1
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [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: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
2
|
Wang Q, Li A, Li Q, Li J, Wang Q, Wu S, Meng J, Liu C, Wang D, Chen Y. Carbon monoxide attenuates cellular senescence-mediated pulmonary fibrosis via modulating p53/PAI-1 pathway. Eur J Pharmacol 2024; 980:176843. [PMID: 39068977 DOI: 10.1016/j.ejphar.2024.176843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE Idiopathic pulmonary fibrosis (IPF) is a fatal progressive condition often requiring lung transplantation. Accelerated senescence of type II alveolar epithelial cells (AECII) plays a crucial role in pulmonary fibrosis progression through the secretion of the senescence-associated secretory phenotype (SASP). Low-dose carbon monoxide (CO) possesses anti-inflammatory, anti-oxidative, and anti-aging properties. This study aims to explore the preventive effects of CO-releasing molecule 2 (CORM2) in a bleomycin-induced pulmonary fibrosis model. METHODS We established an pulmonary fibrosis model in C57BL/6J mice and evaluated the impact of CORM2 on fibrosis pathology using Masson's trichrome staining, fluorescence staining, and pulmonary function tests. Fibrogenic marker expression and SASP secretion in tissues and AECII cells were analyzed using qRT-PCR, Western blot, and ELISA assays both in vivo and in vitro. Additionally, we investigated DNA damage and cellular senescence through immunofluorescence and SA-β-gal staining. RESULTS CORM2 showed a preventive effect on bleomycin-induced lung fibrosis by improving pulmonary function and reducing the expression of fibrosis-related genes, such as TGF-β, α-SMA, Collagen I/III. CORM2 decreased the DNA damage response by inhibiting γ-H2AX, p53, and p21. We identified PAI-1 as a new target gene that was downregulated by CORM2, and which was associated with cellular senescence and fibrosis. CORM2 effectively inhibited cellular senescence and delayed EMT occurrence in AECII cells. CONCLUSION Our study highlights the potential of CORM2 in preventing DNA damage-induced cellular senescence in bleomycin-induced pulmonary fibrosis through modulation of the p53/PAI-1 signaling pathway. These findings underscore the promising prospects of CORM2 in targeting cellular senescence and the p53/PAI-1 pathway as a potential preventive strategy for IPF.
Collapse
Affiliation(s)
- Qianqian Wang
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China; Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622, Liaoning, China
| | - Aohan Li
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Qian Li
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Jiaxin Li
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China; Case Statistics Office, The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Harbin, 150011, China
| | - Qi Wang
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Siyuan Wu
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Jiaojiao Meng
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Changpeng Liu
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Dan Wang
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China; Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622, Liaoning, China.
| | - Yingqing Chen
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China; Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622, Liaoning, China.
| |
Collapse
|
3
|
Rinaldi A, Cippà PE, Nemazanyy I, Anglicheau D, Pallet N. Taurine Deficiency Is a Hallmark of Injured Kidney Allografts. Transplantation 2024; 108:e218-e228. [PMID: 39167563 DOI: 10.1097/tp.0000000000004987] [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] [Indexed: 03/21/2024]
Abstract
BACKGROUND Taurine is one of the most abundant amino acids in humans. Low taurine levels are associated with cellular senescence, mitochondrial dysfunction, DNA damage, and inflammation in mouse, all of which can be reversed by supplementation. It is unknown whether taurine metabolism is associated with kidney allograft function and survival. METHODS We performed urine metabolomic profiling of kidney transplant recipients in the early and late phases after transplantation combined with transcriptomic analysis of human kidney allografts. Single-nucleus RNA sequencing data sets of mouse kidneys after ischemia-reperfusion injury were analyzed. We analyzed the association of urinary taurine levels and taurine metabolism genes with kidney function, histology, and graft survival. RESULTS Urine taurine concentrations were significantly lower in kidney transplant recipients who experienced delayed graft function. In a mouse model of ischemia-reperfusion injury, the taurine biosynthesis gene, CSAD , but not the taurine transporter SLC6A6 , was repressed. In the late stage of transplantation, low level of taurine in urine was associated with impaired kidney function and chronic structural changes. Urine taurine level in the lowest tertile was predictive of graft loss. Expression of the taurine transporter SLC6A6 in the upper median, but not CSAD , was associated with chronic kidney injury and was predictive of graft loss. CONCLUSIONS Low urine taurine level is a marker of injury in the kidney allograft, is associated with poor kidney function, is associated with chronic histological changes, and is predictive of graft survival. The differential expression of CSAD and SLC6A6 , depending on the time after transplantation and marks of injury, highlights different mechanisms affecting taurine metabolism.
