1
|
Li R, Tao H, Pan K, Li R, Guo Z, Chen X, Li Z. Extracellular vesicles derived from mesenchymal stem cells alleviate renal fibrosis via the miR-99b-5p/mTOR/autophagy axis in diabetic kidney disease. Stem Cell Res Ther 2025; 16:142. [PMID: 40103007 PMCID: PMC11921689 DOI: 10.1186/s13287-025-04265-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/04/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease (ESRD) globally, presenting a significant therapeutic challenge. Extracellular vesicles (EVs) from mesenchymal stem cells (MSCs) have emerged as promising therapeutic agents. This study explored the therapeutic effects and mechanisms of EVs derived from human placental mesenchymal stem cells (hP-MSCs) on DKD. METHODS EVs were isolated from cultured hP-MSCs and administered to streptozotocin (STZ)-induced diabetic mice and high glucose-treated glomerular mesangial cells. The therapeutic impact of EVs was assessed through histological analysis and biochemical assays. miR-99b-5p expression in EVs and its role in modulating the mechanistic target of rapamycin (mTOR)/autophagy pathway were examined via western blotting and RT‒qPCR. RESULTS Treatment with hP-MSC-derived EVs significantly alleviated renal fibrosis and improved renal function in DKD models. These EVs were enriched with miR-99b-5p, which targeted and inhibited mTOR signaling, thereby increasing autophagic activity and reducing cellular proliferation and extracellular matrix accumulation in renal tissues. CONCLUSIONS hP-MSC-derived EVs can mitigate renal injury in DKD by modulating the miR-99b-5p/mTOR/autophagy pathway. These findings suggest a potential cell-free therapeutic strategy for managing DKD.
Collapse
Affiliation(s)
- Rongrong Li
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, No. 17 Jingnan 5th Road, 450016, Zhengzhou, China
- School of Medicine, Nankai University, 94 Weijin Road, 300071, Tianjin, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China, 601 Jinsui Road, 453003
| | - Hongyan Tao
- School of Medicine, Nankai University, 94 Weijin Road, 300071, Tianjin, China
| | - Kai Pan
- School of Medicine, Nankai University, 94 Weijin Road, 300071, Tianjin, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China, 601 Jinsui Road, 453003
| | - Rui Li
- School of Medicine, Nankai University, 94 Weijin Road, 300071, Tianjin, China
| | - Zhikun Guo
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, No. 17 Jingnan 5th Road, 450016, Zhengzhou, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China, 601 Jinsui Road, 453003
| | - Xiaoniao Chen
- Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, 69 Yongding Road, Beijing, 100039, China.
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, 100853, Beijing, China.
| | - Zongjin Li
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, No. 17 Jingnan 5th Road, 450016, Zhengzhou, China.
- School of Medicine, Nankai University, 94 Weijin Road, 300071, Tianjin, China.
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China, 601 Jinsui Road, 453003.
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, 100853, Beijing, China.
| |
Collapse
|
2
|
Lee HJ, Sun Y, Das F, Ju W, Nair V, Kevil CG, Varadarajan S, Zhang G, Ghosh Choudhury G, Singh BB, Kretzler M, Nelson RG, Sharma K, Kasinath BS. The CLCA1/TMEM16A/Cl- current axis associates with H2S deficiency in diabetic kidney injury. JCI Insight 2025; 10:e174848. [PMID: 39782685 PMCID: PMC11721299 DOI: 10.1172/jci.insight.174848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/13/2024] [Indexed: 01/30/2025] Open
Abstract
The role played by anionic channels in diabetic kidney disease (DKD) is not known. Chloride channel accessory 1 (CLCA1) facilitates the activity of TMEM16A (Anoctamin-1), a Ca2+-dependent Cl- channel. We examined if CLCA1/TMEM16A had a role in DKD. In mice with type 2 diabetes, renal cortical CLCA1 and TMEM16A content was increased. CLCA1 and TMEM16A content was associated with hydrogen sulfide (H2S) deficiency, mTOR complex 1 (mTORC1) activation, albuminuria, and matrix increase. Administering sodium hydrosulfide (NaHS), a source of H2S, mitigated these changes. In proximal tubular epithelial (MCT) cells, high glucose rapidly increased CLCA1 by recruiting the IL-6/STAT3 axis and augmented TMEM16A expression by stimulating its mRNA translation; these changes were abolished by NaHS. Patch clamp experiments showed that high glucose increased Cl- current in MCT cells that was ameliorated by NaHS and a TMEM16A chemical inhibitor. siRNA against CLCA1 or TMEM16A and TMEM16A inhibitor abolished high glucose-induced mTORC1 activation and matrix protein increase. Tubular expression of TMEM16A correlated with albuminuria in kidney biopsies from people with type 2 diabetes. We report a pathway for DKD in which H2S deficiency results in kidney injury by the recruitment of the CLCA1/TMEM16A/Cl- current system.
Collapse
Affiliation(s)
- Hak Joo Lee
- Center for Precision Medicine, Department of Medicine, and
| | - Yuyang Sun
- Department of Periodontics, University of Texas Health, San Antonio, San Antonio, Texas, USA
| | - Falguni Das
- Center for Precision Medicine, Department of Medicine, and
| | - Wenjun Ju
- Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Christopher G. Kevil
- Department of Pathology, Louisiana State University Health Science Center, Shreveport, Louisiana, USA
| | - Shankara Varadarajan
- Department of Periodontics, University of Texas Health, San Antonio, San Antonio, Texas, USA
| | - Guanshi Zhang
- Center for Precision Medicine, Department of Medicine, and
| | - Goutam Ghosh Choudhury
- Center for Precision Medicine, Department of Medicine, and
- Research service and
- Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Brij B. Singh
- Department of Periodontics, University of Texas Health, San Antonio, San Antonio, Texas, USA
| | - Matthias Kretzler
- Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Robert G. Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
- Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Kumar Sharma
- Center for Precision Medicine, Department of Medicine, and
- Research service and
| | - Balakuntalam S. Kasinath
- Center for Precision Medicine, Department of Medicine, and
- Research service and
- Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| |
Collapse
|
3
|
Deng X, Tang C, Fang T, Li T, Li X, Liu Y, Zhang X, Sun B, Sun H, Chen L. Disruption of branched-chain amino acid homeostasis promotes the progression of DKD via enhancing inflammation and fibrosis-associated epithelial-mesenchymal transition. Metabolism 2025; 162:156037. [PMID: 39317264 DOI: 10.1016/j.metabol.2024.156037] [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: 07/01/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND AND AIMS The disrupted homeostasis of branched-chain amino acids (BCAAs, including leucine, isoleucine, and valine) has been strongly correlated with diabetes with a potential causal role. However, the relationship between BCAAs and diabetic kidney disease (DKD) remains to be established. Here, we show that the elevated BCAAs from BCAAs homeostatic disruption promote DKD progression unexpectedly as an independent risk factor. METHODS AND RESULTS Similar to other tissues, the suppressed BCAAs catabolic gene expression and elevated BCAAs abundance were detected in the kidneys of type 2 diabetic mice and individuals with DKD. Genetic and nutritional studies demonstrated that the elevated BCAAs from systemic disruption of BCAAs homeostasis promoted the progression of DKD. Of note, the elevated BCAAs promoted DKD progression without exacerbating diabetes in the animal models of type 2 DKD. Mechanistic studies demonstrated that the elevated BCAAs promoted fibrosis-associated epithelial-mesenchymal transition (EMT) by enhancing the activation of proinflammatory macrophages through mTOR signaling. Furthermore, pharmacological enhancement of systemic BCAAs catabolism using small molecule inhibitor attenuated type 2 DKD. Finally, the elevated BCAAs also promoted DKD progression in type 1 diabetic mice without exacerbating diabetes. CONCLUSION BCAA homeostatic disruption serves as an independent risk factor for DKD and restoring BCAA homeostasis pharmacologically or dietarily represents a promising therapeutic strategy to ameliorate the progression of DKD.
Collapse
Affiliation(s)
- Xiaoqing Deng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Chao Tang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Affiliated Huzhou Hospital, Zhejiang University School of Medicine, China
| | - Ting Fang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Ting Li
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xiaoyu Li
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Yajin Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xuejiao Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Haipeng Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Center for Cardiovascular Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
4
|
Wang Z, Chen G, Li H, Liu J, Yang Y, Zhao C, Li Y, Shi J, Chen H, Chen G. Zotarolimus alleviates post-trabeculectomy fibrosis via dual functions of anti-inflammation and regulating AMPK/mTOR axis. Int Immunopharmacol 2024; 142:113176. [PMID: 39303539 DOI: 10.1016/j.intimp.2024.113176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE Postoperative scar formation is the primary cause of uncontrolled intraocular pressure following trabeculectomy failure. This study aimed to evaluate the efficacy of zotarolimus as an adjuvant anti-scarring agent in the experimental trabeculectomy. METHODS We performed differential gene and Gene Ontology enrichment analysis on rabbit follicular transcriptome sequencing data (GSE156781). New Zealand white Rabbits were randomly assigned into three groups: Surgery only, Surgery with mitomycin-C treatment, Surgery with zotarolimus treatment. Rabbits were euthanized 3 days or 28 days post-trabeculectomy. Pathological sections were analyzed using immunohistochemistry, immunofluorescence, and Masson staining. In vitro, primary human tenon's capsule fibroblasts (HTFs) were stimulated by transforming growth factor-β1 (TGF-β1) and treated with either mitomycin-C or zotarolimus. Cell proliferation and migration were evaluated using cell counting kit-8, cell cycle, and scratch assays. Mitochondrial membrane potential was detected with the JC-1 probe, and reactive oxygen species were detected using the DCFH-DA probe. RNA and protein expressions were quantified using RT-qPCR and immunofluorescence. RESULTS Transcriptome sequencing analysis revealed the involvement of complex immune factors and metabolic disorders in trabeculectomy outcomes. Zotarolimus effectively inhibited fibrosis, reduced proinflammatory factor release and immune cell infiltration, and improved the surgical outcomes of trabeculectomy. In TGF-β1-induced HTFs, zotarolimus reduced fibrosis, proliferation, and migration without cytotoxicity via the dual regulation of the TGF-β1/Smad2/3 and AMPK/AKT/mTOR pathways. CONCLUSION Our study demonstrates that zotarolimus mitigates fibrosis by reducing immune infiltration and correcting metabolic imbalances, offering a potential treatment for improving trabeculectomy surgical outcomes.
Collapse
Affiliation(s)
- Zhiruo Wang
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Gong Chen
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Haoyu Li
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Jingyuan Liu
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Yuanyuan Yang
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Cong Zhao
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Yunping Li
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Jingming Shi
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Huihui Chen
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China; Clinical Immunology Research Center of Central South University, Changsha, China.
