevents DN podocyte damage using in vivo and in vitro experiments. We randomly divided 24 male C57BLKS/J mice into three groups: db/m (n = 8), db/db (n = 8), and celastrol groups (db/db + celastrol, 1 mg/kg/d, gavage administration, n = 8). In vivo experiments lasted 12 weeks and intervention lasted ten weeks. Serum samples and kidney tissues were collected for biochemical tests, pathological staining, transmission electron microscopy, fluorescencequantitation polymerase chain reaction, and western blotting analysis. In vitro experiments to elaborate the mechanism of celastrol protection were performed on high glucose (HG)-induced podocyte injury. Celastrol reduced blood glucose levels and renal function index in db/db mice, attenuated renal histomorphological injury and glomerular podocyte foot injuries, and induced significant anti-inflammatory effects. Celastrol upregulated silent information regulator 2 related enzyme 1(SIRT1) expression and downregulated enhancer of zeste homolog (EZH2), inhibiting the wnt/β-catenin pathway-related molecules, such as wnt1, wnt7a, and β-catenin. SIRT1 repressed the promoter activity of EZH2, and was co-immunoprecipitated with EZH2 in mouse podocyte cells (MPC5). SIRT1 knockdown aggravated the protective effects of celastrol on MPC5 cells. Celastrol protected podocyte injury via SIRT1/EZH2, which participates in the wnt/β-catenin pathway. Overall, celastrol-mediated SIRT1 upregulation inhibited the EZH2-related wnt/β-catenin signaling pathway to attenuate DN and podocyte injury, providing a theoretical basis for celastrol clinical application.
Collapse
Affiliation(s)
- Yuewen Tang
- Laboratory of Nephropathy, Hangzhou Hospital of Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Feng Wan
- Laboratory of Nephropathy, Hangzhou Hospital of Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xuanli Tang
- Laboratory of Nephropathy, Hangzhou Hospital of Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yi Lin
- Laboratory of Nephropathy, Hangzhou Hospital of Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Huaqin Zhang
- Laboratory of Nephropathy, Hangzhou Hospital of Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jiawei Cao
- Laboratory of Nephropathy, Hangzhou Hospital of Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ruchun Yang
- Laboratory of Nephropathy, Hangzhou Hospital of Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
42
|
Yan H, Huang X, Xu J, Zhang Y, Chen J, Xu Z, Li H, Wang Z, Yang X, Yang B, He Q, Luo P. Chloroquine Intervenes Nephrotoxicity of Nilotinib through Deubiquitinase USP13-Mediated Stabilization of Bcl-XL. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302002. [PMID: 37452432 PMCID: PMC10502815 DOI: 10.1002/advs.202302002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Nephrotoxicity has become prominent due to the increase in the clinical use of nilotinib, a second-generation BCR-ABL1 inhibitor in the first-line treatment of Philadelphia chromosome-positive chronic myeloid leukemia. To date, the mechanism of nilotinib nephrotoxicity is still unknown, leading to a lack of clinical intervention strategies. Here, it is found that nilotinib could induce glomerular atrophy, renal tubular degeneration, and kidney fibrosis in an animal model. Mechanistically, nilotinib induces intrinsic apoptosis by specifically reducing the level of BCL2 like 1 (Bcl-XL) in both vascular endothelial cells and renal tubular epithelial cells, as well as in vivo. It is confirmed that chloroquine (CQ) intervenes with nilotinib-induced apoptosis and improves mitochondrial integrity, reactive oxygen species accumulation, and DNA damage by reversing the decreased Bcl-XL. The intervention effect is dependent on the alleviation of the nilotinib-induced reduction in ubiquitin specific peptidase 13 (USP13) and does not rely on autophagy inhibition. Additionally, it is found that USP13 abrogates cell apoptosis by preventing excessive ubiquitin-proteasome degradation of Bcl-XL. In conclusion, the research reveals the molecular mechanism of nilotinib's nephrotoxicity, highlighting USP13 as an important regulator of Bcl-XL stability in determining cell fate, and provides CQ analogs as a clinical intervention strategy for nilotinib's nephrotoxicity.