Collapse
Affiliation(s)
- Anna Rinaldi
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Pietro E Cippà
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, Institut National de la Santé et de la Recherche Médicale (INSERM) US24/CNRS UMS3633, Paris, France
| | - Dany Anglicheau
- INSERM U1151, Université Paris Cité, Paris, France
- Service de Néphrologie et Transplantation, Assistance Publique Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Nicolas Pallet
- Service de Biochimie, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
- Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Service de Néphrologie, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
4
|
Melatonin Promotes the Development of Secondary Hair Follicles in Adult Cashmere Goats by Activating the Keap1-Nrf2 Signaling Pathway and Inhibiting the Inflammatory Transcription Factors NFκB and AP-1. Int J Mol Sci 2023; 24:ijms24043403. [PMID: 36834812 PMCID: PMC9964152 DOI: 10.3390/ijms24043403] [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: 12/17/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/11/2023] Open
Abstract
Exogenous melatonin (MT) has been used to promote the growth of secondary hair follicles and improve cashmere fiber quality, but the specific cellular-level mechanisms involved are unclear. This study was carried out to investigate the effect of MT on the development of secondary hair follicles and on cashmere fiber quality in cashmere goats. The results showed that MT improved secondary follicle numbers and function as well as enhanced cashmere fiber quality and yield. The MT-treated goat groups had high secondary-to-primary ratios (S:P) for hair follicles, greater in the elderly group (p < 0.05). Antioxidant capacities of secondary hair follicles improved fiber quality and yield in comparison with control groups (p < 0.05/0.01). Levels of reactive oxygen and nitrogen species (ROS, RNS) and malondialdehyde (MDA) were lowered (p < 0.05/0.01) by MT. There was significant upregulation of antioxidant genes (for SOD-3; GPX-1; NFE2L2) and the protein of nuclear factor (Nrf2), and downregulation of the Keap1 protein. There were significant differences in the expression of genes for secretory senescence-associated phenotype (SASP) cytokines (IL-1β, IL-6, MMP-9, MMP-27, CCL-21, CXCL-12, CXCL-14, TIMP-1,2,3) plus their protein of key transcription factors, nuclear factor kappa B (NFκB) and activator protein-1 (AP-1), in comparison with the controls. We concluded that MT could enhance antioxidant capacity and reduce ROS and RNS levels of secondary hair follicles through the Keap1-Nrf2 pathway in adult cashmere goats. Furthermore, MT reduced the expression of the SASP cytokines genes by inhibiting the protein of NFκB and AP-1 in the secondary hair follicles in older cashmere goats, thus delaying skin aging, improving follicle survival, and increasing the number of secondary hair follicles. Collectively, these effects of exogenous MT enhanced the quality and yield of cashmere fibers, especially at 5-7 years old.
Collapse
|
5
|
Zhang JQ, Li YY, Zhang XY, Tian ZH, Liu C, Wang ST, Zhang FR. Cellular senescence of renal tubular epithelial cells in renal fibrosis. Front Endocrinol (Lausanne) 2023; 14:1085605. [PMID: 36926022 PMCID: PMC10011622 DOI: 10.3389/fendo.2023.1085605] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/14/2023] [Indexed: 03/08/2023] Open
Abstract
Renal fibrosis (RF) is the common pathological manifestation of virtually all chronic kidney diseases (CKD) and one of the major causes of end-stage renal disease (ESRD), but the pathogenesis of which is still unclear. Renal tubulointerstitial lesions have been identified as a key pathological hallmark of RF pathology. Renal tubular epithelial cells are the resident cells of the tubulointerstitium and play an important role in kidney recovery versus renal fibrosis following injury. Studies in recent years have shown that senescence of renal tubular epithelial cells can accelerate the progression of renal fibrosis. Oxidative stress(OS), telomere attrition and DNA damage are the major causes of renal tubular epithelial cell senescence. Current interventions and therapeutic strategies for cellular senescence include calorie restriction and routine exercise, Klotho, senolytics, senostatics, and other related drugs. This paper provides an overview of the mechanisms and the key signaling pathways including Wnt/β-catenin/RAS, Nrf2/ARE and STAT-3/NF-κB pathway involved in renal tubular epithelial cell senescence in RF and therapies targeting renal tubular epithelial cell senescence future therapeutic potential for RF patients. These findings may offer promise for the further treatment of RF and CKD.
