1
|
Andonian BJ, Hippensteel JA, Abuabara K, Boyle EM, Colbert JF, Devinney MJ, Faye AS, Kochar B, Lee J, Litke R, Nair D, Sattui SE, Sheshadri A, Sherman AN, Singh N, Zhang Y, LaHue SC. Inflammation and aging-related disease: A transdisciplinary inflammaging framework. GeroScience 2024:10.1007/s11357-024-01364-0. [PMID: 39352664 DOI: 10.1007/s11357-024-01364-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Inflammaging, a state of chronic, progressive low-grade inflammation during aging, is associated with several adverse clinical outcomes, including frailty, disability, and death. Chronic inflammation is a hallmark of aging and is linked to the pathogenesis of many aging-related diseases. Anti-inflammatory therapies are also increasingly being studied as potential anti-aging treatments, and clinical trials have shown benefits in selected aging-related diseases. Despite promising advances, significant gaps remain in defining, measuring, treating, and integrating inflammaging into clinical geroscience research. The Clin-STAR Inflammation Research Interest Group was formed by a group of transdisciplinary clinician-scientists with the goal of advancing inflammaging-related clinical research and improving patient-centered care for older adults. Here, we integrate insights from nine medical subspecialties to illustrate the widespread impact of inflammaging on diseases linked to aging, highlighting the extensive opportunities for targeted interventions. We then propose a transdisciplinary approach to enhance understanding and treatment of inflammaging that aims to improve comprehensive care for our aging patients.
Collapse
Affiliation(s)
- Brian J Andonian
- Division of Rheumatology and Immunology, Duke University School of Medicine, Durham, NC, USA.
| | - Joseph A Hippensteel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katrina Abuabara
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Eileen M Boyle
- Department of Haematology, University College London Cancer Institute, London, UK
| | - James F Colbert
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael J Devinney
- Division of Critical Care, Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Adam S Faye
- Division of Gastroenterology, Department of Population Health, NYU Langone Medical Center, New York, NY, USA
| | - Bharati Kochar
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Jiha Lee
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rachel Litke
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Devika Nair
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sebastian E Sattui
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anoop Sheshadri
- Division of Nephrology, Department of Medicine, University of California, San Francisco, Nephrology Section, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | | | - Namrata Singh
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Yinan Zhang
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sara C LaHue
- Department of Neurology, School of Medicine, and the UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
2
|
Gupta S, Mandal S, Banerjee K, Almarshood H, Pushpakumar SB, Sen U. Complex Pathophysiology of Acute Kidney Injury (AKI) in Aging: Epigenetic Regulation, Matrix Remodeling, and the Healing Effects of H 2S. Biomolecules 2024; 14:1165. [PMID: 39334931 PMCID: PMC11429536 DOI: 10.3390/biom14091165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
The kidney is an essential excretory organ that works as a filter of toxins and metabolic by-products of the human body and maintains osmotic pressure throughout life. The kidney undergoes several physiological, morphological, and structural changes with age. As life expectancy in humans increases, cell senescence in renal aging is a growing challenge. Identifying age-related kidney disorders and their cause is one of the contemporary public health challenges. While the structural abnormalities to the extracellular matrix (ECM) occur, in part, due to changes in MMPs, EMMPRIN, and Meprin-A, a variety of epigenetic modifiers, such as DNA methylation, histone alterations, changes in small non-coding RNA, and microRNA (miRNA) expressions are proven to play pivotal roles in renal pathology. An aged kidney is vulnerable to acute injury due to ischemia-reperfusion, toxic medications, altered matrix proteins, systemic hemodynamics, etc., non-coding RNA and miRNAs play an important role in renal homeostasis, and alterations of their expressions can be considered as a good marker for AKI. Other epigenetic changes, such as histone modifications and DNA methylation, are also evident in AKI pathophysiology. The endogenous production of gaseous molecule hydrogen sulfide (H2S) was documented in the early 1980s, but its ameliorative effects, especially on kidney injury, still need further research to understand its molecular mode of action in detail. H2S donors heal fibrotic kidney tissues, attenuate oxidative stress, apoptosis, inflammation, and GFR, and also modulate the renin-angiotensin-aldosterone system (RAAS). In this review, we discuss the complex pathophysiological interplay in AKI and its available treatments along with future perspectives. The basic role of H2S in the kidney has been summarized, and recent references and knowledge gaps are also addressed. Finally, the healing effects of H2S in AKI are described with special emphasis on epigenetic regulation and matrix remodeling.
Collapse
Affiliation(s)
- Shreyasi Gupta
- Department of Zoology, Trivenidevi Bhalotia College, College Para Rd, Raniganj 713347, West Bengal, India
| | - Subhadeep Mandal
- Department of Zoology, Trivenidevi Bhalotia College, College Para Rd, Raniganj 713347, West Bengal, India
| | - Kalyan Banerjee
- Department of Zoology, Trivenidevi Bhalotia College, College Para Rd, Raniganj 713347, West Bengal, India
| | - Hebah Almarshood
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sathnur B Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
3
|
Chen Y, Dai R, Cheng M, Wang W, Liu C, Cao Z, Ge Y, Wang Y, Zhang L. Status and role of the ubiquitin-proteasome system in renal fibrosis. Biomed Pharmacother 2024; 178:117210. [PMID: 39059348 DOI: 10.1016/j.biopha.2024.117210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024] Open
Abstract
The ubiquitin-proteasome system (UPS) is a basic regulatory mechanism in cells that is essential for maintaining cell homeostasis, stimulating signal transduction, and determining cell fate. These biological processes require coordinated signaling cascades across members of the UPS to achieve substrate ubiquitination and deubiquitination. The role of the UPS in fibrotic diseases has attracted widespread attention, and the aberrant expression of UPS members affects the fibrosis process. In this review, we provide an overview of the UPS and its relevance for fibrotic diseases. Moreover, for the first time, we explore in detail how the UPS promotes or inhibits renal fibrosis by regulating biological processes such as signaling pathways, inflammation, oxidative stress, and the cell cycle, emphasizing the status and role of the UPS in renal fibrosis. Further research on this system may reveal new strategies for preventing renal fibrosis.