| | - Guochun Chen
- Clinical Immunology Research Center of Central South University, Changsha, China; Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
5
|
Alanazi YA, Al‐kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Bahaa MM, Negm WA, AlAnazi FH, Alrouji M, Batiha GE. Role of Autophagy in Type 2 Diabetes Mellitus: The Metabolic Clash. J Cell Mol Med 2024; 28:e70240. [PMID: 39656379 PMCID: PMC11629865 DOI: 10.1111/jcmm.70240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is developed due to the development of insulin resistance (IR) and pancreatic β cell dysfunction with subsequent hyperglycaemia. Hyperglycaemia-induced oxidative stress and endoplasmic reticulum (ER) stress enhances inflammatory disorders, leading to further pancreatic β cell dysfunction. These changes trigger autophagy activation, which recycles cytoplasmic components and injured organelles. Autophagy regulates pancreatic β cell functions by different mechanisms. Though the exact role of autophagy in T2DM is not completely elucidated, that could be beneficial or detrimental. Therefore, this review aims to discuss the exact role of autophagy in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Yousef Abud Alanazi
- Department of Pediatrics, College of MedicineMajmaah UniversityMajmaahSaudi Arabia
| | - Haydar M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliPunjabIndia
- Department of Research & DevelopmentFunogenAthensGreece
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Mostafa M. Bahaa
- Pharmacy Practice Department, Faculty of PharmacyHorus UniversityNew DamiettaEgypt
| | - Walaa A. Negm
- Department of Pharmacognosy, Faculty of PharmacyTanta UniversityTantaEgypt
| | - Faisal Holil AlAnazi
- Department of Internal Medicine, College of MedicineMajmaah UniversityMajmaahSaudi Arabia
| | - Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesShaqra UniversityShaqraSaudi Arabia
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
6
|
Raya AI, Vidal A, López I, Rodríguez M, Aguilera-Tejero E, Pineda C. Phosphorus Restriction Prevents Rapamycin-Induced Kidney Damage in Rats. Am J Nephrol 2024; 56:48-57. [PMID: 39383849 DOI: 10.1159/000541411] [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: 08/01/2024] [Accepted: 09/08/2024] [Indexed: 10/11/2024]
Abstract
INTRODUCTION There are conflicting reports about the effect or rapamycin on the kidneys. Rapamycin is known to promote phosphaturia that may be associated to renal injury. METHODS Detailed histopathological studies were performed on the kidneys of rats with normal (control) and reduced (Nx) renal mass that were treated with rapamycin (1.3 mg/kg for 22 days) or placebo. The effect of rapamycin was also evaluated in control and Nx rats fed different amounts of phosphorus: 0.6% P (NP), 1.2% P (HP), and 0.2% P (LP). Quantitative scores of kidney lesions were obtained for interstitial nephritis (IN), tubular damage (TD), and nephrocalcinosis (NC). RESULTS When compared with placebo, rapamycin administration to Nx rats resulted in significant increases in IN (4.17 ± 0.74 vs. 1.51 ± 0.53%) and TD (14.45 ± 1.51 vs. 8.61 ± 1.83%). Rapamycin also increased NC both in control (0.86 ± 0.23 vs. 0.14 ± 0.06%) and Nx (0.86 ± 0.32 vs. 0.15 ± 0.14%) rats. In control rats receiving rapamycin, feeding HP aggravated IN (3.25 ± 0.48%), TD (22.47 ± 4.56%), and NC (3.66 ± 0.75%), while feeding LP prevented development of any renal lesions. In Nx rats treated with rapamycin, HP intake also increased IN (8.95 ± 1.94%), TD (26.86 ± 3.95%), and NC (2.77 ± 0.60%), whereas feeding LP reduced all lesions to lower levels than in rats fed NP. Rapamycin treatment increased fractional excretion of P (FEP), and an excellent correlation between scores for renal lesions and FEP was found. CONCLUSION Rapamycin has deleterious effects on kidney pathology causing lesions that are located mainly at tubular and tubulointerstitial level. Rapamycin-induced kidney damage is more evident in rats that already have decreased renal function and seems to be related to the phosphaturic effect of the drug. Dietary P restriction prevents kidney damage in rats treated with rapamycin.
Collapse
Affiliation(s)
- Ana I Raya
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Angela Vidal
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Ignacio López
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Mariano Rodríguez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Escolástico Aguilera-Tejero
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Carmen Pineda
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| |
Collapse
|
7
|
Swaroop V, Ozkan E, Herrmann L, Thurman A, Kopasz-Gemmen O, Kunamneni A, Inoki K. mTORC1 signaling and diabetic kidney disease. Diabetol Int 2024; 15:707-718. [PMID: 39469564 PMCID: PMC11512951 DOI: 10.1007/s13340-024-00738-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/26/2024] [Indexed: 10/30/2024]
Abstract
Diabetic kidney disease (DKD) represents the most lethal complication in both type 1 and type 2 diabetes. The disease progresses without obvious symptoms and is often refractory when apparent symptoms have emerged. Although the molecular mechanisms underlying the onset/progression of DKD have been extensively studied, only a few effective therapies are currently available. Pathogenesis of DKD involves multifaced events caused by diabetes, which include alterations of metabolisms, signals, and hemodynamics. While the considerable efficacy of sodium/glucose cotransporter-2 (SGLT2) inhibitors or angiotensin II receptor blockers (ARBs) for DKD has been recognized, the ever-increasing number of patients with diabetes and DKD warrants additional practical therapeutic approaches that prevent DKD from diabetes. One plausible but promising target is the mechanistic target of the rapamycin complex 1 (mTORC1) signaling pathway, which senses cellular nutrients to control various anabolic and catabolic processes. This review introduces the current understanding of the mTOR signaling pathway and its roles in the development of DKD and other chronic kidney diseases (CKDs), and discusses potential therapeutic approaches targeting this pathway for the future treatment of DKD.
Collapse
Affiliation(s)
- Vinamra Swaroop
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Eden Ozkan
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Lydia Herrmann
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Aaron Thurman
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | | | | | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, USA
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, USA
| |
Collapse
|
8
|
Lee HJ, Min L, Gao J, Matta S, Drel V, Saliba A, Tamayo I, Montellano R, Hejazi L, Maity S, Xu G, Grajeda BI, Roy S, Hallows KR, Choudhury GG, Kasinath BS, Sharma K. Female Protection Against Diabetic Kidney Disease Is Regulated by Kidney-Specific AMPK Activity. Diabetes 2024; 73:1167-1177. [PMID: 38656940 PMCID: PMC11189830 DOI: 10.2337/db23-0807] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
Reduced kidney AMPK activity is associated with nutrient stress-induced chronic kidney disease (CKD) in male mice. In contrast, female mice resist nutrient stress-induced CKD. The role of kidney AMPK in sex-related organ protection against nutrient stress and metabolite changes was evaluated in diabetic kidney tubule-specific AMPKγ2KO (KTAMPKγ2ΚΟ) male and female mice. In wild-type (WT) males, diabetes increased albuminuria, urinary kidney injury molecule-1, hypertension, kidney p70S6K phosphorylation, and kidney matrix accumulation; these features were not exacerbated with KTAMPKγ2ΚΟ. Whereas WT females had protection against diabetes-induced kidney injury, KTAMPKγ2ΚΟ led to loss of female protection against kidney disease. The hormone 17β-estradiol ameliorated high glucose-induced AMPK inactivation, p70S6K phosphorylation, and matrix protein accumulation in kidney tubule cells. The mechanism for female protection against diabetes-induced kidney injury is likely via an estrogen-AMPK pathway, as inhibition of AMPK led to loss of estrogen protection to glucose-induced mTORC1 activation and matrix production. RNA sequencing and metabolomic analysis identified a decrease in the degradation pathway of phenylalanine and tyrosine resulting in increased urinary phenylalanine and tyrosine levels in females. The metabolite levels correlated with loss of female protection. The findings provide new insights to explain evolutionary advantages to females during states of nutrient challenges. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Hak Joo Lee
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
- South Texas Veterans Health Care System, San Antonio, TX
| | - Liang Min
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Jingli Gao
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Shane Matta
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Viktor Drel
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Afaf Saliba
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Ian Tamayo
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Richard Montellano
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Leila Hejazi
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Soumya Maity
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Guogang Xu
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Brian I. Grajeda
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas, El Paso, TX
| | - Sourav Roy
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas, El Paso, TX
| | - Kenneth R. Hallows
- USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Goutam Ghosh Choudhury
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
- South Texas Veterans Health Care System, San Antonio, TX
| | - Balakuntalam S. Kasinath
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
- South Texas Veterans Health Care System, San Antonio, TX
| | - Kumar Sharma
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
- South Texas Veterans Health Care System, San Antonio, TX
| |
Collapse
|
9
|
Das F, Ghosh-Choudhury N, Kasinath BS, Sharma K, Choudhury GG. High glucose-induced downregulation of PTEN-Long is sufficient for proximal tubular cell injury in diabetic kidney disease. Exp Cell Res 2024; 440:114116. [PMID: 38830568 DOI: 10.1016/j.yexcr.2024.114116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 04/24/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
During the progression of diabetic kidney disease, proximal tubular epithelial cells respond to high glucose to induce hypertrophy and matrix expansion leading to renal fibrosis. Recently, a non-canonical PTEN has been shown to be translated from an upstream initiation codon CUG (leucine) to produce a longer protein called PTEN-Long (PTEN-L). Interestingly, the extended sequence present in PTEN-L contains cell secretion/penetration signal. Role of this non-canonical PTEN-L in diabetic renal tubular injury is not known. We show that high glucose decreases expression of PTEN-L. As a mechanism of its function, we find that reduced PTEN-L activates Akt-2, which phosphorylates and inactivate tuberin and PRAS40, resulting in activation of mTORC1 in tubular cells. Antibacterial agent acriflavine and antiviral agent ATA regulate translation from CUG codon. Acriflavine and ATA, respectively, decreased and increased expression of PTEN-L to altering Akt-2 and mTORC1 activation in the absence of change in expression of canonical PTEN. Consequently, acriflavine and ATA modulated high glucose-induced tubular cell hypertrophy and lamininγ1 expression. Importantly, expression of PTEN-L inhibited high glucose-stimulated Akt/mTORC1 activity to abrogate these processes. Since PTEN-L contains secretion/penetration signals, addition of conditioned medium containing PTEN-L blocked Akt-2/mTORC1 activity. Notably, in renal cortex of diabetic mice, we found reduced PTEN-L concomitant with Akt-2/mTORC1 activation, leading to renal hypertrophy and lamininγ1 expression. These results present first evidence for involvement of PTEN-L in diabetic kidney disease.
Collapse
Affiliation(s)
- Falguni Das
- VA Research, South Texas Veterans Health Care System, San Antonio, TX, USA; Department of Medicine, TX, USA
| | | | | | - Kumar Sharma
- VA Research, South Texas Veterans Health Care System, San Antonio, TX, USA; Department of Medicine, TX, USA
| | - Goutam Ghosh Choudhury
- VA Research, South Texas Veterans Health Care System, San Antonio, TX, USA; Department of Medicine, TX, USA; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA.
| |
Collapse
|
10
|
Farooqui Z, Banday AA. Angiotensin 1-7 exerts antioxidant effects, suppresses Mammalian Target of Rapamycin (mTOR) signaling, and inhibits apoptosis in renal proximal tubular cells. Peptides 2024; 172:171136. [PMID: 38104660 DOI: 10.1016/j.peptides.2023.171136] [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: 10/31/2023] [Revised: 12/09/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
Oxidative stress is one of the crucial pathogenic factors involved in the progression of renal injury. Angiotensin (ANG) 1-7, a bioactive heptapeptide of the renin-angiotensin-aldosterone system is known to exert antioxidant and nephroprotective effects. However, the cellular mechanism involved in the beneficial effect of ANG 1-7 is not clear. Here, we assessed ANG 1-7's effect on H2O2-mediated oxidative damage in the human proximal tubular (HK2) cells and the underlying mechanisms. HK2 cells were incubated with H2O2 (500 µM, 4 h) pre-treated with and without ANG 1-7 (100 nM, 24 h), and reactive oxygen species (ROS) generation, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, apoptosis and mammalian target of rapamycin (mTOR) signaling were determined H2O2 induced an increase in oxidative and ER stress together with loss of mitochondrial membrane potential, decreased ATP levels, and induced apoptosis in HK2 cells. Moreover, H2O2 treatment resulted in the activation of mTOR complexes (mTORC1 and mTORC2) in these cells. ANG 1-7 significantly attenuated H2O2-induced ROS generation, ER stress and apoptosis, and also improved mitochondrial function. Additionally, pre-treatment of ANG 1-7 inhibited the H2O2-mediated mTOR activation. These effects of ANG 1-7 were blocked by co-treatment with the Mas receptor (MasR) inhibitor, A779. Furthermore, transfection of HK2 cells with Mas receptor siRNA also abolished the inhibitory effect of ANG 1-7 on mTOR activities. In conclusion, ANG 1-7 via MasR mitigates oxidative stress, suppresses mTOR signaling, and protects HK2 cells from ER stress, mitochondrial dysfunction, and apoptosis, suggesting ANG 1-7-MasR renoprotective effects.