Collapse
Affiliation(s)
- Jun-Qing Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying-Ying Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xue-Yan Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zeng-Hui Tian
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cheng Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shi-Tao Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fa-Rong Zhang
- Department of Nephrology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Fa-Rong Zhang,
| |
Collapse
|
6
|
Luo Y, Deng D, Lin L, Zhou Y, Wang L, Zou X, Wang X. FGF2 isoforms play distinct roles in tubular epithelial-to-mesenchymal transition in diabetic nephropathy. Exp Cell Res 2022; 420:113355. [PMID: 36115414 DOI: 10.1016/j.yexcr.2022.113355] [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: 03/17/2022] [Revised: 08/17/2022] [Accepted: 09/09/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The role of different isoforms of Fibroblast growth factor-2 (FGF2) in tubular epithelial-to-mesenchymal transition (EMT) in diabetic nephropathy remains unknown. We aimed to evaluate the role of FGF2 isoforms in the pathogenesis of EMT. MATERIALS AND METHODS Western blot and immunofluorescence were used to assess the expression of FGF2 isoforms in db/db mice and high glucose-stimulated HK2 cells. The effects of specific FGF2 isoforms on EMT were explored via overexpression or knockdown of the corresponding isoform in HK2 cells cultivated in high glucose. RESULTS Expression of low molecular weight (LMW) FGF2 was up-regulated while high molecular weight (HMW) FGF2 was down-regulated in the kidney of db/db mice and HK2 cells cultured in high glucose that underwent EMT. Overexpression of the LMW FGF2 enhanced EMT changes, while overexpression of the HMW FGF2 attenuated EMT. Knockdown of HMW FGF2 in HK2 cells promoted the EMT process. CONCLUSIONS The expression and function of LMW and HMW FGF2 differed in the process of EMT in tubular cells. LMW FGF2 contributed to EMT, while HMW FGF2 played a protective role in the EMT process.
Collapse
Affiliation(s)
- Yingying Luo
- School of Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Danfang Deng
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China
| | - Lamei Lin
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China
| | - Yikun Zhou
- School of Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Lan Wang
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China
| | - Xinrong Zou
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China
| | - Xiaoqin Wang
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China.
| |
Collapse
|
7
|
Hoff U, Markmann D, Thurn-Valassina D, Nieminen-Kelhä M, Erlangga Z, Schmitz J, Bräsen JH, Budde K, Melk A, Hegner B. The mTOR inhibitor Rapamycin protects from premature cellular senescence early after experimental kidney transplantation. PLoS One 2022; 17:e0266319. [PMID: 35446876 PMCID: PMC9022825 DOI: 10.1371/journal.pone.0266319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 03/18/2022] [Indexed: 01/07/2023] Open
Abstract
Interstitial fibrosis and tubular atrophy, a major cause of kidney allograft dysfunction, has been linked to premature cellular senescence. The mTOR inhibitor Rapamycin protects from senescence in experimental models, but its antiproliferative properties have raised concern early after transplantation particularly at higher doses. Its effect on senescence has not been studied in kidney transplantation, yet. Rapamycin was applied to a rat kidney transplantation model (3 mg/kg bodyweight loading dose, 1.5 mg/kg bodyweight daily dose) for 7 days. Low Rapamycin trough levels (2.1-6.8 ng/mL) prevented the accumulation of p16INK4a positive cells in tubules, interstitium, and glomerula. Expression of the cytokines MCP-1, IL-1β, and TNF-α, defining the proinflammatory senescence-associated secretory phenotype, was abrogated. Infiltration with monocytes/macrophages and CD8+ T-lymphocytes was reduced and tubular function was preserved by Rapamycin. Inhibition of mTOR was not associated with impaired structural recovery, higher glucose levels, or weight loss. mTOR inhibition with low-dose Rapamycin in the immediate posttransplant period protected from premature cellular senescence without negative effects on structural and functional recovery from preservation/reperfusion damage, glucose homeostasis, and growth in a rat kidney transplantation model. Reduced senescence might maintain the renal regenerative capacity rendering resilience to future injuries resulting in protection from interstitial fibrosis and tubular atrophy.