Collapse
Affiliation(s)
- Yizhen Chen
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Rong Dai
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Meng Cheng
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Weili Wang
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Chuanjiao Liu
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Zeping Cao
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Yong Ge
- First Clinical Medical College, Anhui University of Chinese Medicine, Hefei, China
| | - Yiping Wang
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.
| | - Lei Zhang
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
4
|
Li Y, Tan P, Liu Q, Liu M, Wang Y, Kong W, Sun H, Shao X. MiRNA-133a-3p Attenuates Renal Tubular Epithelial Cell Injury via Targeting MALM1 and Suppressing the Notch Signaling Pathway in Diabetic Nephropathy. Cell Biochem Biophys 2024; 82:2401-2411. [PMID: 38878099 DOI: 10.1007/s12013-024-01351-4] [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] [Accepted: 06/04/2024] [Indexed: 10/02/2024]
Abstract
Diabetic nephropathy (DN) is a serious microvascular complication of diabetes characterized by structural and functional changes of kidneys. Human renal tubular epithelial (HK-2) cells are important for kidney recovery post injury and usually used for establishment of DN cell models. The study explored the role of microRNA (miR)-133a-3p in DN cell model and animal model. A cell model for DN was established via high glucose (HG) stimulation to HK-2 cells. Cell viability and apoptotic rate were measured by cell counting kit 8 and flow cytometry. Polymerase chain reaction was performed to quantify levels of miR-133a-3p and targets. Luciferase reporter assay was conducted to verify the binding of miR-133a-3p and MAML1. After establishment of a mouse model of DN, levels of renal function indicators were measured by biochemical analysis. Hematoxylin-eosin and periodic acid-schiff staining of kidney samples were performed to analyze histological changes. Western blotting was conducted to quantify levels of apoptotic markers, MAML1, and factors related to Notch signaling. Results showed that HG induced HK-2 cell apoptosis and the reduction of cell viability. MiR-133a-3p was lowly expressed in HG-stimulated HK-2 cells. Overexpressed miR-133a-3p improved HK-2 cell injury by increasing cell viability and hampering apoptosis under HG condition. In addition, miR-133a-3p directly targets MAML1 3'-untranslated region. MAML1 overexpression countervailed the repressive impact of miR-133a-3p on cell apoptosis in the context of HG. Moreover, miR-133a-3p inhibited the activity of Notch pathway by downregulating MAML1. MiR-133a-3p inhibits DN progression in mice, as evidenced by reduced fasting blood glucose level, improved levels of renal function parameters, and alleviation of kidney atrophy. In conclusion, miR-133a-3p improves HG-induced HK-2 cell injury and inhibits DN progression by targeting MAML1 and inactivating Notch signaling.
Collapse
Affiliation(s)
- Yuting Li
- Department of Geriatrics, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medcine, Suzhou, 215000, China
| | - Peng Tan
- Department of Nephrology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medcine, Suzhou, 215000, China
| | - Qianpan Liu
- Department of Nephrology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medcine, Suzhou, 215000, China
| | - Man Liu
- Department of Nephrology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medcine, Suzhou, 215000, China
| | - Yue Wang
- Department of Nephrology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medcine, Suzhou, 215000, China
| | - Weixin Kong
- Department of Nephrology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medcine, Suzhou, 215000, China
| | - Huaixin Sun
- Department of Nephrology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medcine, Suzhou, 215000, China
| | - Xiang Shao
- Department of Nephrology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medcine, Suzhou, 215000, China.
- Centralab, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medcine, Suzhou, 215000, China.