Collapse
Affiliation(s)
- Zeba Farooqui
- Heart and Kidney Institute, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Anees Ahmad Banday
- Heart and Kidney Institute, College of Pharmacy, University of Houston, Houston, TX 77204, USA.
| |
Collapse
|
11
|
Espartero A, Vidal A, Lopez I, Raya AI, Rodriguez M, Aguilera-Tejero E, Pineda C. Rapamycin downregulates α-klotho in the kidneys of female rats with normal and reduced renal function. PLoS One 2023; 18:e0294791. [PMID: 38015969 PMCID: PMC10684065 DOI: 10.1371/journal.pone.0294791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/07/2023] [Indexed: 11/30/2023] Open
Abstract
Both mTOR and α-klotho play a role in the pathophysiology of renal disease, influence mineral metabolism and participate in the aging process. The influence of mTOR inhibition by rapamycin on renal α-klotho expression is unknown. Rats with normal (controls) and reduced (Nx) renal function were treated with rapamycin, 1.3 mg/kg/day, for 22 days. The experiments were conducted with rats fed 0.6% P diet (NP) and 0.2% P diet (LP). Treatment with rapamycin promoted phosphaturia in control and Nx rats fed NP and LP. A decrease in FGF23 was identified in controls after treatment with rapamycin. In rats fed NP, rapamycin decreased mRNA α-klotho/GADPH ratio both in controls, 0.6±0.1 vs 1.1±0.1, p = 0.001, and Nx, 0.3±0.1 vs 0.7±0.1, p = 0.01. At the protein level, a significant reduction in α-klotho was evidenced after treatment with rapamycin both by Western Blot: 0.6±0.1 vs 1.0±0.1, p = 0.01, in controls, 0.7±0.1 vs 1.1±0.1, p = 0.02, in Nx; and by immunohistochemistry staining. Renal α-klotho was inversely correlated with urinary P excretion (r = -0.525, p = 0.0002). The decrease in α-klotho after treatment with rapamycin was also observed in rats fed LP. In conclusion, rapamycin increases phosphaturia and down-regulates α-klotho expression in rats with normal and decreased renal function. These effects can be observed in animals ingesting normal and low P diet.
Collapse
Affiliation(s)
- Azahara Espartero
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Angela Vidal
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Ignacio Lopez
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Ana I. Raya
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Mariano Rodriguez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Escolastico Aguilera-Tejero
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Carmen Pineda
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| |
Collapse
|
12
|
Chevalier RL. Why is chronic kidney disease progressive? Evolutionary adaptations and maladaptations. Am J Physiol Renal Physiol 2023; 325:F595-F617. [PMID: 37675460 DOI: 10.1152/ajprenal.00134.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/08/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023] Open
Abstract
Despite significant advances in renal physiology, the global prevalence of chronic kidney disease (CKD) continues to increase. The emergence of multicellular organisms gave rise to increasing complexity of life resulting in trade-offs reflecting ancestral adaptations to changing environments. Three evolutionary traits shape CKD over the lifespan: 1) variation in nephron number at birth, 2) progressive nephron loss with aging, and 3) adaptive kidney growth in response to decreased nephron number. Although providing plasticity in adaptation to changing environments, the cell cycle must function within constraints dictated by available energy. Prioritized allocation of energy available through the placenta can restrict fetal nephrogenesis, a risk factor for CKD. Moreover, nephron loss with aging is a consequence of cell senescence, a pathway accelerated by adaptive nephron hypertrophy that maintains metabolic homeostasis at the expense of increased vulnerability to stressors. Driven by reproductive fitness, natural selection operates in early life but diminishes thereafter, leading to an exponential increase in CKD with aging, a product of antagonistic pleiotropy. A deeper understanding of the evolutionary constraints on the cell cycle may lead to manipulation of the balance between progenitor cell renewal and differentiation, regulation of cell senescence, and modulation of the balance between cell proliferation and hypertrophy. Application of an evolutionary perspective may enhance understanding of adaptation and maladaptation by nephrons in the progression of CKD, leading to new therapeutic advances.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, The University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
13
|
Liu T, Jin Q, Yang L, Mao H, Ma F, Wang Y, Li P, Zhan Y. Regulation of autophagy by natural polyphenols in the treatment of diabetic kidney disease: therapeutic potential and mechanism. Front Endocrinol (Lausanne) 2023; 14:1142276. [PMID: 37635982 PMCID: PMC10448531 DOI: 10.3389/fendo.2023.1142276] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Diabetic kidney disease (DKD) is a major microvascular complication of diabetes and a leading cause of end-stage renal disease worldwide. Autophagy plays an important role in maintaining cellular homeostasis in renal physiology. In DKD, the accumulation of advanced glycation end products induces decreased renal autophagy-related protein expression and transcription factor EB (TFEB) nuclear transfer, leading to impaired autophagy and lysosomal function and blockage of autophagic flux. This accelerates renal resident cell injury and apoptosis, mediates macrophage infiltration and phenotypic changes, ultimately leading to aggravated proteinuria and fibrosis in DKD. Natural polyphenols show promise in treating DKD by regulating autophagy and promoting nuclear transfer of TFEB and lysosomal repair. This review summarizes the characteristics of autophagy in DKD, and the potential application and mechanisms of some known natural polyphenols as autophagy regulators in DKD, with the goal of contributing to a deeper understanding of natural polyphenol mechanisms in the treatment of DKD and promoting the development of their applications. Finally, we point out the limitations of polyphenols in current DKD research and provide an outlook for their future research.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Jin
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Yongli Zhan
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Huynh C, Ryu J, Lee J, Inoki A, Inoki K. Nutrient-sensing mTORC1 and AMPK pathways in chronic kidney diseases. Nat Rev Nephrol 2023; 19:102-122. [PMID: 36434160 DOI: 10.1038/s41581-022-00648-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2022] [Indexed: 11/27/2022]
Abstract
Nutrients such as glucose, amino acids and lipids are fundamental sources for the maintenance of essential cellular processes and homeostasis in all organisms. The nutrient-sensing kinases mechanistic target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) are expressed in many cell types and have key roles in the control of cell growth, proliferation, differentiation, metabolism and survival, ultimately contributing to the physiological development and functions of various organs, including the kidney. Dysregulation of these kinases leads to many human health problems, including cancer, neurodegenerative diseases, metabolic disorders and kidney diseases. In the kidney, physiological levels of mTOR and AMPK activity are required to support kidney cell growth and differentiation and to maintain kidney cell integrity and normal nephron function, including transport of electrolytes, water and glucose. mTOR forms two functional multi-protein kinase complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Hyperactivation of mTORC1 leads to podocyte and tubular cell dysfunction and vulnerability to injury, thereby contributing to the development of chronic kidney diseases, including diabetic kidney disease, obesity-related kidney disease and polycystic kidney disease. Emerging evidence suggests that targeting mTOR and/or AMPK could be an effective therapeutic approach to controlling or preventing these diseases.
Collapse
Affiliation(s)
- Christopher Huynh
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jaewhee Ryu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jooho Lee
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Ayaka Inoki
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Santos AL, Sinha S. Ageing, Metabolic Dysfunction, and the Therapeutic Role of Antioxidants. Subcell Biochem 2023; 103:341-435. [PMID: 37120475 DOI: 10.1007/978-3-031-26576-1_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
The gradual ageing of the world population has been accompanied by a dramatic increase in the prevalence of obesity and metabolic diseases, especially type 2 diabetes. The adipose tissue dysfunction associated with ageing and obesity shares many common physiological features, including increased oxidative stress and inflammation. Understanding the mechanisms responsible for adipose tissue dysfunction in obesity may help elucidate the processes that contribute to the metabolic disturbances that occur with ageing. This, in turn, may help identify therapeutic targets for the treatment of obesity and age-related metabolic disorders. Because oxidative stress plays a critical role in these pathological processes, antioxidant dietary interventions could be of therapeutic value for the prevention and/or treatment of age-related diseases and obesity and their complications. In this chapter, we review the molecular and cellular mechanisms by which obesity predisposes individuals to accelerated ageing. Additionally, we critically review the potential of antioxidant dietary interventions to counteract obesity and ageing.
Collapse
Affiliation(s)
- Ana L Santos
- IdISBA - Fundación de Investigación Sanitaria de las Islas Baleares, Palma, Spain.
| | | |
Collapse
|
16
|
Watany MM, El-Horany HE, Elhosary MM, Elhadidy AA. Clinical application of RUBCN/SESN2 mediated inhibition of autophagy as biomarkers of diabetic kidney disease. Mol Med 2022; 28:147. [PMID: 36476132 PMCID: PMC9730641 DOI: 10.1186/s10020-022-00580-8] [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: 07/04/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Deregulated autophagy in diabetes has been a field of many experimental studies recently. Impaired autophagy in diabetic kidneys orchestrates every step of diabetic nephropathy (DN) pathogenesis. This study aimed to evaluate three autophagy regulators; RUBCN, mTOR, and SESN2 as clinically applicable indicators of DN progression and as early predictors of DN. METHODS This retrospective study included 120 participants in 4 groups; G1: diabetic patients without albuminuria, G2: diabetic patients with microalbuminuria, G3: diabetic patients with macroalbuminuria and G4: healthy controls. RUBCN and SESN2 genes expression were tested by RT-qPCR. RUBCN, mTOR, and SESN2 serum proteins were quantitated by ELISA. RESULTS RUBCN mRNA was over-expressed in diabetic patients relative to controls with the highest level found in G3 followed by G2 then G1; (9.04 ± 0.64, 5.18 ± 0.73, 1.94 ± 0.41 respectively. P < 0.001). SESN2 mRNA expression was at its lowest level in G3 followed by G2 then G1 (0.1 ± 0.06, 0.48 ± 0.11, 0.78 ± 0.13 respectively. P < 0.001). Similar parallel reduction in serum SENS2 was observed. Serum RUBCN and mTOR were significantly elevated in diabetic patients compared to controls, with the increase parallel to albuminuria degree. RUBCN expression, serum RUBCN and mTOR strongly correlated with albuminuria (r = 0.912, 0.925 and 0.867 respectively). SESN2 expression and serum level negatively correlated with albuminuria (r = - 0.897 and -0.828 respectively); (All p < 0.001). Regression analysis showed that serum RUBCN, mTOR, RUBCN and SESN2 mRNAs could successfully predict DN. CONCLUSIONS The study proves the overexpression of RUBCN and mTOR in DN and the down-expression of SESN2. The three markers can be clinically used to predict DN and to monitor disease progression.
Collapse
Affiliation(s)
- Mona M. Watany
- grid.412258.80000 0000 9477 7793Clinical Pathology Department, Faculty of Medicine, Tanta University, El Geish Street, Tanta, 31527 El-Gharbia Governorate Egypt
| | - Hemat E. El-Horany
- grid.412258.80000 0000 9477 7793Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, 31527 Egypt ,grid.443320.20000 0004 0608 0056Biochemistry Department, College of Medicine, Ha’il University, Ha’il, 55211 Saudi Arabia
| | - Marwa M. Elhosary
- grid.412258.80000 0000 9477 7793Msc Immunology from Tanta Faculty of Science, Tanta, 31527 Egypt
| | - Ahmed A. Elhadidy
- grid.412258.80000 0000 9477 7793Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta, 31527 Egypt
| |
Collapse
|
17
|
Abou Daher A, Alkhansa S, Azar WS, Rafeh R, Ghadieh HE, Eid AA. Translational Aspects of the Mammalian Target of Rapamycin Complexes in Diabetic Nephropathy. Antioxid Redox Signal 2022; 37:802-819. [PMID: 34544257 DOI: 10.1089/ars.2021.0217] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Despite the many efforts put into understanding diabetic nephropathy (DN), direct treatments for DN have yet to be discovered. Understanding the mechanisms behind DN is an essential step in the development of novel therapeutic regimens. The mammalian target of rapamycin (mTOR) pathway has emerged as an important candidate in the quest for drug discovery because of its role in regulating growth, proliferation, as well as protein and lipid metabolism. Recent Advances: Kidney cells have been found to rely on basal autophagy for survival and for conserving kidney integrity. Recent studies have shown that diabetes induces renal autophagy deregulation, leading to kidney injury. Hyper-activation of the mTOR pathway and oxidative stress have been suggested to play a role in diabetes-induced autophagy imbalance. Critical Issues: A detailed understanding of the role of mTOR signaling in diabetes-associated complications is of major importance in the search for a cure. In this review, we provide evidence that mTOR is heavily implicated in diabetes-induced kidney injury. We suggest possible mechanisms through which mTOR exerts its negative effects by increasing insulin resistance, upregulating oxidative stress, and inhibiting autophagy. Future Directions: Both increased oxidative stress and autophagy deregulation are deeply embedded in DN. However, the mechanisms controlling oxidative stress and autophagy are not well understood. Although Akt/mTOR signaling seems to play an important role in oxidative stress and autophagy, further investigation is required to uncover the details of this signaling pathway. Antioxid. Redox Signal. 37, 802-819.