Collapse
Affiliation(s)
- Uwe Hoff
- Department of Nephrology and Critical Care Medicine, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Denise Markmann
- Nieren- und Dialysezentrum Schöneberg-Tempelhof, Berlin, Germany
| | | | - Melina Nieminen-Kelhä
- Departement of Neurosurgery, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | | | - Jessica Schmitz
- Nephropathology Unit, Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Jan Hinrich Bräsen
- Nephropathology Unit, Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Klemens Budde
- Department of Nephrology and Critical Care Medicine, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Anette Melk
- Childrens’ Hospital, Hannover Medical School, Hannover, Germany
| | - Björn Hegner
- Department of Nephrology and Critical Care Medicine, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Vitanas Hospital for Geriatric Medicine, Berlin, Germany
- * E-mail:
| |
Collapse
|
8
|
Liu L, Sun Q, Davis F, Mao J, Zhao H, Ma D. Epithelial-mesenchymal transition in organ fibrosis development: current understanding and treatment strategies. BURNS & TRAUMA 2022; 10:tkac011. [PMID: 35402628 PMCID: PMC8990740 DOI: 10.1093/burnst/tkac011] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/16/2021] [Indexed: 01/10/2023]
Abstract
Organ fibrosis is a process in which cellular homeostasis is disrupted and extracellular matrix is excessively deposited. Fibrosis can lead to vital organ failure and there are no effective treatments yet. Although epithelial–mesenchymal transition (EMT) may be one of the key cellular mechanisms, the underlying mechanisms of fibrosis remain largely unknown. EMT is a cell phenotypic process in which epithelial cells lose their cell-to-cell adhesion and polarization, after which they acquire mesenchymal features such as infiltration and migration ability. Upon injurious stimulation in different organs, EMT can be triggered by multiple signaling pathways and is also regulated by epigenetic mechanisms. This narrative review summarizes the current understanding of the underlying mechanisms of EMT in fibrogenesis and discusses potential strategies for attenuating EMT to prevent and/or inhibit fibrosis. Despite better understanding the role of EMT in fibrosis development, targeting EMT and beyond in developing therapeutics to tackle fibrosis is challenging but likely feasible.
Collapse
Affiliation(s)
- Lexin Liu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK.,Department of Nephrology and Urology, Pediatric Urolith Center, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, 310003, China
| | - Qizhe Sun
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Frank Davis
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Jianhua Mao
- Department of Nephrology, The Children Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| |
Collapse
|
9
|
Hu MC, Moe OW. Phosphate and Cellular Senescence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:55-72. [PMID: 35288873 PMCID: PMC10513121 DOI: 10.1007/978-3-030-91623-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cellular senescence is one type of permeant arrest of cell growth and one of increasingly recognized contributor to aging and age-associated disease. High phosphate and low Klotho individually and synergistically lead to age-related degeneration in multiple organs. Substantial evidence supports the causality of high phosphate in cellular senescence, and potential contribution to human aging, cancer, cardiovascular, kidney, neurodegenerative, and musculoskeletal diseases. Phosphate can induce cellular senescence both by direct phosphotoxicity, and indirectly through downregulation of Klotho and upregulation of plasminogen activator inhibitor-1. Restriction of dietary phosphate intake and blockage of intestinal absorption of phosphate help suppress cellular senescence. Supplementation of Klotho protein, cellular senescence inhibitor, and removal of senescent cells with senolytic agents are potential novel strategies to attenuate phosphate-induced cellular senescence, retard aging, and ameliorate age-associated, and phosphate-induced disorders.