| |
Collapse
|
5
|
Almalki WH, Salman Almujri S. Oxidative stress and senescence in aging kidneys: the protective role of SIRT1. EXCLI JOURNAL 2024; 23:1030-1067. [PMID: 39391060 PMCID: PMC11464868 DOI: 10.17179/excli2024-7519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/07/2024] [Indexed: 10/12/2024]
Abstract
Aging leads to a gradual decline in kidney function, making the kidneys increasingly vulnerable to various diseases. Oxidative stress, together with cellular senescence, has been established as paramount in promoting the aging process of the kidney. Oxidative stress, defined as an imbalance between ROS formation and antioxidant defense mechanisms, has been implicated in the kidney's cellular injury, inflammation, and premature senescence. Concurrently, the accumulation of SCs in the kidney also exacerbates oxidative stress via the secretion of pro-inflammatory and tissue-damaging factors as the senescence-associated secretory phenotype (SASP). Recently, SIRT1, a nicotinamide adenine dinucleotide (NAD)-dependent deacetylase, has been pivotal in combating oxidative stress and cellular senescence in the aging kidney. SIRT1 acts as a potential antioxidant molecule through myriad pathways that influence diverse transcription factors and enzymes essential in maintaining redox homeostasis. SIRT1 promotes longevity and renal health by modulating the acetylation of cell cycle and senescence pathways. This review covers the complex relationship between oxidative stress and cellular senescence in the aging kidney, emphasizing the protective role of SIRT1. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Aseer, Saudi Arabia
| |
Collapse
|
6
|
Wu H, Qiu Z, Wang L, Li W. Renal Fibrosis: SIRT1 Still of Value. Biomedicines 2024; 12:1942. [PMID: 39335456 PMCID: PMC11428497 DOI: 10.3390/biomedicines12091942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Chronic kidney disease (CKD) is a major global health concern. Renal fibrosis, a prevalent outcome regardless of the initial cause, ultimately leads to end-stage renal disease. Glomerulosclerosis and renal interstitial fibrosis are the primary pathological features. Preventing and slowing renal fibrosis are considered effective strategies for delaying CKD progression. However, effective treatments are lacking. Sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase belonging to class III histone deacetylases, is implicated in the physiological regulation and protection of the kidney and is susceptible to a diverse array of pathological influences, as demonstrated in previous studies. Interestingly, controversial conclusions have emerged as research has progressed. This review provides a comprehensive summary of the current understanding and advancements in the field; specifically, the biological roles and mechanisms of SIRT1 in regulating renal fibrosis progression. These include aspects such as lipid metabolism, epithelial-mesenchymal transition, oxidative stress, aging, inflammation, and autophagy. This manuscript explores the potential of SIRT1 as a therapeutic target for renal fibrosis and offers new perspectives on treatment approaches and prognostic assessments.
Collapse
Affiliation(s)
- Huailiang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.W.); (Z.Q.)
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.W.); (Z.Q.)
| | - Liyan Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.W.); (Z.Q.)
| |
Collapse
|
7
|
Li J, Zou Y, Kantapan J, Su H, Wang L, Dechsupa N. TGF‑β/Smad signaling in chronic kidney disease: Exploring post‑translational regulatory perspectives (Review). Mol Med Rep 2024; 30:143. [PMID: 38904198 PMCID: PMC11208996 DOI: 10.3892/mmr.2024.13267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Abstract
The TGF‑β/Smad signaling pathway plays a pivotal role in the onset of glomerular and tubulointerstitial fibrosis in chronic kidney disease (CKD). The present review delves into the intricate post‑translational modulation of this pathway and its implications in CKD. Specifically, the impact of the TGF‑β/Smad pathway on various biological processes was investigated, encompassing not only renal tubular epithelial cell apoptosis, inflammation, myofibroblast activation and cellular aging, but also its role in autophagy. Various post‑translational modifications (PTMs), including phosphorylation and ubiquitination, play a crucial role in modulating the intensity and persistence of the TGF‑β/Smad signaling pathway. They also dictate the functionality, stability and interactions of the TGF‑β/Smad components. The present review sheds light on recent findings regarding the impact of PTMs on TGF‑β receptors and Smads within the CKD landscape. In summary, a deeper insight into the post‑translational intricacies of TGF‑β/Smad signaling offers avenues for innovative therapeutic interventions to mitigate CKD progression. Ongoing research in this domain holds the potential to unveil powerful antifibrotic treatments, aiming to preserve renal integrity and function in patients with CKD.
Collapse
Affiliation(s)
- Jianchun Li
- Department of Radiologic Technology, Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yuanxia Zou
- Department of Radiologic Technology, Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jiraporn Kantapan
- Department of Radiologic Technology, Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Hongwei Su
- Department of Urology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Nathupakorn Dechsupa
- Department of Radiologic Technology, Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
8
|
Yin Q, Tang TT, Lu XY, Ni WJ, Yin D, Zhang YL, Jiang W, Zhang Y, Li ZL, Wen Y, Gan WH, Zhang AQ, Lv LL, Wang B, Liu BC. Macrophage-derived exosomes promote telomere fragility and senescence in tubular epithelial cells by delivering miR-155. Cell Commun Signal 2024; 22:357. [PMID: 38987851 PMCID: PMC11238407 DOI: 10.1186/s12964-024-01708-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/08/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is highly prevalent worldwide, and its global burden is substantial and growing. CKD displays a number of features of accelerated senescence. Tubular cell senescence is a common biological process that contributes to CKD progression. Tubulointerstitial inflammation is a driver of tubular cell senescence and a common characteristic of CKD. However, the mechanism by which the interstitial inflammation drives tubular cell senescence remains unclear. This paper aims to explore the role of exosomal miRNAs derived from macrophages in the development of tubular cell senescence. METHODS Among the identified inflammation-related miRNAs, miR-155 is considered to be one of the most important miRNAs involved in the inflammatory response. Macrophages, the primary immune cells that mediate inflammatory processes, contain a high abundance of miR-155 in their released exosomes. We assessed the potential role of miR-155 in tubular cell senescence and renal fibrosis. We subjected miR-155-/- mice and wild-type controls, as well as tubular epithelial cells (TECs), to angiotensin II (AngII)-induced kidney injury. We assessed kidney function and injury using standard techniques. TECs were evaluated for cell senescence and telomere dysfunction in vivo and in vitro. Telomeres were measured by the fluorescence in situ hybridization. RESULTS Compared with normal controls, miR-155 was up-regulated in proximal renal tubule cells in CKD patients and mouse models of CKD. Moreover, the expression of miR-155 was positively correlated with the extent of renal fibrosis, eGFR decline and p16INK4A expression. The overexpression of miR-155 exacerbated tubular senescence, evidenced by increased detection of p16INK4A/p21expression and senescence-associated β-galactosidase activity. Notably, miR-155 knockout attenuates renal fibrosis and tubule cell senescence in vivo. Interestingly, once released, macrophages-derived exosomal miR-155 was internalized by TECs, leading to telomere shortening and dysfunction through targeting TRF1. A dual-luciferase reporter assay confirmed that TRF1 was the direct target of miR-155. Thus, our study clearly demonstrates that exosomal miR-155 may mediate communication between macrophages and TECs, subsequently inducing telomere dysfunction and senescence in TECs. CONCLUSIONS Our work suggests a new mechanism by which macrophage exosomes are involved in the development of tubule senescence and renal fibrosis, in part by delivering miR-155 to target TRF1 to promote telomere dysfunction. Our study may provide novel strategies for the treatment of AngII-induced kidney injury.