Collapse
Affiliation(s)
- Alaa Abou Daher
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Sahar Alkhansa
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - William S Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,Department of Physiology and Biophysics, Georgetown University Medical School, Washington, District of Columbia, USA
| | - Rim Rafeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Hilda E Ghadieh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
18
|
Zhou X, Zhang B, Zhao X, Lin Y, Zhuang Y, Guo J, Wang S. Chlorogenic Acid Prevents Hyperuricemia Nephropathy via Regulating TMAO-Related Gut Microbes and Inhibiting the PI3K/AKT/mTOR Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10182-10193. [PMID: 35950815 DOI: 10.1021/acs.jafc.2c03099] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Hyperuricemia is an independent hazard factor of renal injury and can induce renal fibrosis, promoting the development of chronic kidney disease (CKD). This study aimed to explore the probability of chlorogenic acid (CGA) as a potential substance for preventing hyperuricemia nephropathy (HN). Pretreatment with CGA downregulated SUA, BUN, and CR levels, relieved oxidative stress and inflammatory response, alleviated kidney fibrosis, and contributed to the prevention of HN. In the gut microbiota, Blautia, Enterococcus, and Faecalibaculum related to trimethylamine N-oxide (TMAO) synthesis were significantly increased in HN rats. In addition, it showed a significant increase in serum TMAO content in HN rats. However, CGA regulated the cascade response of the microbiota-TMAO signaling to reverse the increase of serum TMAO. CGA also decreased the protein expression of protein kinase B (AKT) phosphorylation, phosphatidylinositide 3-kinase (PI3K), and mammalian target of rapamycin (mTOR) by reducing the production of TMAO. CGA delayed kidney fibrosis in HN rats as evidenced by regulating the cascade response of the microbiota-TMAO-PI3K/AKT/mTOR signaling pathway. In summary, CGA can be an excellent candidate for HN prevention.
Collapse
Affiliation(s)
- Xiaofei Zhou
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Bowei Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiuli Zhao
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yongxi Lin
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yuan Zhuang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jingting Guo
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shuo Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
19
|
Lin Y, Yang Q, Wang J, Chen X, Liu Y, Zhou T. An overview of the efficacy and signaling pathways activated by stem cell-derived extracellular vesicles in diabetic kidney disease. Front Endocrinol (Lausanne) 2022; 13:962635. [PMID: 35966088 PMCID: PMC9366010 DOI: 10.3389/fendo.2022.962635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/06/2022] [Indexed: 02/05/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of complications of diabetes mellitus with severe microvascular lesion and the most common cause of end-stage chronic kidney disease (ESRD). Controlling serum glucose remains the primary approach to preventing and slowing the progression of DKD. Despite considerable efforts to control diabetes, people with diabetes develop not only DKD but also ESRD. The pathogenesis of DKD is very complex, and current studies indicate that mesenchymal stromal cells (MSCs) regulate complex disease processes by promoting pro-regenerative mechanisms and inhibiting multiple pathogenic pathways. Extracellular vesicles (EVs) are products of MSCs. Current data indicate that MSC-EVs-based interventions not only protect renal cells, including renal tubular epithelial cells, podocytes and mesangial cells, but also improve renal function and reduce damage in diabetic animals. As an increasing number of clinical studies have confirmed, MSC-EVs may be an effective way to treat DKD. This review explores the potential efficacy and signaling pathways of MSC-EVs in the treatment of DKD.
Collapse
Affiliation(s)
- Yongda Lin
- Department of Nephrology, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | | | | | | | | | - Tianbiao Zhou
- Department of Nephrology, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| |
Collapse
|
20
|
Das F, Ghosh-Choudhury N, Maity S, Kasinath BS, Choudhury GG. Oncoprotein DJ-1 interacts with mTOR complexes to effect transcription factor Hif1α-dependent expression of collagen I (α2) during renal fibrosis. J Biol Chem 2022; 298:102246. [PMID: 35835217 PMCID: PMC9399488 DOI: 10.1016/j.jbc.2022.102246] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/27/2022] Open
Abstract
Proximal tubular epithelial cells respond to transforming growth factor β (TGFβ) to synthesize collagen I (α2) during renal fibrosis. The oncoprotein DJ-1 has previously been shown to promote tumorigenesis and prevent apoptosis of dopaminergic neurons; however, its role in fibrosis signaling is unclear. Here, we show TGFβ-stimulation increased expression of DJ-1, which promoted noncanonical mTORC1 and mTORC2 activities. We show DJ-1 augmented the phosphorylation/activation of PKCβII, a direct substrate of mTORC2. In addition, coimmunoprecipitation experiments revealed association of DJ-1 with Raptor and Rictor, exclusive subunits of mTORC1 and mTORC2, respectively, as well as with mTOR kinase. Interestingly, siRNAs against DJ-1 blocked TGFβ-stimulated expression of collagen I (α2), while expression of DJ-1 increased expression of this protein. In addition, expression of dominant negative PKCβII and siRNAs against PKCβII significantly inhibited TGFβ-induced collagen I (α2) expression. In fact, constitutively active PKCβII abrogated the effect of siRNAs against DJ-1, suggesting a role of PKCβII downstream of this oncoprotein. Moreover, we demonstrate expression of collagen I (α2) stimulated by DJ-1 and its target PKCβII is dependent on the transcription factor hypoxia-inducible factor 1α (Hif1α). Finally, we show in the renal cortex of diabetic rats that increased TGFβ was associated with enhanced expression of DJ-1 and activation of mTOR and PKCβII, concomitant with increased Hif1α and collagen I (α2). Overall, we identified that DJ-1 affects TGFβ-induced expression of collagen I (α2) via an mTOR-, PKCβII-, and Hif1α-dependent mechanism to regulate renal fibrosis.
Collapse
Affiliation(s)
- Falguni Das
- VA Research, South Texas Veterans Health Care System, San Antonio, Texas; Department of Medicine, UT Health San Antonio, Texas
| | | | - Soumya Maity
- Department of Medicine, UT Health San Antonio, Texas
| | | | - Goutam Ghosh Choudhury
- VA Research, South Texas Veterans Health Care System, San Antonio, Texas; Department of Medicine, UT Health San Antonio, Texas; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas.
| |
Collapse
|
21
|
Mechanisms of podocyte injury and implications for diabetic nephropathy. Clin Sci (Lond) 2022; 136:493-520. [PMID: 35415751 PMCID: PMC9008595 DOI: 10.1042/cs20210625] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/25/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Albuminuria is the hallmark of both primary and secondary proteinuric glomerulopathies, including focal segmental glomerulosclerosis (FSGS), obesity-related nephropathy, and diabetic nephropathy (DN). Moreover, albuminuria is an important feature of all chronic kidney diseases (CKDs). Podocytes play a key role in maintaining the permselectivity of the glomerular filtration barrier (GFB) and injury of the podocyte, leading to foot process (FP) effacement and podocyte loss, the unifying underlying mechanism of proteinuric glomerulopathies. The metabolic insult of hyperglycemia is of paramount importance in the pathogenesis of DN, while insults leading to podocyte damage are poorly defined in other proteinuric glomerulopathies. However, shared mechanisms of podocyte damage have been identified. Herein, we will review the role of haemodynamic and oxidative stress, inflammation, lipotoxicity, endocannabinoid (EC) hypertone, and both mitochondrial and autophagic dysfunction in the pathogenesis of the podocyte damage, focussing particularly on their role in the pathogenesis of DN. Gaining a better insight into the mechanisms of podocyte injury may provide novel targets for treatment. Moreover, novel strategies for boosting podocyte repair may open the way to podocyte regenerative medicine.
Collapse
|
22
|
Yang L, Zhang Z, Wang D, Jiang Y, Liu Y. Targeting mTOR Signaling in Type 2 Diabetes Mellitus and Diabetes Complications. Curr Drug Targets 2022; 23:692-710. [PMID: 35021971 DOI: 10.2174/1389450123666220111115528] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 11/22/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is a pivotal regulator of cell metabolism and growth. In the form of two different multi-protein complexes, mTORC1 and mTORC2, mTOR integrates cellular energy, nutrient and hormonal signals to regulate cellular metabolic homeostasis. In type 2 diabetes mellitus (T2DM) aberrant mTOR signaling underlies its pathological conditions and end-organ complications. Substantial evidence suggests that two mTOR-mediated signaling schemes, mTORC1-p70S6 kinase 1 (S6K1) and mTORC2-protein kinase B (AKT), play a critical role in insulin sensitivity and that their dysfunction contributes to development of T2DM. This review summaries our current understanding of the role of mTOR signaling in T2DM and its associated complications, as well as the potential use of mTOR inhibitors in treatment of T2DM.
Collapse
Affiliation(s)
- Lin Yang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhixin Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Doudou Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Ying Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
23
|
High glucose-stimulated enhancer of zeste homolog-2 (EZH2) forces suppression of deptor to cause glomerular mesangial cell pathology. Cell Signal 2021; 86:110072. [PMID: 34224844 DOI: 10.1016/j.cellsig.2021.110072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 11/24/2022]
Abstract
Function of mTORC1 and mTORC2 has emerged as a driver of mesangial cell pathologies in diabetic nephropathy. The mechanism of mTOR activation is poorly understood in this disease. Deptor is a constitutive subunit and a negative regulator of both mTOR complexes. Mechanistic investigation in mesangial cells revealed that high glucose decreased the expression of deptor concomitant with increased mTORC1 and mTORC2 activities, induction of hypertrophy and, expression of fibronectin and PAI-1. shRNAs against deptor mimicked these pathologic outcomes of high glucose. Conversely, overexpression of deptor significantly inhibited all effects of high glucose. To determine the mechanism of deptor suppression, we found that high glucose significantly increased the expression of EZH2, resulting in lysine-27 tri-methylation of histone H3 (H3K27Me3). Employing approaches including pharmacological inhibition, shRNA-mediated downregulation and overexpression of EZH2, we found that EZH2 regulates high glucose-induced deptor suppression along with activation of mTOR, mesangial cell hypertrophy and fibronectin/PAI-1 expression. Moreover, expression of hyperactive mTORC1 reversed shEZH2-mediated inhibition of hypertrophy and expression of fibronectin and PAI-1 by high glucose. Finally, in renal cortex of diabetic mice, we found that enhanced expression of EZH2 is associated with decreased deptor levels and increased mTOR activity and, expression of fibronectin and PAI-1. Together, our findings provide a novel mechanism for mTOR activation via EZH2 to induce mesangial cell hypertrophy and matrix expansion during early progression of diabetic nephropathy. These results suggest a strategy for leveraging the intrinsic effect of deptor to suppress mTOR activity via reducing EZH2 as a novel therapy for diabetic nephropathy.
Collapse
|
24
|
Dissecting the Involvement of Ras GTPases in Kidney Fibrosis. Genes (Basel) 2021; 12:genes12060800. [PMID: 34073961 PMCID: PMC8225075 DOI: 10.3390/genes12060800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/30/2022] Open
Abstract
Many different regulatory mechanisms of renal fibrosis are known to date, and those related to transforming growth factor-β1 (TGF-β1)-induced signaling have been studied in greater depth. However, in recent years, other signaling pathways have been identified, which contribute to the regulation of these pathological processes. Several studies by our team and others have revealed the involvement of small Ras GTPases in the regulation of the cellular processes that occur in renal fibrosis, such as the activation and proliferation of myofibroblasts or the accumulation of extracellular matrix (ECM) proteins. Intracellular signaling mediated by TGF-β1 and Ras GTPases are closely related, and this interaction also occurs during the development of renal fibrosis. In this review, we update the available in vitro and in vivo knowledge on the role of Ras and its main effectors, such as Erk and Akt, in the cellular mechanisms that occur during the regulation of kidney fibrosis (ECM synthesis, accumulation and activation of myofibroblasts, apoptosis and survival of tubular epithelial cells), as well as the therapeutic strategies for targeting the Ras pathway to intervene on the development of renal fibrosis.