Collapse
Affiliation(s)
- Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
10
|
Gong W, Luo C, Peng F, Xiao J, Zeng Y, Yin B, Chen X, Li S, He X, Liu Y, Cao H, Xu J, Long H. Brahma-related gene-1 promotes tubular senescence and renal fibrosis through Wnt/β-catenin/autophagy axis. Clin Sci (Lond) 2021; 135:1873-1895. [PMID: 34318888 PMCID: PMC8358963 DOI: 10.1042/cs20210447] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/09/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
Although accelerated cellular senescence is closely related to the progression of chronic kidney disease (CKD) and renal fibrosis, the underlying mechanisms remain largely unknown. Here, we reported that tubular aberrant expression of Brahma-related gene 1 (BRG1), an enzymatic subunit of the SWItch/Sucrose Non-Fermentable complex, is critically involved in tubular senescence and renal fibrosis. BRG1 was significantly up-regulated in the kidneys, predominantly in tubular epithelial cells, of both CKD patients and unilateral ureteral obstruction (UUO) mice. In vivo, shRNA-mediated knockdown of BRG1 significantly ameliorated renal fibrosis, improved tubular senescence, and inhibited UUO-induced activation of Wnt/β-catenin pathway. In mouse renal tubular epithelial cells (mTECs) and primary renal tubular cells, inhibition of BRG1 diminished transforming growth factor-β1 (TGF-β1)-induced cellular senescence and fibrotic responses. Correspondingly, ectopic expression of BRG1 in mTECs or normal kidneys increased p16INK4a, p19ARF, and p21 expression and senescence-associated β-galactosidase (SA-β-gal) activity, indicating accelerated tubular senescence. Additionally, BRG1-mediated pro-fibrotic responses were largely abolished by small interfering RNA (siRNA)-mediated p16INK4a silencing in vitro or continuous senolytic treatment with ABT-263 in vivo. Moreover, BRG1 activated the Wnt/β-catenin pathway, which further inhibited autophagy. Pharmacologic inhibition of the Wnt/β-catenin pathway (ICG-001) or rapamycin (RAPA)-mediated activation of autophagy effectively blocked BRG1-induced tubular senescence and fibrotic responses, while bafilomycin A1 (Baf A1)-mediated inhibition of autophagy abolished the effects of ICG-001. Further, BRG1 altered the secretome of senescent tubular cells, which promoted proliferation and activation of fibroblasts. Taken together, our results indicate that BRG1 induces tubular senescence by inhibiting autophagy via the Wnt/β-catenin pathway, which ultimately contributes to the development of renal fibrosis.
Collapse
Affiliation(s)
- Wangqiu Gong
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jing Xiao
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yiqun Zeng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Bohui Yin
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaowen Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaoyang He
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yanxia Liu
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Huihui Cao
- Traditional Chinese Pharmacological Laboratory, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jiangping Xu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
11
|
Xu J, Zhou L, Liu Y. Cellular Senescence in Kidney Fibrosis: Pathologic Significance and Therapeutic Strategies. Front Pharmacol 2020; 11:601325. [PMID: 33362554 PMCID: PMC7759549 DOI: 10.3389/fphar.2020.601325] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/21/2020] [Indexed: 01/10/2023] Open
Abstract
Age-related disorders such as chronic kidney disease (CKD) are increasingly prevalent globally and pose unprecedented challenges. In many aspects, CKD can be viewed as a state of accelerated and premature aging. Aging kidney and CKD share many common characteristic features with increased cellular senescence, a conserved program characterized by an irreversible cell cycle arrest with altered transcriptome and secretome. While developmental senescence and acute senescence may positively contribute to the fine-tuning of embryogenesis and injury repair, chronic senescence, when unresolved promptly, plays a crucial role in kidney fibrogenesis and CKD progression. Senescent cells elicit their fibrogenic actions primarily by secreting an assortment of inflammatory and profibrotic factors known as the senescence-associated secretory phenotype (SASP). Increasing evidence indicates that senescent cells could be a promising new target for therapeutic intervention known as senotherapy, which includes depleting senescent cells, modulating SASP and restoration of senescence inhibitors. In this review, we discuss current understanding of the role and mechanism of cellular senescence in kidney fibrosis. We also highlight potential options of targeting senescent cells for the treatment of CKD.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
12
|
Sheng L, Zhuang S. New Insights Into the Role and Mechanism of Partial Epithelial-Mesenchymal Transition in Kidney Fibrosis. Front Physiol 2020; 11:569322. [PMID: 33041867 PMCID: PMC7522479 DOI: 10.3389/fphys.2020.569322] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is described as the process in which injured renal tubular epithelial cells undergo a phenotype change, acquiring mesenchymal characteristics and morphing into fibroblasts. Initially, it was widely thought of as a critical mechanism of fibrogenesis underlying chronic kidney disease. However, evidence that renal tubular epithelial cells can cross the basement membrane and become fibroblasts in the renal interstitium is rare, leading to debate about the existence of EMT. Recent research has demonstrated that after injury, renal tubular epithelial cells acquire mesenchymal characteristics and the ability to produce a variety of profibrotic factors and cytokines, but remain attached to the basement membrane. On this basis, a new concept of “partial epithelial-mesenchymal transition (pEMT)” was proposed to explain the contribution of renal epithelial cells to renal fibrogenesis. In this review, we discuss the concept of pEMT and the most recent findings related to this process, including cell cycle arrest, metabolic alternation of epithelial cells, infiltration of immune cells, epigenetic regulation as well as the novel signaling pathways that mediate this disturbed epithelial-mesenchymal communication. A deeper understanding of the role and the mechanism of pEMT may help in developing novel therapies to prevent and halt fibrosis in kidney disease.