Collapse
Affiliation(s)
- Qing Yin
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China
| | - Xiao-Yu Lu
- Department of Pediatric Nephrology, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu, China
| | - Wei-Jie Ni
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China
| | - Di Yin
- Department of Nephrology, Taixing People's Hospital, Taixing, Jiangsu, China
| | - Yi-Lin Zhang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China
| | - Wei Jiang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China
| | - Yue Zhang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China
| | - Yi Wen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China
| | - Wei-Hua Gan
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ai-Qing Zhang
- Department of Pediatric Nephrology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China
| | - Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China.
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Nanjing, Jiangsu, China.
| |
Collapse
|
9
|
Berezin AE, Berezina TA, Hoppe UC, Lichtenauer M, Berezin AA. An overview of circulating and urinary biomarkers capable of predicting the transition of acute kidney injury to chronic kidney disease. Expert Rev Mol Diagn 2024; 24:627-647. [PMID: 39007888 DOI: 10.1080/14737159.2024.2379355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Acute kidney injury (AKI) defined by a substantial decrease in kidney function within hours to days and is often irreversible with higher risk to chronic kidney disease (CKD) transition. AREAS COVERED The authors discuss the diagnostic and predictive utilities of serum and urinary biomarkers on AKI and on the risk of AKI-to-CKD progression. The authors focus on the relevant literature covering evidence of circulating and urinary biomarkers' capability to predict the transition of AKI to CKD. EXPERT OPINION Based on the different modalities of serum and urinary biomarkers, multiple biomarker panel seems to be potentially useful to distinguish between various types of AKI, to detect the severity and the risk of AKI progression, to predict the clinical outcome and evaluate response to the therapy. Serum/urinary neutrophil gelatinase-associated lipocalin (NGAL), serum/urinary uromodulin, serum extracellular high mobility group box-1 (HMGB-1), serum cystatin C and urinary liver-type fatty acid-binding protein (L-FABP) were the most effective in the prediction of AKI-to-CKD transition regardless of etiology and the presence of critical state in patients. The current clinical evidence on the risk assessments of AKI progression is mainly based on the utility of combination of functional, injury and stress biomarkers, mainly NGAL, L-FABP, HMGB-1 and cystatin C.
Collapse
Affiliation(s)
- Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Tetiana A Berezina
- Department of Internal Medicine & Nephrology, VitaCenter, Zaporozhye, Ukraine
| | - Uta C Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | | |
Collapse
|
10
|
Wang B, Yu W, Zhang W, Zhang M, Niu Y, Jin X, Zhang J, Sun D, Li H, Zhang Z, Luo Q, Cheng X, Niu J, Cai G, Chen X, Chen Y. Enhanced TRPC3 transcription through AT1R/PKA/CREB signaling contributes to mitochondrial dysfunction in renal tubular epithelial cells in D-galactose-induced accelerated aging mice. Aging Cell 2024; 23:e14130. [PMID: 38415902 PMCID: PMC11166371 DOI: 10.1111/acel.14130] [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: 04/10/2023] [Revised: 02/05/2024] [Accepted: 02/11/2024] [Indexed: 02/29/2024] Open
Abstract
Aging-associated renal dysfunction promotes the pathogenesis of chronic kidney disease. Mitochondrial dysfunction in renal tubular epithelial cells is a hallmark of senescence and leads to accelerated progression of renal disorders. Dysregulated calcium profiles in mitochondria contribute to aging-associated disorders, but the detailed mechanism of this process is not clear. In this study, modulation of the sirtuin 1/angiotensin II type 1 receptor (Sirt1/AT1R) pathway partially attenuated renal glomerular sclerosis, tubular atrophy, and interstitial fibrosis in D-galactose (D-gal)-induced accelerated aging mice. Moreover, modulation of the Sirt1/AT1R pathway improved mitochondrial dysfunction induced by D-gal treatment. Transient receptor potential channel, subtype C, member 3 (TRPC3) upregulation mediated dysregulated cellular and mitochondrial calcium homeostasis during aging. Furthermore, knockdown or knockout (KO) of Trpc3 in mice ameliorated D-gal-induced mitochondrial reactive oxygen species production, membrane potential deterioration, and energy metabolism disorder. Mechanistically, activation of the AT1R/PKA pathway promoted CREB phosphorylation and nucleation of CRE2 binding to the Trpc3 promoter (-1659 to -1648 bp) to enhance transcription. Trpc3 KO significantly improved the renal disorder and cell senescence in D-gal-induced mice. Taken together, these results indicate that TRPC3 upregulation mediates age-related renal disorder and is associated with mitochondrial calcium overload and dysfunction. TRPC3 is a promising therapeutic target for aging-associated renal disorders.