Collapse
Affiliation(s)
- Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Xiangliang Huang
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Jiangxin Xu
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Ying Zhang
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Jiajia Chen
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Hui Li
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Zeng Wang
- Department of PharmacyZhejiang Cancer HospitalHangzhou310005China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Bo Yang
- Institute of Pharmacology & ToxicologyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityHangzhou310018China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang UniversityCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Department of CardiologySecond Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009China
| |
Collapse
|
43
|
Cailotto F, Santulli G. Editorial: Wnt signaling in endocrine and metabolic disorders. Front Endocrinol (Lausanne) 2023; 14:1254977. [PMID: 37608791 PMCID: PMC10441232 DOI: 10.3389/fendo.2023.1254977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023] Open
Affiliation(s)
- Frederic Cailotto
- UMR7365 Ingénierie Moléculaire et Physiopathologie Articulaire (IMOPA) Centre National de la Recherche Scientifique - Université de Lorraine (CNRS-UL), Biopôle de l’Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY, United States
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Fibroblast growth factor (FGF) 23 is a bone-derived hormone that regulates phosphate and vitamin D metabolism by targeting the kidney. When highly elevated, such as in chronic kidney disease (CKD), FGF23 can also target the heart and induce pathologic remodeling. Here we discuss the mechanisms that underlie the physiologic and pathologic actions of FGF23, with focus on its FGF receptors (FGFR) and co-receptors. RECENT FINDINGS Klotho is a transmembrane protein that acts as an FGFR co-receptor for FGF23 on physiologic target cells. Klotho also exists as a circulating variant, and recent studies suggested that soluble klotho (sKL) can mediate FGF23 effects in cells that do not express klotho. Furthermore, it has been assumed that the actions of FGF23 do not require heparan sulfate (HS), a proteoglycan that acts as a co-receptor for other FGF isoforms. However, recent studies revealed that HS can be part of the FGF23:FGFR signaling complex and modulate FGF23-induced effects. SUMMARY sKL and HS have appeared as circulating FGFR co-receptors that modulate the actions of FGF23. Experimental studies suggest that sKL protects from and HS accelerates CKD-associated heart injury. However, the in vivo relevance of these findings is still speculative.
Collapse
Affiliation(s)
- S Madison Thomas
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | |
Collapse
|
45
|
Zhang N, Shen H, Chen B, Hu H, Liu C, Chen Y, Cong W. The recent progress of peptide regulators for the Wnt/β-catenin signaling pathway. Front Med (Lausanne) 2023; 10:1164656. [PMID: 37396899 PMCID: PMC10311566 DOI: 10.3389/fmed.2023.1164656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/16/2023] [Indexed: 07/04/2023] Open
Abstract
Wnt signaling plays an important role in many biological processes such as stem cell self-renewal, cell proliferation, migration, and differentiation. The β-catenin-dependent signaling pathway mainly regulates cell proliferation, differentiation, and migration. In the Wnt/β-catenin signaling pathway, the Wnt family ligands transduce signals through LRP5/6 and Frizzled receptors to the Wnt/β-catenin signaling cascades. Wnt-targeted therapy has garnered extensive attention. The most commonly used approach in targeted therapy is small-molecule regulators. However, it is difficult for small-molecule regulators to make great progress due to their inherent defects. Therapeutic peptide regulators targeting the Wnt signaling pathway have become an alternative therapy, promising to fill the gaps in the clinical application of small-molecule regulators. In this review, we describe recent advances in peptide regulators for Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Nan Zhang
- School of Medicine or Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Huaxing Shen
- School of Medicine or Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Baobao Chen
- School of Medicine or Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Honggang Hu
- School of Medicine or Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Chao Liu
- School of Medicine or Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Yan Chen
- Department of Pharmacy, Medical Supplies Center of People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Wei Cong
- School of Medicine or Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
46
|
Li SS, Sheng MJ, Sun ZY, Liang Y, Yu LX, Liu QF. Upstream and downstream regulators of Klotho expression in chronic kidney disease. Metabolism 2023; 142:155530. [PMID: 36868370 DOI: 10.1016/j.metabol.2023.155530] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023]
Abstract
Klotho is a critical protein that protects the kidney. Klotho is severely downregulated in chronic kidney disease (CKD), and its deficiency is implicated in the pathogenesis and progression of CKD. Conversely, an increase in Klotho levels results in improved kidney function and delays CKD progression, supporting the notion that modulating Klotho levels could represent a possible therapeutic strategy for CKD treatment. Nevertheless, the regulatory mechanisms responsible for the loss of Klotho remain elusive. Previous studies have demonstrated that oxidative stress, inflammation, and epigenetic modifications can modulate Klotho levels. These mechanisms result in a decrease in Klotho mRNA transcript levels and reduced translation, thus can be grouped together as upstream regulatory mechanisms. However, therapeutic strategies that aim to rescue Klotho levels by targeting these upstream mechanisms do not always result in increased Klotho, indicating the involvement of other regulatory mechanisms. Emerging evidence has shown that endoplasmic reticulum (ER) stress, the unfolded protein response, and ER-associated degradation also affect the modification, translocation, and degradation of Klotho, and thus are proposed to be downstream regulatory mechanisms. Here, we discuss the current understanding of upstream and downstream regulatory mechanisms of Klotho and examine potential therapeutic strategies to upregulate Klotho expression for CKD treatment.