Collapse
|
25
|
Osoro EK, Du X, Liang D, Lan X, Farooq R, Huang F, Zhu W, Ren J, Sadiq M, Tian L, Yang X, Li D, Lu S. Induction of PDCD4 by albumin in proximal tubule epithelial cells potentiates proteinuria-induced dysfunctional autophagy by negatively targeting Atg5. Biochem Cell Biol 2021; 99:617-628. [PMID: 33831322 DOI: 10.1139/bcb-2021-0028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The precise molecular mechanism of autophagy dysfunction in type 1 diabetes is not known. Herein, the role of programmed cell death 4 (PDCD4) in autophagy regulation in the pathogenesis of diabetic kidney disease (DKD) in vivo and in vitro was described. It was found that Pdcd4 mRNA and protein was upregulated in the streptozotocin (STZ)-induced DKD rats. In addition, a unilateral ureteral obstruction mouse model displayed an upregulation of PDCD4 in the disease group. kidney biopsy samples of human DKD patients showed an upregulation of PDCD4. Furthermore, western blotting of the STZ-induced DKD rat tissues displayed a low microtubule-associated protein 1A/1B-light chain 3 (LC3)-II, as compared to the control. It was found that albumin overload in cultured PTEC could upregulate the expression of PDCD4 and p62, and decrease the expression of LC3-II and autophagy-related 5 (Atg5) proteins. The knockout of Pdcd4 in cultured PTECs could lessen albumin-induced dysfunctional autophagy as evidenced by the recovery of Atg5 and LC3-II protein. The forced expression of PDCD4 could further suppress the expression of crucial autophagy-related gene Atg5. Herein, endogenous PDCD4 was shown to promote proteinuria-induced dysfunctional autophagy by negatively regulating Atg5. PDCD4 might therefore be a potential therapeutic target in DKD.
Collapse
Affiliation(s)
- Ezra Kombo Osoro
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Xiaojuan Du
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Dong Liang
- Xi'an Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Xi Lan
- Xi'an Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Riaz Farooq
- Xi'an Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Fumeng Huang
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Wenhua Zhu
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Jiajun Ren
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Muhammad Sadiq
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Lifang Tian
- Xi'an Jiaotong University, 12480, Department of Nephrology, the Second Affiliated Hospital, Xi'an, Shaanxi, China;
| | - Xudong Yang
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Dongmin Li
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Shemin Lu
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| |
Collapse
|
26
|
Hu C, Zhao Y, Wang X, Zhu T. Intratumoral Fibrosis in Facilitating Renal Cancer Aggressiveness: Underlying Mechanisms and Promising Targets. Front Cell Dev Biol 2021; 9:651620. [PMID: 33777960 PMCID: PMC7991742 DOI: 10.3389/fcell.2021.651620] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 02/05/2021] [Indexed: 01/01/2023] Open
Abstract
Intratumoral fibrosis is a histologic manifestation of fibrotic tumor stroma. The interaction between cancer cells and fibrotic stroma is intricate and reciprocal, involving dysregulations from multiple biological processes. Different components of tumor stroma are implicated via distinct manners. In the kidney, intratumoral fibrosis is frequently observed in renal cell carcinoma (RCC). However, the underlying mechanisms remain largely unclear. In this review, we recapitulate evidence demonstrating how fibrotic stroma interacts with cancer cells and mechanisms shared between RCC tumorigenesis and renal fibrogenesis, providing promising targets for future studies.
Collapse
Affiliation(s)
- Chao Hu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yufeng Zhao
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Xuanchuan Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| |
Collapse
|
27
|
Kim KH, Hong GL, Jung DY, Karunasagara S, Jeong WI, Jung JY. IL-17 deficiency aggravates the streptozotocin-induced diabetic nephropathy through the reduction of autophagosome formation in mice. Mol Med 2021; 27:25. [PMID: 33691614 PMCID: PMC7945049 DOI: 10.1186/s10020-021-00285-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 03/02/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is one of the most important medical complications of diabetes mellitus. Autophagy is an important mediator of pathological response and plays a critical role in inflammation during the progression of diabetic nephropathy. Interleukin (IL)-17A favorably modulates inflammatory disorders including DN. In this study, we examined whether IL-17A deficiency affected the autophagy process in the kidneys of mice with streptozotocin (STZ)-induced DN. METHODS The autophagic response of IL-17A to STZ-induced nephrotoxicity was evaluated by analyzing STZ-induced functional and histological renal injury in IL-17A knockout (KO) mice. RESULTS IL-17A KO STZ-treated mice developed more severe nephropathy than STZ-treated wild-type (WT) mice, with increased glomerular damage and renal interstitial fibrosis at 12 weeks. IL-17A deficiency also increased the up-regulation of proinflammatory cytokines and fibrotic gene expression after STZ treatment. Meanwhile, autophagy-associated proteins were induced in STZ-treated WT mice. However, IL-17A KO STZ-treated mice displayed a significant decrease in protein expression. Especially, the levels of LC3 and ATG7, which play crucial roles in autophagosome formation, were notably decreased in the IL-17A KO STZ-treated mice compared with their WT counterparts. CONCLUSIONS IL-17 deficiency aggravates of STZ-induced DN via attenuation of autophagic response. Our study demonstrated that IL-17A mediates STZ-induced renal damage and represents a potential therapeutic target in DN.
Collapse
Affiliation(s)
- Kyung-Hyun Kim
- Department of Veterinary Medicine, Institute of Veterinary Science, Chungnam National University, 99, Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Geum-Lan Hong
- Department of Veterinary Medicine, Institute of Veterinary Science, Chungnam National University, 99, Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Da-Young Jung
- Department of Veterinary Medicine, Institute of Veterinary Science, Chungnam National University, 99, Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Shanika Karunasagara
- Department of Veterinary Medicine, Institute of Veterinary Science, Chungnam National University, 99, Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Ju-Young Jung
- Department of Veterinary Medicine, Institute of Veterinary Science, Chungnam National University, 99, Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
28
|
Roles of mTOR in Diabetic Kidney Disease. Antioxidants (Basel) 2021; 10:antiox10020321. [PMID: 33671526 PMCID: PMC7926630 DOI: 10.3390/antiox10020321] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease and the number of patients affected is increasing worldwide. Thus, there is a need to establish a new treatment for DKD to improve the renal prognosis of diabetic patients. Recently, it has shown that intracellular metabolic abnormalities are involved in the pathogenesis of DKD. In particular, the activity of mechanistic target of rapamycin complex 1 (mTORC1), a nutrient-sensing signaling molecule, is hyperactivated in various organs of diabetic patients, which suggests the involvement of excessive mTORC1 activation in the pathogenesis of diabetes. In DKD, hyperactivated mTORC1 may be involved in the pathogenesis of podocyte damage, which causes proteinuria, and tubular cell injury that decreases renal function. Therefore, elucidating the role of mTORC1 in DKD and developing new therapeutic agents that suppress mTORC1 hyperactivity may shed new light on DKD treatments in the future.
Collapse
|
29
|
Uil M, Hau CM, Ahdi M, Mills JD, Kers J, Saleem MA, Florquin S, Gerdes VEA, Nieuwland R, Roelofs JJTH. Cellular origin and microRNA profiles of circulating extracellular vesicles in different stages of diabetic nephropathy. Clin Kidney J 2021; 14:358-365. [PMID: 33564439 PMCID: PMC7857783 DOI: 10.1093/ckj/sfz145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/13/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a major complication of diabetes and the main cause of end-stage renal disease. Extracellular vesicles (EVs) are small cell-derived vesicles that can alter disease progression by microRNA (miRNA) transfer. METHODS In this study, we aimed to characterize the cellular origin and miRNA content of EVs in plasma samples of type 2 diabetes patients at various stages of DN. Type 2 diabetes patients were classified in three groups: normoalbuminuria, microalbuminuria and macroalbuminuria. The concentration and cellular origin of plasma EVs were measured by flow cytometry. A total of 752 EV miRNAs were profiled in 18 subjects and differentially expressed miRNAs were validated. RESULTS Diabetic patients with microalbuminuria and/or macroalbuminuria showed elevated concentrations of total EVs and EVs from endothelial cells, platelets, leucocytes and erythrocytes compared with diabetic controls. miR-99a-5p was upregulated in macroalbuminuric patients compared with normoalbuminuric and microalbuminuric patients. Transfection of miR-99a-5p in cultured human podocytes downregulated mammalian target of rapamycin (mTOR) protein expression and downregulated the podocyte injury marker vimentin. CONCLUSIONS Type 2 diabetes patients with microalbuminuria and macroalbuminuria display differential EV profiles. miR-99a-5p expression is elevated in EVs from macroalbuminuria and mTOR is its validated mRNA target.
Collapse
Affiliation(s)
- Melissa Uil
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Chi M Hau
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Vesicle Observation Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mohamed Ahdi
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - James D Mills
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jesper Kers
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Van’t Hoff Institute for Molecular Sciences (HIMS), University of Amsterdam, Amsterdam, The Netherlands
| | - Moin A Saleem
- Academic Renal Unit, University of Bristol, Southmead Hospital, Bristol, UK
| | - Sandrine Florquin
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Victor E A Gerdes
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Vesicle Observation Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Chen N, Mu L, Yang Z, Du C, Wu M, Song S, Yuan C, Shi Y. Carbohydrate response element-binding protein regulates lipid metabolism via mTOR complex1 in diabetic nephropathy. J Cell Physiol 2021; 236:625-640. [PMID: 32583421 DOI: 10.1002/jcp.29890] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
Lipid deposition caused by the disorder of renal lipid metabolism is involved in diabetic nephropathy (DN). Carbohydrate response element-binding protein (ChREBP) is a key transcription factor in high glucose-induced cellular fat synthesis. At present, the regulation and mechanism of ChREBP on fat metabolism in diabetic kidneys are still unclear. In this study, we showed that lack of ChREBP significantly improved renal injury, inhibited oxidative stress, lipid deposition, fatty acid synthase (FASN), acetyl-CoA carboxylase (ACC) and thioredoxin-interacting protein (TXNIP) expression, as well as the activity of mammalian target of rapamycin complex 1 (mTORC1) in diabetic kidneys. Meanwhile, ChREBP deficiency upregulated the expression of peroxisome proliferator-activated receptor-α (PPARα), carnitine palmitoyltransferaser 1A (CPT1A) and acyl-coenzyme A oxidase 1 (ACOX1) in diabetic kidneys. In vitro, knockdown of ChREBP attenuated lipid deposition, mTORC1 activation, and expression of FASN and ACC, increased PPARα, CPT1A, and ACOX1 expression in HK-2 cells and podocytes under high glucose (HG) conditions. Moreover, HG-induced lipid deposition, increased expression of FASN and ACC and decreased expression of PPARα, CPT1A, and ACOX1 were reversed by rapamycin, a specific inhibitor of mTORC1, in HK-2 cells. These results indicate that ChREBP deficiency alleviates diabetes-associated renal lipid accumulation by inhibiting mTORC1 activity and suggest that reduction of ChREBP is a potential therapeutic strategy to treat DN.