Collapse
Affiliation(s)
- Bin Wang
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
- Senior Department of Nephrology, The First Medical Center of Chinese PLA General HospitalChinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases ResearchBeijingChina
| | - Wenpei Yu
- Department of Chemical Defense Medicine, School of Military Preventive MedicineArmy Medical University (Third Military Medical University)ChongqingChina
- Department of Clinical MedicineDazhou Vocational and Technical CollegeDazhouSichuanChina
| | - Weiguang Zhang
- Senior Department of Nephrology, The First Medical Center of Chinese PLA General HospitalChinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases ResearchBeijingChina
| | - Min Zhang
- Senior Department of Nephrology, The First Medical Center of Chinese PLA General HospitalChinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases ResearchBeijingChina
| | - Yue Niu
- Senior Department of Nephrology, The First Medical Center of Chinese PLA General HospitalChinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases ResearchBeijingChina
| | - Xinye Jin
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
| | - Jie Zhang
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
| | - Ding Sun
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
- Graduate SchoolChinese PLA General HospitalBeijingChina
| | - Hao Li
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
- Graduate SchoolChinese PLA General HospitalBeijingChina
| | - Zehao Zhang
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
- Graduate SchoolChinese PLA General HospitalBeijingChina
| | - Qing Luo
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
| | - Xiaowei Cheng
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
| | - Jingxue Niu
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
| | - Guangyan Cai
- Senior Department of Nephrology, The First Medical Center of Chinese PLA General HospitalChinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases ResearchBeijingChina
| | - Xiangmei Chen
- Senior Department of Nephrology, The First Medical Center of Chinese PLA General HospitalChinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases ResearchBeijingChina
| | - Yizhi Chen
- Department of Nephrology, The Hainan Academician Team Innovation CenterHainan Hospital of Chinese PLA General HospitalSanyaChina
- Senior Department of Nephrology, The First Medical Center of Chinese PLA General HospitalChinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases ResearchBeijingChina
- Graduate SchoolChinese PLA General HospitalBeijingChina
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
11
|
Bao YN, Yang Q, Shen XL, Yu WK, Zhou L, Zhu QR, Shan QY, Wang ZC, Cao G. Targeting tumor suppressor p53 for organ fibrosis therapy. Cell Death Dis 2024; 15:336. [PMID: 38744865 PMCID: PMC11094089 DOI: 10.1038/s41419-024-06702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Fibrosis is a reparative and progressive process characterized by abnormal extracellular matrix deposition, contributing to organ dysfunction in chronic diseases. The tumor suppressor p53 (p53), known for its regulatory roles in cell proliferation, apoptosis, aging, and metabolism across diverse tissues, appears to play a pivotal role in aggravating biological processes such as epithelial-mesenchymal transition (EMT), cell apoptosis, and cell senescence. These processes are closely intertwined with the pathogenesis of fibrotic disease. In this review, we briefly introduce the background and specific mechanism of p53, investigate the pathogenesis of fibrosis, and further discuss p53's relationship and role in fibrosis affecting the kidney, liver, lung, and heart. In summary, targeting p53 represents a promising and innovative therapeutic approach for the prevention and treatment of organ fibrosis.
Collapse
Affiliation(s)
- Yi-Ni Bao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Xin-Lei Shen
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Wen-Kai Yu
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Li Zhou
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Qing-Ru Zhu
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Qi-Yuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Zhi-Chao Wang
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
12
|
Deng D, Tian D, Wang Y, Bai Y, Diao Z, Liu W. Secreted frizzled-related protein 5 protects against renal fibrosis by inhibiting Wnt/β-catenin pathway. Open Med (Wars) 2024; 19:20240934. [PMID: 38584843 PMCID: PMC10997006 DOI: 10.1515/med-2024-0934] [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: 11/19/2023] [Revised: 01/28/2024] [Accepted: 02/22/2024] [Indexed: 04/09/2024] Open
Abstract
Renal fibrosis (RF) is an important pathogenesis for renal function deterioration in chronic kidney disease. Secreted frizzled-related protein 5 (SFRP5) is an anti-fibrotic adipokine but its direct role on RF remains unknown. It was aimed to study the protective effect of SFRP5 against RF and interference with Wnt/β-catenin signaling pathway for the first time. First, the therapeutic efficacy of SFRP5 was evaluated by adenovirus overexpression in rats with unilateral ureteral obstruction (UUO) in vivo. Thirty-six rats were randomly divided into the sham, UUO, and SFRP5 (UUO + Ad-SFRP5) groups. Half rats in each group were selected at random for euthanasia at 7 days and the others until 14 days. Then, the transforming growth factor (TGF)-β1-induced epithelial-mesenchymal transition (EMT) was established in HK-2 cells in vitro. The cells were divided into four groups: the control group, the TGF-β1 group, the TGF-β1 + SFRP5 group, and the TGF-β1 + SFRP5 + anti-SFRP5 group. The makers of EMT and Wnt/β-catenin pathway proteins were investigated. In the UUO model, expression of SFRP5 showed compensatory upregulation, and adenoviral-mediated SFRP5 over-expression remarkably attenuated RF, as demonstrated by maintenance of E-cadherin and suppression of α-smooth muscle actin (SMA). In vitro, SFRP5 was shown to inhibit TGF-β1-mediated positive regulation of α-SMA, fibronectin, collagen I but negative regulation of E-cadherin. Furthermore, SFRP5 abrogated activation of Wnt/β-catenin, which was the essential pathway in EMT and RF pathogenesis. The changes after a neutralizing antibody to SFRP5 confirmed the specificity of SFRP5 for inhibition. These findings suggest that SFRP5 can directly ameliorate EMT and protect against RF by inhibiting Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Dai Deng