Collapse
Affiliation(s)
- Sha-Sha Li
- Clinical Research & Lab Centre, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China
| | - Ming-Jie Sheng
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China
| | - Zhuo-Yi Sun
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China
| | - Yan Liang
- Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China
| | - Li-Xia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China.
| | - Qi-Feng Liu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China; Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China.
| |
Collapse
|
47
|
Tang A, Zhang Y, Wu L, Lin Y, Lv L, Zhao L, Xu B, Huang Y, Li M. Klotho's impact on diabetic nephropathy and its emerging connection to diabetic retinopathy. Front Endocrinol (Lausanne) 2023; 14:1180169. [PMID: 37143722 PMCID: PMC10151763 DOI: 10.3389/fendo.2023.1180169] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/04/2023] [Indexed: 05/06/2023] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease worldwide and is a significant burden on healthcare systems. α-klotho (klotho) is a protein known for its anti-aging properties and has been shown to delay the onset of age-related diseases. Soluble klotho is produced by cleavage of the full-length transmembrane protein by a disintegrin and metalloproteases, and it exerts various physiological effects by circulating throughout the body. In type 2 diabetes and its complications DN, a significant decrease in klotho expression has been observed. This reduction in klotho levels may indicate the progression of DN and suggest that klotho may be involved in multiple pathological mechanisms that contribute to the onset and development of DN. This article examines the potential of soluble klotho as a therapeutic agent for DN, with a focus on its ability to impact multiple pathways. These pathways include anti-inflammatory and oxidative stress, anti-fibrotic, endothelial protection, prevention of vascular calcification, regulation of metabolism, maintenance of calcium and phosphate homeostasis, and regulation of cell fate through modulation of autophagy, apoptosis, and pyroptosis pathways. Diabetic retinopathy shares similar pathological mechanisms with DN, and targeting klotho may offer new insights into the prevention and treatment of both conditions. Finally, this review assesses the potential of various drugs used in clinical practice to modulate klotho levels through different mechanisms and their potential to improve DN by impacting klotho levels.
Collapse
Affiliation(s)
- Anqi Tang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Yu Zhang
- Department of Nephrology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an, China
| | - Ling Wu
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Yong Lin
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Lizeyu Lv
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Liangbin Zhao
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Bojun Xu
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Youqun Huang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Mingquan Li
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
- *Correspondence: Mingquan Li,
| |
Collapse
|
48
|
Speer T, Schunk SJ. Klotho in diabetic kidney disease: more than dust in the Wnt. Kidney Int 2022; 102:469-471. [PMID: 35988933 DOI: 10.1016/j.kint.2022.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 10/15/2022]
Abstract
Activation of the Wnt/β-catenin pathway represents a hallmark in the development of kidney fibrosis. Herein, Chen et al. report that Klotho-derived peptide 6, a peptide mimicking the function of the protein Klotho, directly binds to endogenous Wnt ligands and, thereby, serves as a small-molecule inhibitor of canonical Wnt/β-catenin signaling. In diabetic kidney disease, Klotho-derived peptide 6 reduces glomerular injury and preserves kidney function, highlighting Klotho-derived peptide 6 as a novel therapeutic agent.
Collapse
Affiliation(s)
- Thimoteus Speer
- Translational Cardio-Renal Medicine, Saarland University, Homburg/Saar, Germany.
| | - Stefan J Schunk
- Department of Nephrology and Hypertension, Saarland University, Homburg/Saar, Germany
| |
Collapse
|