Collapse
Affiliation(s)
- Nan Chen
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
| | - Lin Mu
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
- Department of Nephrology, Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Zhifen Yang
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Chunyang Du
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
| | - Ming Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
| | - Chen Yuan
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
| |
Collapse
|
31
|
Chen A, Wang H, Su Y, Zhang C, Qiu Y, Zhou Y, Wan Y, Hu B, Li Y. Exosomes: Biomarkers and Therapeutic Targets of Diabetic Vascular Complications. Front Endocrinol (Lausanne) 2021; 12:720466. [PMID: 34456875 PMCID: PMC8387814 DOI: 10.3389/fendo.2021.720466] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/22/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetic vascular complications (DVC) including macrovascular and microvascular lesions, have a significant impact on public health, and lead to increased patient mortality. Disordered intercellular cascades play a vital role in diabetic systemic vasculopathy. Exosomes participate in the abnormal signal transduction of local vascular cells and mediate the transmission of metabolic disorder signal molecules in distant organs and cells through the blood circulation. They can store different signaling molecules in the membrane structure and release them into the blood, urine, and tears. In recent years, the carrier value and therapeutic effect of exosomes derived from stem cells have garnered attention. Exosomes are not only a promising biomarker but also a potential target and tool for the treatment of DVC. This review explored changes in the production process of exosomes in the diabetic microenvironment and exosomes' early warning role in DVC from different systems and their pathological processes. On the basis of these findings, we discussed the future direction of exosomes in the treatment of DVC, and the current limitations of exosomes in DVC research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bo Hu
- *Correspondence: Yanan Li, ; Bo Hu,
| | - Yanan Li
- *Correspondence: Yanan Li, ; Bo Hu,
| |
Collapse
|
32
|
Srivastava SP, Kanasaki K, Goodwin JE. Loss of Mitochondrial Control Impacts Renal Health. Front Pharmacol 2020; 11:543973. [PMID: 33362536 PMCID: PMC7756079 DOI: 10.3389/fphar.2020.543973] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Disruption of mitochondrial biosynthesis or dynamics, or loss of control over mitochondrial regulation leads to a significant alteration in fuel preference and metabolic shifts that potentially affect the health of kidney cells. Mitochondria regulate metabolic networks which affect multiple cellular processes. Indeed, mitochondria have established themselves as therapeutic targets in several diseases. The importance of mitochondria in regulating the pathogenesis of several diseases has been recognized, however, there is limited understanding of mitochondrial biology in the kidney. This review provides an overview of mitochondrial dysfunction in kidney diseases. We describe the importance of mitochondria and mitochondrial sirtuins in the regulation of renal metabolic shifts in diverse cells types, and review this loss of control leads to increased cell-to-cell transdifferentiation processes and myofibroblast-metabolic shifts, which affect the pathophysiology of several kidney diseases. In addition, we examine mitochondrial-targeted therapeutic agents that offer potential leads in combating kidney diseases.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Keizo Kanasaki
- Internal Medicine 1, Shimane University Faculty of Medicine, Izumo, Japan
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
33
|
Nie Y, Fu C, Zhang H, Zhang M, Xie H, Tong X, Li Y, Hou Z, Fan X, Yan M. Celastrol slows the progression of early diabetic nephropathy in rats via the PI3K/AKT pathway. BMC Complement Med Ther 2020; 20:321. [PMID: 33097050 PMCID: PMC7583204 DOI: 10.1186/s12906-020-03050-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022] Open
Abstract
Background Diabetic nephropathy serves as one of the most regular microvascular complications of diabetes mellitus and is the main factor that causes end-stage renal disease and incident mortality. As the beneficial effect and minute adverse influence of Celastrol on the renal system requires further elucidation, the renoprotective function of Celastrol in early diabetic nephropathy was investigated. Methods In high-fat and high-glucose diet/streptozotocin-induced diabetic rats which is the early diabetic nephropathy model, ALT, AST, 24 h urinary protein, blood urea nitrogen, and serum creatinine content were observed. Periodic acid-Schiff staining, enzyme-linked immunosorbent assay, immunohistochemical analysis, reverse transcription-polymerase chain reaction, and western blot analysis were used to explore the renoprotective effect of Celastrol to diabetic nephropathy rats and the underlying mechanism. Results High dose of Celastrol (1.5 mg/kg/d) not only improved the kidney function of diabetic nephropathy (DN) rats, and decreased the blood glucose and 24 h urinary albumin, but also increased the expression of LC3II and nephrin, and downregulated the expression of PI3K, p-AKT, and the mRNA level of NF-κB and mTOR. Conclusion Celastrol functions as a potential therapeutic substance, acting via the PI3K/AKT pathway to attenuate renal injury, inhibit glomerular basement membrane thickening, and achieve podocyte homeostasis in diabetic nephropathy.
Collapse
Affiliation(s)
- Yusong Nie
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.,Xianyang Central Hospital, Xianyang, 712000, Shaanxi, China
| | - Chengxiao Fu
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Huimin Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Min Zhang
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.,First clinical medical college, Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Hui Xie
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Xiaopei Tong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yao Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Zhenyan Hou
- Department of Pharmacy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Xinrong Fan
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China. .,First clinical medical college, Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China. .,Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
34
|
Tang PMK, Zhang YY, Hung JSC, Chung JYF, Huang XR, To KF, Lan HY. DPP4/CD32b/NF-κB Circuit: A Novel Druggable Target for Inhibiting CRP-Driven Diabetic Nephropathy. Mol Ther 2020; 29:365-375. [PMID: 32956626 DOI: 10.1016/j.ymthe.2020.08.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/17/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetic nephropathy (DN) is a major cause of end-stage renal disease, but treatment remains ineffective. C-reactive protein (CRP) is pathogenic in DN, which significantly correlated with dipeptidyl peptidase-4 (DPP4) expression in diabetic patients with unknown reason. Here, using our unique CRPtg-db/db mice, we observed human CRP markedly induced renal DPP4 associated with enhanced kidney injury compared with db/db mice. Interestingly, linagliptin, a US Food and Drug Administration (FDA)-approved specific DPP4 inhibitor, effectively blocked this CRP-driven DN in the CRPtg-db/db mice. Mechanistically, CRP evoked DPP4 in cultured renal tubular epithelial cells, where CD32b/nuclear factor κB (NF-κB) signaling markedly enriched p65 binding on the DPP4 promoter region to increase its transcription. Unexpectedly, we further discovered that CRP triggers dimerization of DPP4 with CD32b at protein level, forming a novel DPP4/CD32b/NF-κB signaling circuit for promoting CRP-mediated DN. More importantly, linagliptin effectively blocked the circuit, thereby inhibiting the CRP/CD32b/NF-κB-driven renal inflammation and fibrosis. Thus, DPP4 may represent a precise druggable target for CRP-driven DN.
Collapse
Affiliation(s)
- Patrick Ming-Kuen Tang
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong; Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong.
| | - Ying-Ying Zhang
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong; Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jessica Shuk-Chun Hung
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Jeff Yat-Fai Chung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Xiao-Ru Huang
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong; Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Hui-Yao Lan
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong; Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
35
|
Fluitt MB, Shivapurkar N, Kumari M, Singh S, Li L, Tiwari S, Ecelbarger CM. Systemic inhibition of miR-451 increases fibrotic signaling and diminishes autophagic response to exacerbate renal damage in Tallyho/Jng mice. Am J Physiol Renal Physiol 2020; 319:F476-F486. [PMID: 32715758 PMCID: PMC7509278 DOI: 10.1152/ajprenal.00594.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
miRNAs provide fine tuning of gene expression via inhibition of translation. miR-451 has a modulatory role in cell cycling via downregulation of mechanistic target of rapamycin. We aimed to test whether chronic systemic inhibition of miR-451 would enhance renal fibrosis (associated with deranged autophagy). Adult TallyHo/Jng mice (obese insulin resistant) were randomized to two treatment groups to receive either miR-451 inhibition [via a locked nucleic acid construct] or a similar scrambled locked nucleic acid control for 8 wk. All mice were fed a high-fat diet (60% kcal from fat) ad libitum and humanely euthanized after 12 wk. Kidneys and blood were collected for analysis. Renal expression of miR-451 was sixfold lower in inhibitor-treated mice compared with control mice. miR-451 inhibition increased kidney weight and collagen and glycogen deposition. Blood chemistry revealed significantly higher Na+ and anion gap (relative metabolic acidosis) in inhibitor-treated mice. Western blot analysis and immunohistochemistry of the kidney revealed that the inhibitor increased markers of renal injury and fibrosis, e.g., kidney injury molecule 1, neutrophil gelatinase-associated lipocalin, transforming growth factor-β, 14-3-3 protein-ζ, mechanistic target of rapamycin, AMP-activated protein kinase-α, calcium-binding protein 39, matrix metallopeptidase-9, and the autophagy receptor sequestosome 1. In contrast, the inhibitor reduced the epithelial cell integrity marker collagen type IV and the autophagy markers microtubule-associated protein 1A/1B light chain 3B and beclin-1. Taken together, these results support a protective role for miR-451 in reducing renal fibrosis by enhancing autophagy in obese mice.
Collapse
Affiliation(s)
- Maurice B. Fluitt
- 1Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Narayan Shivapurkar
- 1Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Manju Kumari
- 2Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Sarojini Singh
- 2Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Lijun Li
- 1Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Swasti Tiwari
- 2Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Carolyn M. Ecelbarger
- 1Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, District of Columbia
| |
Collapse
|
36
|
Karunasagara S, Hong GL, Park SR, Lee NH, Jung DY, Kim TW, Jung JY. Korean red ginseng attenuates hyperglycemia-induced renal inflammation and fibrosis via accelerated autophagy and protects against diabetic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2020; 254:112693. [PMID: 32112899 DOI: 10.1016/j.jep.2020.112693] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/10/2020] [Accepted: 02/20/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax ginseng C.A. Mey. (Korean ginseng) has been widely used in traditional medicine to treat diabetes mellitus for thousands of years. It also plays a key role in health maintenance owing to its anti-oxidant and anti-fatigue properties, and is quite popular as a dietary supplement. AIM OF THE STUDY This study was designed to offer a complementary and alternative medicine to manage the diabetic kidney disease (DKD), which causes long-term damage to the renal structure. We also investigated the regulation of the autophagy mechanism, which is the underlying the pathogenesis of DKD. MATERIALS AND METHODS The effect of Korean red ginseng (KRG) on DKD was evaluated using human kidney proximal tubular cells and streptozotocin (STZ)-treated Sprague-Dawley rat models. In vitro experiments were conducted to evaluate the proteins related to fibrosis and autophagy. This was followed by in vivo experiments involving rats treated with single intraperitoneal administration of STZ (60 mg/kg) and then with KRG solution orally for 4 weeks. Proteins related to renal injury, fibrosis, and autophagy were determined by immunoblotting. Hematoxylin and eosin (H&E), Periodic acid-Schiff (PAS), Sirius red, and immunostaining were processed for histological studies. RESULTS KRG diminished the levels of metabolic measurements and blood parameters. Western blotting showed a decreased expression of proteins, such as TGF-β1, KIM1, and AGE, which are responsible for renal inflammation, injury, and fibrosis. Histological studies also supported these results and revealed that the KRG-treated groups recovered from renal injury and fibrosis. Furthermore, the autophagy marker, LC3, was upregulated, whereas p62 was downregulated. The levels of proteins related to the autophagy mechanism, such as ATG7, increased, while mammalian target of rapamycin (mTOR) decreased with the KRG treatment and exhibited accelerated autophagy compared to the STZ alone group. CONCLUSIONS KRG can suppress renal inflammation, injury, and fibrosis by blocking TGF-β1 activation and can induce cellular autophagy. Therefore, this study strongly suggests that KRG exhibits a renoprotective effect against the STZ-induced DKD.
Collapse
Affiliation(s)
- Shanika Karunasagara
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Geum-Lan Hong
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Se-Ra Park
- Samsung Biomedical Research Institute, Seoul, 06351, Republic of Korea
| | - Na-Hyun Lee
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Da-Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Tae-Won Kim
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Ju-Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
37
|
Autophagy and mTOR Pathways Mediate the Potential Renoprotective Effects of Vitamin D on Diabetic Nephropathy. Int J Nephrol 2020; 2020:7941861. [PMID: 32455017 PMCID: PMC7243019 DOI: 10.1155/2020/7941861] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Not only is diabetic nephropathy (DN) the most common cause of end-stage renal disease worldwide, but it also increases the risk of mortality up to fourteen times compared to normoalbuminuric diabetic patients. Aim The aim of the current study was the evaluation of the renoprotective effects of vitamin D in DN and the possible interplay between autophagy and mTOR pathways. Materials and Methods Fifty male Wistar albino rats were divided (10/group) into control, DN group, insulin-treated DN group, vitamin D-treated DN group, and combined insulin and vitamin D-treated DN group. Assessments of systolic blood pressure, albuminuria, creatinine clearance, serum glucose, insulin, urea, creatinine, inflammatory cytokines, oxidative stress markers, and rat kidney gene expression of mTOR were performed. Histopathological and immunohistochemical assessments of autophagy marker LC3 in rat kidneys were also performed. Results DN was associated with significant increases in SBP, urinary albumin, serum glucose, urea, creatinine, inflammatory cytokines, MDA, and mTOR gene expression (P < 0.05). However, there was significant decrease in creatinine clearance, serum insulin, GSH, and H score value of LC3 when compared with control group (P < 0.05). The combination of insulin and vitamin D treatment significantly restored DN changes when compared with the other treated groups, except in oxidative stress markers where there was an insignificant difference between the combination-treated and insulin-treated groups (P > 0.05). Conclusion It has been concluded that vitamin D is a potent adjuvant therapy in treatment of DN via downregulation of mTOR gene expression, stimulation of autophagy, and antioxidant, anti-inflammatory, and hypotensive effects.