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, 100050, Beijing, China
| | - Dongli Tian
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, 100050, Beijing, China
| | - Yahui Wang
- Department of Emergency, China Rehabilitation Research Center, Beijing, China
| | - Yu Bai
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, 100050, Beijing, China
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, 100050, Beijing, China
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, 95 Yong’an Road, Xicheng District, 100050, Beijing, China
| |
Collapse
|
13
|
Tao P, Liu H, Hou G, Lu J, Xu Y. Kangxianling formula attenuates renal fibrosis by regulating gut microbiota. Eur J Med Res 2024; 29:183. [PMID: 38500195 PMCID: PMC10949625 DOI: 10.1186/s40001-024-01778-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 03/09/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Renal fibrosis (RF) produced adverse effect on kidney function. Recently, intestinal dysbiosis is a key regulator that promotes the formation of renal fibrosis. This study will focus on exploring the protective mechanism of Kangxianling Formula (KXL) on renal fibrosis from the perspective of intestinal flora. METHODS Unilateral Ureteral Obstruction (UUO) was used to construct rats' model with RF, and receive KXL formula intervention for 1 week. The renal function indicators were measured. Hematoxylin-eosin (HE), Masson and Sirus red staining were employed to detect the pathological changes of renal tissue in each group. The expression of α-SMA, Col-III, TGF-β, FN, ZO-1, and Occuludin was detected by immunofluorescence and immunohistochemistry. Rat feces samples were collected and analyzed for species' diversity using high-throughput sequencing 16S rRNA. RESULTS Rats in UUO groups displayed poor renal function as well as severe RF. The pro-fibrotic protein expression in renal tissues including α-SMA, Col-III, TGF-β and FN was increased in UUO rats, while ZO-1 and Occuludin -1 expression was downregulated in colon tissues. The above changes were attenuated by KXL treatment. 16S rRNA sequencing results revealed that compared with the sham group, the increased abundance of pathogenic bacteria including Acinetobacter, Enterobacter and Proteobacteria and the decreased abundance of beneficial bacteria including Actinobacteriota, Bifidobacteriales, Prevotellaceae, and Lactobacillus were found in UUO group. After the administration of KXL, the growth of potential pathogenic bacteria was reduced and the abundance of beneficial bacteria was enhanced. CONCLUSION KXL displays a therapeutical potential in protecting renal function and inhibiting RF, and its mechanism of action may be associated with regulating intestinal microbiota.
Collapse
Affiliation(s)
- Pengyu Tao
- Department of Nephrology, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haiyan Liu
- Department of Ultrasound, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Guangjian Hou
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jianrao Lu
- Department of Nephrology, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yukun Xu
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
14
|
Wu Q, Chen Q, Xu D, Wang X, Ye H, Li X, Xiong Y, Li J, Zhou S, Miao J, Shen W, Liu Y, Niu H, Tang Y, Zhou L. C-X-C chemokine receptor type 4 promotes tubular cell senescence and renal fibrosis through β-catenin-inhibited fatty acid oxidation. J Cell Mol Med 2024; 28:e18075. [PMID: 38213100 PMCID: PMC10844696 DOI: 10.1111/jcmm.18075] [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: 03/31/2023] [Revised: 11/01/2023] [Accepted: 11/24/2023] [Indexed: 01/13/2024] Open
Abstract
The prevalence of chronic kidney disease (CKD) is highly increasing. Renal fibrosis is a common pathological feature in various CKD. Previous studies showed tubular cell senescence is highly involved in the pathogenesis of renal fibrosis. However, the inducers of tubular senescence and the underlying mechanisms have not been fully investigated. C-X-C motif chemokine receptor 4 (CXCR4), a G-protein-coupled seven-span transmembrane receptor, increases renal fibrosis and plays an important role in tubular cell injury. Whereas, whether CXCR4 could induce tubular cell senescence and the detailed mechanisms have not studied yet. In this study, we adopted adriamycin nephropathy and 5/6 nephrectomy models, and cultured tubular cell line. Overexpression or knockdown of CXCR4 was obtained by injection of related plasmids. We identified CXCR4 increased in injury tubular cells. CXCR4 was expressed predominantly in renal tubular epithelial cells and co-localized with adipose differentiation-related protein (ADRP) as well as the senescence-related protein P16INK4A . Furthermore, we found overexpression of CXCR4 greatly induced the activation of β-catenin, while knockdown of CXCR4 inhibited it. We also found that CXCR4 inhibited fatty acid oxidation and triggered lipid deposition in tubular cells. To inhibit β-catenin by ICG-001, an inhibitor of β-catenin, could significantly block CXCR4-suppressed fatty acid oxidation. Taken together, our results indicate that CXCR4 is a key mediator in tubular cell senescence and renal fibrosis. CXCR4 promotes tubular cell senescence and renal fibrosis by inducing β-catenin and inhibiting fatty acid metabolism. Our findings provide a new theory for tubular cell injury in renal fibrosis.