Collapse
Affiliation(s)
- Lili Sheng
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
13
|
Recent Advances on Biomarkers of Early and Late Kidney Graft Dysfunction. Int J Mol Sci 2020; 21:ijms21155404. [PMID: 32751357 PMCID: PMC7432796 DOI: 10.3390/ijms21155404] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
New biomarkers of early and late graft dysfunction are needed in renal transplant to improve management of complications and prolong graft survival. A wide range of potential diagnostic and prognostic biomarkers, measured in different biological fluids (serum, plasma, urine) and in renal tissues, have been proposed for post-transplant delayed graft function (DGF), acute rejection (AR), and chronic allograft dysfunction (CAD). This review investigates old and new potential biomarkers for each of these clinical domains, seeking to underline their limits and strengths. OMICs technology has allowed identifying many candidate biomarkers, providing diagnostic and prognostic information at very early stages of pathological processes, such as AR. Donor-derived cell-free DNA (ddcfDNA) and extracellular vesicles (EVs) are further promising tools. Although most of these biomarkers still need to be validated in multiple independent cohorts and standardized, they are paving the way for substantial advances, such as the possibility of accurately predicting risk of DGF before graft is implanted, of making a “molecular” diagnosis of subclinical rejection even before histological lesions develop, or of dissecting etiology of CAD. Identification of “immunoquiescent” or even tolerant patients to guide minimization of immunosuppressive therapy is another area of active research. The parallel progress in imaging techniques, bioinformatics, and artificial intelligence (AI) is helping to fully exploit the wealth of information provided by biomarkers, leading to improved disease nosology of old entities such as transplant glomerulopathy. Prospective studies are needed to assess whether introduction of these new sets of biomarkers into clinical practice could actually reduce the need for renal biopsy, integrate traditional tools, and ultimately improve graft survival compared to current management.
Collapse
|
14
|
Kooman JP, Dekker MJ, Usvyat LA, Kotanko P, van der Sande FM, Schalkwijk CG, Shiels PG, Stenvinkel P. Inflammation and premature aging in advanced chronic kidney disease. Am J Physiol Renal Physiol 2017; 313:F938-F950. [PMID: 28701312 DOI: 10.1152/ajprenal.00256.2017] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/29/2017] [Accepted: 07/06/2017] [Indexed: 12/22/2022] Open
Abstract
Systemic inflammation in end-stage renal disease is an established risk factor for mortality and a catalyst for other complications, which are related to a premature aging phenotype, including muscle wasting, vascular calcification, and other forms of premature vascular disease, depression, osteoporosis, and frailty. Uremic inflammation is also mechanistically related to mechanisms involved in the aging process, such as telomere shortening, mitochondrial dysfunction, and altered nutrient sensing, which can have a direct effect on cellular and tissue function. In addition to uremia-specific causes, such as abnormalities in the phosphate-Klotho axis, there are remarkable similarities between the pathophysiology of uremic inflammation and so-called "inflammaging" in the general population. Potentially relevant, but still somewhat unexplored in this respect, are abnormal or misplaced protein structures, as well as abnormalities in tissue homeostasis, which evoke danger signals through damage-associated molecular patterns, as well as the senescence-associated secretory phenotype. Systemic inflammation, in combination with the loss of kidney function, can impair the resilience of the body to external and internal stressors by reduced functional and structural tissue reserves, and by impairing normal organ crosstalk, thus providing an explanation for the greatly increased risk of homeostatic breakdown in this population. In this review, the relationship between uremic inflammation and a premature aging phenotype, as well as potential causes and consequences, are discussed.
Collapse
Affiliation(s)
- Jeroen P Kooman
- Maastricht University Medical Center, Maastricht, Netherlands;
| | | | - Len A Usvyat
- Fresenius Medical Care North America, Waltham, Massachusetts
| | - Peter Kotanko
- Renal Research Institute, New York, New York.,Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Paul G Shiels
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; and
| | - Peter Stenvinkel
- Divsion of Renal Medicine, Department of Clinical Science Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|