Collapse
|
38
|
Klen J, Goričar K, Horvat S, Stojan J, Dolžan V. DEPTOR polymorphisms influence late complications in Type 2 diabetes patients. Pharmacogenomics 2020; 20:879-890. [PMID: 31453770 DOI: 10.2217/pgs-2019-0058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: We investigated if DEPTOR polymorphisms influence metabolic parameters and risk for vascular complications in Type 2 diabetes (T2D) patients. Methods: T2D patients were genotyped for DEPTOR rs7840156, rs2271900 and rs4871827. We built low homology model of DEPTOR to check the position of two investigated substitutions within the protein 3D structure. Results: Carriers of polymorphic DEPTOR rs4871827 A allele had higher HDL cholesterol than noncarriers (p = 0.008). Risk for macrovascular and microvascular complications was increased in rs4871827 GG normal genotype carriers (p = 0.006 and p = 0.021, respectively). Low homology model of DEPTOR has shown that p.Ser389Asn substitution resulting from rs4871827 polymorphism is located at the interaction surface with mTOR. Conclusion: Our data suggest role of DEPTOR polymorphism in T2D vascular complication. First draft submitted: xxx; Accepted for publication: xxx; Published online: TBC.
Collapse
Affiliation(s)
- Jasna Klen
- At time of writing: General Hospital Trbovlje, Rudarska cesta 9, 1420 Trbovlje, Slovenia.,At time of publication: Division of Surgery, Department of Abdominal Surgery, University Medical Centre Ljubljana, Zaloška c. 2, 1000 Ljubljana, Slovenia
| | - Katja Goričar
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Simon Horvat
- Animal Science Department, Biotechnical Faculty, University of Ljubljana, 1230 Domžale, Slovenia
| | - Jure Stojan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Vita Dolžan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
39
|
Cao H, Luo J, Zhang Y, Mao X, Wen P, Ding H, Xu J, Sun Q, He W, Dai C, Zen K, Zhou Y, Yang J, Jiang L. Tuberous sclerosis 1 (Tsc1) mediated mTORC1 activation promotes glycolysis in tubular epithelial cells in kidney fibrosis. Kidney Int 2020; 98:686-698. [PMID: 32739207 DOI: 10.1016/j.kint.2020.03.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 03/12/2020] [Accepted: 03/19/2020] [Indexed: 10/24/2022]
Abstract
Energy reprogramming to glycolysis is closely associated with the development of chronic kidney disease. As an important negative regulatory factor of the mammalian target of rapamycin complex 1 (mTORC1) signal, tuberous sclerosis complex 1 (Tsc1) is also a key regulatory point of glycolysis. Here, we investigated whether Tsc1 could mediate the progression of kidney interstitial fibrosis by regulating glycolysis in proximal tubular epithelial cells. We induced mTORC1 signal activation in tubular epithelial cells in kidneys with fibrosis via unilateral ureteral occlusion. This resulted in increased tubular epithelial cell proliferation and glycolytic enzyme upregulation. Prior incubation with rapamycin inhibited mTORC1 activation and abolished the enhanced glycolysis and tubular epithelial cell proliferation. Furthermore, knockdown of Tsc1 expression promoted glycolysis in the rat kidney epithelial cell line NRK-52E. Specific deletion of Tsc1 in the proximal tubules of mice resulted in enlarged kidneys characterized by a high proportion of proliferative tubular epithelial cells, dilated tubules with cyst formation, and a large area of interstitial fibrosis in conjunction with elevated glycolysis. Treatment of the mice with the glycolysis inhibitor 2-deoxyglucose notably ameliorated tubular epithelial cell proliferation, cystogenesis, and kidney fibrosis. Thus, our findings suggest that Tsc1-associated mTORC1 signaling mediates the progression of kidney interstitial fibrosis by regulating glycolysis in proximal tubular epithelial cells.
Collapse
Affiliation(s)
- Hongdi Cao
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Luo
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Zhang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoming Mao
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ping Wen
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Ding
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Xu
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qi Sun
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weichun He
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunsun Dai
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Nanjing, Jiangsu, China
| | - Yang Zhou
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Junwei Yang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Lei Jiang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
40
|
Wang J, Chai L, Lu Y, Lu H, Liu Y, Zhang Y. Attenuation of mTOR Signaling Is the Major Response Element in the Rescue Pathway of Chronic Kidney Disease in Rats. Neuroimmunomodulation 2020; 27:9-18. [PMID: 32526762 DOI: 10.1159/000505095] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 11/28/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Modern lifestyle changes and the interlinking of non-communicable diseases result in the development of chronic kidney disease (CKD). While research has focused on attenuating the CKD, the role of mTOR in the progression of CKD is still unclear. OBJECTIVES The current investigation was undertaken to study the role of mTOR-mediated signaling in CKD using Wistar male rats and adenine-induced CKD as an experimental model. METHOD The animals were divided into 3 groups, representing control, CKD, and rapamycin-pretreated rats. At the end of the experimental period, blood biochemical indexes on kidney function and expression levels of fibrotic markers, including TGF-β, PAI-1, α-smooth muscle action, fibronectin, CTGF, and collagen-1, were analyzed. In addition, kidney injury markers such as kim-1, cystatin-C, NAG, and NGAL, indicating a progressive fibrotic response, were also studied. RESULTS The results suggest that mTOR inhibition significantly attenuated the induction of fibrosis, with restored serum creatinine and blood urea nitrogen levels. Intriguingly, the microRNA (miRNA) analysis revealed an increased expression of miR-193-5p, miR-221, miR-212, and miR-183-5p in CKD, while an increased mRNA expression of anti-inflammatory cytokines and reduced level of pS6K with attenuated miRNA was found in rapamycin-treated rats compared to the CKD animals. CONCLUSION Activation of mTOR is the major responsive element with activation of miRNAs as an elementary role in the progression of kidney disease. Hence, targeting mTOR would be a possible strategy of treatment for CKD.
Collapse
Affiliation(s)
- Jing Wang
- Department of Nephrology, Xingtai People's Hospital, Xingtai, China
| | - Lichao Chai
- Department of Nuclear Medicine, Xingtai People's Hospital, Xingtai, China,
| | - Yi Lu
- Department of Endocrinology, Xingtai People's Hospital, Xingtai, China
| | - Hua Lu
- Department of Nephrology, Xingtai People's Hospital, Xingtai, China
| | - Yanling Liu
- Department of Nephrology, Xingtai People's Hospital, Xingtai, China
| | - Yingying Zhang
- Department of Oncology, Affiliated Hospital of Hebei University of Engineering, Handan, China
| |
Collapse
|
41
|
Mroueh FM, Noureldein M, Zeidan YH, Boutary S, Irani SAM, Eid S, Haddad M, Barakat R, Harb F, Costantine J, Kanj R, Sauleau EA, Ouhtit A, Azar ST, Eid AH, Eid AA. Unmasking the interplay between mTOR and Nox4: novel insights into the mechanism connecting diabetes and cancer. FASEB J 2019; 33:14051-14066. [PMID: 31661292 DOI: 10.1096/fj.201900396rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 09/17/2019] [Indexed: 01/03/2025]
Abstract
Cancer was recently annexed to diabetic complications. Furthermore, recent studies suggest that cancer can increase the risk of diabetes. Consequently, diabetes and cancer share many risk factors, but the cellular and molecular pathways correlating diabetes and colon and rectal cancer (CRC) remain far from understood. In this study, we assess the effect of hyperglycemia on cancer cell aggressiveness in human colon epithelial adenocarcinoma cells in vitro and in an experimental animal model of CRC. Our results show that Nox (NADPH oxidase enzyme) 4-induced reactive oxygen species (ROS) production is deregulated in both diabetes and CRC. This is paralleled by inactivation of the AMPK and activation of the mammalian target of rapamycin (mTOR) C1 signaling pathways, resulting in 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) accumulation, induction of DNA damage, and exacerbation of cancer cell aggressiveness, thus contributing to the genomic instability and predisposition to increased tumorigenesis in the diabetic milieu. Pharmacologic activation of AMPK, inhibition of mTORC1, or blockade of Nox4 reduce ROS production, restore the homeostatic signaling of 8-oxoguanine DNA glycosylase/8-oxodG, and lessen the progression of CRC malignancy in a diabetic milieu. Taken together, our results identify the AMPK/mTORC1/Nox4 signaling axis as a molecular switch correlating diabetes and CRC. Modulating this pathway may be a strategic target of therapeutic potential aimed at reversing or slowing the progression of CRC in patients with or without diabetes.-Mroueh, F. M., Noureldein, M., Zeidan, Y. H., Boutary, S., Irani, S. A. M., Eid, S., Haddad, M., Barakat, R., Harb, F., Costantine, J., Kanj, R., Sauleau, E.-A., Ouhtit, A., Azar, S. T., Eid, A. H., Eid, A. A. Unmasking the interplay between mTOR and Nox4: novel insights into the mechanism connecting diabetes and cancer.
Collapse
Affiliation(s)
- Fatima Mohsen Mroueh
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, Beirut, Lebanon
| | - Mohamed Noureldein
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, Beirut, Lebanon
| | - Youssef H Zeidan
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, Beirut, Lebanon
- Department of Radiation Oncology, Faculty of Medicine and Medical Center, Beirut, Lebanon
| | - Suzan Boutary
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, Beirut, Lebanon
| | - Sara Abou Merhi Irani
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, Beirut, Lebanon
| | - Stéphanie Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, Beirut, Lebanon
| | - Mary Haddad
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, Beirut, Lebanon
| | - Rasha Barakat
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, Beirut, Lebanon
| | - Frederic Harb
- Department of Life and Earth Sciences, Faculty of Sciences, Lebanese University, Fanar, Lebanon
| | - Joseph Costantine
- Department of Electrical and Computer Engineering, Maroun Semaan Faculty of Engineering and Architecture, Beirut, Lebanon
| | - Rouwaida Kanj
- Department of Electrical and Computer Engineering, Maroun Semaan Faculty of Engineering and Architecture, Beirut, Lebanon
| | - Erik-Andre Sauleau
- Department of Biostatistics, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7357 ICube, University of Strasbourg, Strasbourg, France
| | - Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Sami T Azar
- Department of Internal Medicine, Faculty of Medicine and Medical Center, Beirut, Lebanon
- American University of Beirut (AUB) Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Assaad A Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, Beirut, Lebanon
- American University of Beirut (AUB) Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
42
|
Blagosklonny MV. Rapamycin for longevity: opinion article. Aging (Albany NY) 2019; 11:8048-8067. [PMID: 31586989 PMCID: PMC6814615 DOI: 10.18632/aging.102355] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/03/2019] [Indexed: 12/31/2022]
Abstract
From the dawn of civilization, humanity has dreamed of immortality. So why didn't the discovery of the anti-aging properties of mTOR inhibitors change the world forever? I will discuss several reasons, including fear of the actual and fictional side effects of rapamycin, everolimus and other clinically-approved drugs, arguing that no real side effects preclude their use as anti-aging drugs today. Furthermore, the alternative to the reversible (and avoidable) side effects of rapamycin/everolimus are the irreversible (and inevitable) effects of aging: cancer, stroke, infarction, blindness and premature death. I will also discuss why it is more dangerous not to use anti-aging drugs than to use them and how rapamycin-based drug combinations have already been implemented for potential life extension in humans. If you read this article from the very beginning to its end, you may realize that the time is now.
Collapse
|
43
|
Yuan X, Wang X, Li Y, Li X, Zhang S, Hao L. Aldosterone promotes renal interstitial fibrosis via the AIF‑1/AKT/mTOR signaling pathway. Mol Med Rep 2019; 20:4033-4044. [PMID: 31545432 PMCID: PMC6797939 DOI: 10.3892/mmr.2019.10680] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/05/2019] [Indexed: 01/25/2023] Open
Abstract
A number of studies have shown that aldosterone serves an important role in promoting renal interstitial fibrosis, although the specific mechanism remains to be elucidated. A previous study revealed that the fibrotic effect of aldosterone was associated with the expression of allograft inflammatory factor 1 (AIF‑1) in RAW264.7 macrophage cells, in a time‑ and concentration‑dependent manner. However, the exact mechanism through which aldosterone promotes renal interstitial fibrosis remains unknown. In the present study, the effects of aldosterone on renal inflammatory cell infiltration, collagen deposition and the expression levels of AIF‑1, phosphatidylinositol 3‑kinase (PI3K), AKT serine/threonine kinase (AKT), mammalian target of rapamycin (mTOR), the oxidative stress factor NADPH oxidase 2 (NOX2) and nuclear transcription factor erythroid‑related factor 2 (Nrf2) were assessed in normal rats, rats treated with aldosterone, rats treated with aldosterone and spironolactone and those treated with spironolactone only (used as the control). The effect of aldosterone on these factors was also investigated in the renal interstitium of unilateral ureteral obstruction (UUO) rats. Additionally, the AIF‑1 gene was overexpressed and knocked down in macrophage RAW264.7 cells, and the effects of aldosterone on PI3K, AKT, mTOR, NOX2 and Nrf2 were subsequently investigated. The results showed that aldosterone promoted inflammatory cell infiltration, collagen deposition and the expression of AIF‑1, PI3K, AKT, mTOR and NOX2, but inhibited the expression of Nrf2. In the UUO rats, aldosterone also promoted renal interstitial inflammatory cell infiltration, collagen deposition and the expression of AIF‑1, NOX2, PI3K, AKT and mTOR, whereas the expression of Nrf2 was downregulated by aldosterone compared with that in the UUO‑only group; the influence of aldosterone was counteracted by spironolactone in the normal and UUO rats. In vitro, aldosterone upregulated the expression levels of AKT, mTOR, NOX2 and Nrf2 in RAW264.7 cells compared with those in untreated cells. Suppressing the expression of AIF‑1 inhibited the effects of aldosterone, whereas the overexpression of AIF‑1 enhanced these effects in RAW264.7 cells. These findings indicated that aldosterone promoted renal interstitial fibrosis by upregulating the expression of AIF‑1 and that the specific mechanism may involve AKT/mTOR and oxidative stress signaling.