Collapse
Affiliation(s)
- Qinyu Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of NephrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Qiurong Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Dan Xu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaoxu Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Huiyun Ye
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaolong Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yabing Xiong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jiemei Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Weiwei Shen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Hongxin Niu
- Special Medical Service Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ying Tang
- Department of NephrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
15
|
Kim Y, Kang D, Choi GE, Kim SD, Yang SJ, Kim H, You D, Kim CS, Suh N. Therapeutic potential of BMSC-conditioned medium in an in vitro model of renal fibrosis using the RPTEC/TERT1 cell line. BMB Rep 2024; 57:116-121. [PMID: 38303564 PMCID: PMC10910087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
We investigated the therapeutic potential of bone marrow-derived mesenchymal stem cell-conditioned medium (BMSC-CM) on immortalized renal proximal tubule epithelial cells (RPTEC/ TERT1) in a fibrotic environment. To replicate the increased stiffness characteristic of kidneys in chronic kidney disease, we utilized polyacrylamide gel platforms. A stiff matrix was shown to increase α-smooth muscle actin (α-SMA) levels, indicating fibrogenic activation in RPTEC/TERT1 cells. Interestingly, treatment with BMSC-CM resulted in significant reductions in the levels of fibrotic markers (α-SMA and vimentin) and increases in the levels of the epithelial marker E-cadherin and aquaporin 7, particularly under stiff conditions. Furthermore, BMSC-CM modified microRNA (miRNA) expression and reduced oxidative stress levels in these cells. Our findings suggest that BMSC-CM can modulate cellular morphology, miRNA expression, and oxidative stress in RPTEC/TERT1 cells, highlighting its therapeutic potential in fibrotic kidney disease. [BMB Reports 2024; 57(2): 116-121].
Collapse
Affiliation(s)
- Yunji Kim
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
| | - Dayeon Kang
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soonchunhyang University, Asan 31538, Korea
| | - Ga-eun Choi
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
| | - Sang Dae Kim
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
| | | | - Hyosang Kim
- Division of Nephrology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Dalsan You
- Department of Urology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Choung Soo Kim
- Urology Institute, Ewha Womans University Mokdong Hospital, Seoul 07985, Korea
| | - Nayoung Suh
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soonchunhyang University, Asan 31538, Korea
| |
Collapse
|
16
|
Zhang X, Li L, Tan H, Hong X, Yuan Q, Hou FF, Zhou L, Liu Y. Klotho-derived peptide 1 inhibits cellular senescence in the fibrotic kidney by restoring Klotho expression via posttranscriptional regulation. Theranostics 2024; 14:420-435. [PMID: 38164143 PMCID: PMC10750200 DOI: 10.7150/thno.89105] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 11/16/2023] [Indexed: 01/03/2024] Open
Abstract
Background: Klotho deficiency is a common feature of premature aging and chronic kidney disease (CKD). As such, restoring Klotho expression could be a logic strategy for protecting against various nephropathies. In this study, we demonstrate that KP1, a Klotho-derived peptide, inhibits cellular senescence by restoring endogenous Klotho expression. Methods: The effects of KP1 on cellular senescence and Klotho expression were assessed in mouse models of CKD. RNA-sequencing was employed to identify the microRNA involved in regulating Klotho by KP1. Gain- or loss-of-function approaches were used to assess the role of miR-223-3p and IncRNA-TUG1 in regulating Klotho and cellular senescence. Results: KP1 inhibited senescence markers p21, p16 and γ-H2AX in tubular epithelial cells of diseased kidneys, which was associated with its restoration of Klotho expression at the posttranscriptional level. Profiling of kidney microRNAs by RNA sequencing identified miR-223-3p that bound to Klotho mRNA and inhibited its protein expression. Overexpression of miR-223-3p inhibited Klotho and induced p21, p16 and γ-H2AX, which were negated by KP1. Conversely, inhibition of miR-223-3p restored Klotho expression, inhibited cellular senescence. Furthermore, miR-223-3p interacted with lncRNA-TUG1 and inhibited its expression. Knockdown of lncRNA-TUG1 increased miR-223-3p, aggravated Klotho loss and worsened cellular senescence, whereas KP1 mitigated all these changes. Conclusion: These studies demonstrate that KP1 inhibits cellular senescence and induces Klotho expression via posttranscriptional regulation mediated by miR-223-3p and lncRNA-TUG1. By restoring endogenous Klotho, KP1 elicits a broad spectrum of protective actions and could serve as a promising therapeutic agent for fibrotic kidney disorders.