Collapse
Affiliation(s)
- Xueying Yuan
- Department of Nephropathy and Hemodialysis, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xingzhi Wang
- Department of Nephropathy and Hemodialysis, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yushu Li
- Department of Nephropathy and Hemodialysis, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xin Li
- Department of Nephropathy and Hemodialysis, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Shuyu Zhang
- Department of Nephropathy and Hemodialysis, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Lirong Hao
- Department of Nephropathy and Hemodialysis, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
44
|
Decreased Expression of Urinary Mammalian Target of Rapamycin mRNA Is Related to Chronic Renal Fibrosis in IgAN. DISEASE MARKERS 2019; 2019:2424751. [PMID: 31485275 PMCID: PMC6710774 DOI: 10.1155/2019/2424751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/10/2019] [Accepted: 07/29/2019] [Indexed: 12/12/2022]
Abstract
Background Renal fibrosis is a common outcome of all pathological types of chronic kidney disease (CKD). However, the noninvasive detection of renal fibrosis remains a challenge. Methods We collected urine samples from 154 biopsy-proven IgA nephropathy (IgAN) patients and 61 healthy controls. The expression of mTOR was measured and the correlation with renal function parameter and pathological indicators. The receiver operating characteristic (ROC) curve for the diagnosis of IgAN and renal fibrosis was calculated. Results The urinary mammalian target of rapamycin (mTOR) expression was decreased in IgAN patients. The expression of mTOR was correlated with serum creatinine, blood urea nitrogen, estimated glomerular filtration rate, 24 h proteinuria, and cystatin C. Further, the urinary mTOR expression was significantly decreased in severe renal fibrosis patients compared with mild or moderate renal fibrosis patients. Urinary mTOR expression was correlated with score of tubulointerstitial fibrosis (TIF) and score of glomerular sclerosis. The ROC curve showed that mTOR can diagnose IgAN at a cut-off value of 0.930 with the sensitivity of 90.2% and specificity of 73.8% and renal fibrosis at a cut-off value of 0.301 with the sensitivity of 71.7% and specificity of 64.8%. Conclusion Urinary mTOR mRNA expression was a potential biomarker for diagnosis of IgAN and renal fibrosis in IgAN patients.
Collapse
|
45
|
Fasting and rapamycin: diabetes versus benevolent glucose intolerance. Cell Death Dis 2019; 10:607. [PMID: 31406105 PMCID: PMC6690951 DOI: 10.1038/s41419-019-1822-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023]
Abstract
Rapamycin (Sirolimus) slows aging, extends life span, and prevents age-related diseases, including diabetic complications such as retinopathy. Puzzlingly, rapamycin can induce insulin sensitivity, but may also induce insulin resistance or glucose intolerance without insulin resistance. This mirrors the effect of fasting and very low calorie diets, which improve insulin sensitivity and reverse type 2 diabetes, but also can cause a form of glucose intolerance known as benevolent pseudo-diabetes. There is no indication that starvation (benevolent) pseudo-diabetes is detrimental. By contrast, it is associated with better health and life extension. In transplant patients, a weak association between rapamycin/everolimus use and hyperglycemia is mostly due to a drug interaction with calcineurin inhibitors. When it occurs in cancer patients, the hyperglycemia is mild and reversible. No hyperglycemic effects of rapamycin/everolimus have been detected in healthy people. For antiaging purposes, rapamycin/everolimus can be administrated intermittently (e.g., once a week) in combination with intermittent carbohydrate restriction, physical exercise, and metformin.
Collapse
|
46
|
Sun HJ, Wu ZY, Cao L, Zhu MY, Liu TT, Guo L, Lin Y, Nie XW, Bian JS. Hydrogen Sulfide: Recent Progression and Perspectives for the Treatment of Diabetic Nephropathy. Molecules 2019; 24:molecules24152857. [PMID: 31390847 PMCID: PMC6696501 DOI: 10.3390/molecules24152857] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/29/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
Diabetic kidney disease develops in approximately 40% of diabetic patients and is a major cause of chronic kidney diseases (CKD) and end stage kidney disease (ESKD) worldwide. Hydrogen sulfide (H2S), the third gasotransmitter after nitric oxide (NO) and carbon monoxide (CO), is synthesized in nearly all organs, including the kidney. Though studies on H2S regulation of renal physiology and pathophysiology are still in its infancy, emerging evidence shows that H2S production by renal cells is reduced under disease states and H2S donors ameliorate kidney injury. Specifically, aberrant H2S level is implicated in various renal pathological conditions including diabetic nephropathy. This review presents the roles of H2S in diabetic renal disease and the underlying mechanisms for the protective effects of H2S against diabetic renal damage. H2S may serve as fundamental strategies to treat diabetic kidney disease. These H2S treatment modalities include precursors for H2S synthesis, H2S donors, and natural plant-derived compounds. Despite accumulating evidence from experimental studies suggests the potential role of the H2S signaling pathway in the treatment of diabetic nephropathy, these results need further clinical translation. Expanding understanding of H2S in the kidney may be vital to translate H2S to be a novel therapy for diabetic renal disease.
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Zhi-Yuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Lei Cao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Meng-Yuan Zhu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Teng-Teng Liu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Lei Guo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Ye Lin
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, China
| | - Xiao-Wei Nie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
- National University of Singapore (Suzhou) Research Institute, Suzhou 215000, China.
| |
Collapse
|
47
|
Abstract
Designed a century ago to treat epilepsy, the ketogenic diet (KD) is also effective against obesity and diabetes. Paradoxically, some studies in rodents have found that the KD seemingly causes diabetes, contradicting solid clinical data in humans. This paradox can be resolved by applying the concept of starvation pseudo-diabetes, which was discovered in starved animals almost two centuries ago, and has also been observed in some rapamycin-treated rodents. Intriguingly, use of the KD and rapamycin is indicated for a similar spectrum of diseases, including Alzheimer's disease and cancer. Even more intriguingly, benevolent (starvation) pseudo-diabetes may counteract type 2 diabetes or its complications.
Collapse
|
48
|
Tai W, Deng S, Wu W, Li Z, Lei W, Wang Y, Vongphouttha C, Zhang T, Dong Z. Rapamycin attenuates the paraquat-induced pulmonary fibrosis through activating Nrf2 pathway. J Cell Physiol 2019; 235:1759-1768. [PMID: 31301076 PMCID: PMC6899830 DOI: 10.1002/jcp.29094] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
Oxidative stress is a key regulator of idiopathic pulmonary fibrosis. Paraquat (PQ)‐induced pulmonary fibrosis seriously endangers people's health. Rapamycin has been reported to alleviate PQ‐induced pulmonary fibrosis, but its underlying mechanism is unclear. The nuclear factor E2‐related factor 2 (Nrf2) plays an important regulatory role in the antioxidant therapy of PQ‐induced pulmonary fibrosis. In this study, we tried to confirm that rapamycin attenuates PQ‐induced pulmonary fibrosis by regulating Nrf2 pathway. In vivo, we proved that rapamycin could inhibit the degree of PQ‐induced oxidant stress as well as enhanced the expression of Nrf2. In vitro, rapamycin decreased the upregulated effects of cell death and apoptosis, fibrosis‐related factors expression and fibroblast‐to‐myofibroblast transformation by PQ treatment. In vivo, rapamycin treatment reduced fibrosis degree and the expression of fibrosis‐related factors in lung tissues of rat treated PQ. Furthermore, we also found that Nrf2 knockdown reduced the inhibitory effect of rapamycin on PQ‐induced pulmonary fibrosis, as well as decreased Nrf2 transfer from the cytoplasm into the nucleus. Our findings demonstrated that the protective effect of rapamycin is associated with the activation of the Nrf2 pathway in pulmonary fibrosis induced by PQ poisoning.
Collapse
Affiliation(s)
- Wenlin Tai
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Shuhao Deng
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenjuan Wu
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - ZhenKun Li
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wen Lei
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yin Wang
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chanthasone Vongphouttha
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Tao Zhang
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhaoxing Dong
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
49
|
Zhang L, Shen ZY, Wang K, Li W, Shi JM, Osoro EK, Ullah N, Zhou Y, Ji SR. C-reactive protein exacerbates epithelial-mesenchymal transition through Wnt/β-catenin and ERK signaling in streptozocin-induced diabetic nephropathy. FASEB J 2019; 33:6551-6563. [PMID: 30794428 DOI: 10.1096/fj.201801865rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Previous studies have reported the pathogenic role of C-reactive protein (CRP) during diabetic kidney disease (DKD) in human CRP transgenic and Crp-/- mice. However, because humans and mice have inverse acute phase expression patterns of CRP and serum amyloid P component, this could lead to the inaccurate evaluation of CRP function with the above-mentioned CRP transgenic mouse. But different from mice, rats have the same acute phase protein expression pattern as human, which might avoid this problem and be a better choice for CRP function studies. To dispel this doubt and accurately define the role of CRP during diabetic nephropathy, we created the first Crp-/- rat model, which we treated with streptozocin to induce DKD for in vivo studies. Moreover, an established cell line (human kidney 2) was used to further investigate the pathologic mechanisms of CRP. We found that CRP promotes epithelial-mesenchymal transition (EMT) through Wnt/β-catenin and ERK1/2 signaling, which are dependent on CRP binding to FcγRII on apoptotic cells. By promoting EMT, CRP was demonstrated to accelerate the development of DKD. We thus present convincing evidence demonstrating CRP as a therapeutic target for DKD treatment.-Zhang, L., Shen, Z.-Y., Wang, K., Li, W., Shi, J.-M., Osoro, E. K., Ullah, N., Zhou, Y., Ji, S.-R. C-reactive protein exacerbates epithelial-mesenchymal transition through Wnt/β-catenin and ERK signaling in streptozocin-induced diabetic nephropathy.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Zhi-Yuan Shen
- Ministry of Education (MOE) Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Ke Wang
- Ministry of Education (MOE) Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Wei Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Jing-Ming Shi
- Department of Anatomy, School of Basic Medical Sciences, Xizang Minzu University, Xianyang, China
| | - Ezra Kombo Osoro
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Naeem Ullah
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhou
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shang-Rong Ji
- Ministry of Education (MOE) Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
50
|
Zhao XC, Livingston MJ, Liang XL, Dong Z. Cell Apoptosis and Autophagy in Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:557-584. [PMID: 31399985 DOI: 10.1007/978-981-13-8871-2_28] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Renal fibrosis is the final common pathway of all chronic kidney diseases progressing to end-stage renal diseases. Autophagy, a highly conserved lysosomal degradation pathway, plays important roles in maintaining cellular homeostasis in all major types of kidney cells including renal tubular cells as well as podocytes, mesangial cells and endothelial cells in glomeruli. Autophagy dysfunction is implicated in the pathogenesis of various renal pathologies. Here, we analyze the pathological role and regulation of autophagy in renal fibrosis and related kidney diseases in both glomeruli and tubulointerstitial compartments. Further research is expected to gain significant mechanistic insights and discover pathway-specific and kidney-selective therapies targeting autophagy to prevent renal fibrosis and related kidney diseases.
Collapse
Affiliation(s)
- Xing-Chen Zhao
- Division of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Man J Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Xin-Ling Liang
- Division of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
| |
Collapse
|