Collapse
Affiliation(s)
- Xiaoyao Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huishi Tan
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue Hong
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Yuan
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Lili Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
17
|
Shu H, Zhang Z, Liu J, Chen P, Yang C, Wu Y, Wu D, Cao Y, Chu Y, Li L. Circular RNAs: An emerging precise weapon for diabetic nephropathy diagnosis and therapy. Biomed Pharmacother 2023; 168:115818. [PMID: 37939612 DOI: 10.1016/j.biopha.2023.115818] [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: 09/03/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023] Open
Abstract
Diabetic nephropathy (DN) is a prevalent chronic microvascular complication associated with diabetes mellitus and represents a major cause of chronic kidney disease and renal failure. Current treatment strategies for DN primarily focus on symptom alleviation, lacking effective approaches to halt or reverse DN progression. Circular RNA (circRNA), characterized by a closed-loop structure, has emerged as a novel non-coding RNA regulator of gene expression, attributed to its conservation, stability, specificity, and multifunctionality. Dysregulation of circRNA expression is closely associated with DN progression, whereby circRNA impacts kidney cell injury by modulating cell cycle, differentiation, cell death, as well as influencing the release of inflammatory factors and stromal fibronectin expression. Consequently, circRNA is considered a predictive biomarker and a potential therapeutic target for DN. This review provides an overview of the latest research progress in the classification, functions, monitoring methods, and databases related to circRNA. The paper focuses on elucidating the impact and underlying mechanisms of circRNA on kidney cells under diabetic conditions, aiming to offer novel insights into the prevention, diagnosis, and treatment of DN.
Collapse
Affiliation(s)
- Haiying Shu
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China; College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Zhen Zhang
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China; School of First Clinical Medical College, Mudanjiang Medical University, Mudanjiang, China
| | - Jieting Liu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China.
| | - Peijian Chen
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China; College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Can Yang
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China; College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Yan Wu
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China; College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Dan Wu
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China; College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Yanan Cao
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China; College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Yanhui Chu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China.
| | - Luxin Li
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China; College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China.
| |
Collapse
|
18
|
Tsai LT, Weng TI, Chang TY, Lan KC, Chiang CK, Liu SH. Inhibition of Indoxyl Sulfate-Induced Reactive Oxygen Species-Related Ferroptosis Alleviates Renal Cell Injury In Vitro and Chronic Kidney Disease Progression In Vivo. Antioxidants (Basel) 2023; 12:1931. [PMID: 38001784 PMCID: PMC10669521 DOI: 10.3390/antiox12111931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
The accumulation of the uremic toxin indoxyl sulfate (IS) is a key pathological feature of chronic kidney disease (CKD). The effect of IS on ferroptosis and the role of IS-related ferroptosis in CKD are not well understood. We used a renal tubular cell model and an adenine-induced CKD mouse model to explore whether IS induces ferroptosis and injury and affects iron metabolism in the renal cells and the kidneys. Our results showed that exposure to IS induced several characteristics for ferroptosis, including iron accumulation, an impaired antioxidant system, elevated reactive oxygen species (ROS) levels, and lipid peroxidation. Exposure to IS triggered intracellular iron accumulation by upregulating transferrin and transferrin receptors, which are involved in cellular iron uptake. We also observed increased levels of the iron storage protein ferritin. The effects of IS-induced ROS generation, lipid peroxidation, ferroptosis, senescence, ER stress, and injury/fibrosis were effectively alleviated by treatments with an iron chelator deferoxamine (DFO) in vitro and the adsorbent charcoal AST-120 (scavenging the IS precursor) in vivo. Our findings suggest that IS triggers intracellular iron accumulation and ROS generation, leading to the induction of ferroptosis, senescence, ER stress, and injury/fibrosis in CKD kidneys. AST-120 administration may serve as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Li-Ting Tsai
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; (L.-T.T.); (T.-Y.C.); (C.-K.C.)
| | - Te-I Weng
- Department of Forensic Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Ting-Yu Chang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; (L.-T.T.); (T.-Y.C.); (C.-K.C.)
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chih-Kang Chiang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; (L.-T.T.); (T.-Y.C.); (C.-K.C.)
- Departments of Integrated Diagnostics & Therapeutics and Internal Medicine, College of Medicine and Hospital, National Taiwan University, Taipei 100, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; (L.-T.T.); (T.-Y.C.); (C.-K.C.)
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University & Hospital, Taipei 100, Taiwan
| |
Collapse
|
19
|
Roberts JA, Rainbow RD, Sharma P. Mitigation of Cardiovascular Disease and Toxicity through NRF2 Signalling. Int J Mol Sci 2023; 24:ijms24076723. [PMID: 37047696 PMCID: PMC10094784 DOI: 10.3390/ijms24076723] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Cardiovascular toxicity and diseases are phenomena that have a vastly detrimental impact on morbidity and mortality. The pathophysiology driving the development of these conditions is multifactorial but commonly includes the perturbance of reactive oxygen species (ROS) signalling, iron homeostasis and mitochondrial bioenergetics. The transcription factor nuclear factor erythroid 2 (NFE2)-related factor 2 (NRF2), a master regulator of cytoprotective responses, drives the expression of genes that provide resistance to oxidative, electrophilic and xenobiotic stresses. Recent research has suggested that stimulation of the NRF2 signalling pathway can alleviate cardiotoxicity and hallmarks of cardiovascular disease progression. However, dysregulation of NRF2 dynamic responses can be severely impacted by ageing processes and off-target toxicity from clinical medicines including anthracycline chemotherapeutics, rendering cells of the cardiovascular system susceptible to toxicity and subsequent tissue dysfunction. This review addresses the current understanding of NRF2 mechanisms under homeostatic and cardiovascular pathophysiological conditions within the context of wider implications for this diverse transcription factor.
Collapse
Affiliation(s)
- James A. Roberts
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Richard D. Rainbow
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Liverpool Centre for Cardiovascular Science, Liverpool L7 8TX, UK
| | - Parveen Sharma
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Liverpool Centre for Cardiovascular Science, Liverpool L7 8TX, UK
| |
Collapse
|