1
|
Borri M, Jacobs ME, Carmeliet P, Rabelink TJ, Dumas SJ. Endothelial dysfunction in the aging kidney. Am J Physiol Renal Physiol 2025; 328:F542-F562. [PMID: 39933752 DOI: 10.1152/ajprenal.00287.2024] [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: 10/08/2024] [Revised: 11/07/2024] [Accepted: 02/07/2025] [Indexed: 02/13/2025] Open
Abstract
Global population aging is an escalating challenge in modern society, especially as it impairs the function of multiple organs and increases the burden of age-related diseases. The kidneys, in particular, experience function decline, reduced regenerative capacity, and increased susceptibility to injury as they age. As a result, the prevalence of chronic kidney disease (CKD) rises with aging, further contributing to the growing health burden in older populations. One of the key factors in this process is the dysfunction of specialized renal endothelial cells (RECs), which are essential for maintaining kidney health by regulating blood flow and supporting filtration, solute and water reabsorption, and vascular integrity. As the kidneys age, REC dysfunction drives vascular and microenvironmental changes, contributing to the overall decline in kidney function. In this review, we outline the structural and functional effects of aging on the kidney's macrovascular and microvascular compartments and provide a phenotypic description of the aged endothelium. We particularly focus on the molecular and metabolic rewiring driving and sustaining growth-arrested EC senescence phenotype. We finally give an overview of senotherapies acting on ECs, especially of those modulating metabolism. Given that the pathophysiological processes underlying kidney aging largely overlap with those observed in CKD, REC rejuvenation could also benefit patients with CKD. Moreover, such interventions may hold promise in improving the outcomes of aged kidney transplants. Hence, advancing our understanding of REC and kidney aging will create opportunities for innovations that could improve outcomes for both elderly individuals and patients with CKD.
Collapse
Affiliation(s)
- Mila Borri
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Marleen E Jacobs
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ton J Rabelink
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Sébastien J Dumas
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Ma M, Zeng J, Zhu M, Li H, Lin T, Yang H, Wei X, Song T. Human umbilical cord mesenchymal stem cells-derived extracellular vesicles ameliorate kidney ischemia-reperfusion injury by suppression of senescent tubular epithelial cells: experimental study. Int J Surg 2025; 111:394-410. [PMID: 39236098 PMCID: PMC11745712 DOI: 10.1097/js9.0000000000002074] [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: 05/15/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Human umbilical cord mesenchymal stem cells-derived extracellular vesicles (HUMSC-EVs) have drawn much interest in kidney transplantation, mainly because of their renoprotection by alleviating cell injury and stimulating tissue repair. Cellular senescence has been proven to play a dual regulatory role in kidney ischemia-reperfusion injury (IRI), and the regulation of HUMSC-EVs on tubular epithelial cell senescence may be a potential therapeutic target. MATERIALS AND METHODS In vitro , the hypoxia-reoxygenation of human kidney-2 cells was used to simulate kidney IRI, and the regulation of HUMSC-EVs on human kidney-2 cells was detected. Transcriptome sequencing of human kidney-2 cells was used to explore the potential regulatory mechanism. In vivo , adult male mice were divided into five groups: control group, IRI group, HUMSC-EVs treatment group, senolytics treatment group (dasatinib + quercetin), and combined treatments group (HUMSC-EVs and senolytics). Kidney function, senescent features of tubular epithelial cells, acute kidney injury, and chronic interstitial fibrosis in mice were detected to explore the renoprotection effects of HUMSC-EVs. RESULTS Kidney IRI significantly up-regulated expressions of LaminB1, p53, p21, p16, senescence-associated beta-galactosidase, and apoptosis of tubular epithelial cells. In the mouse kidney IRI model, kidney subcapsular injection of HUMSC-EVs significantly improved kidney function, reducing the senescent features of tubular epithelial cells and alleviating acute kidney injury and chronic interstitial fibrosis. HUMSC-EVs mainly achieved renoprotection by regulating Bax/Bcl-2-dependent apoptosis during acute kidney injury and mostly reduced tubular atrophy and kidney interstitial fibrosis by regulating Ras-pERK-Ets1-p53 pathway-dependent cell senescence. Oral administration of senolytics also alleviated kidney injury induced by IRI, while the combined treatments of HUMSC-EVs and senolytics had better renoprotection effects. CONCLUSIONS The combination of HUMSC-EVs and senolytics alleviated acute kidney injury and chronic interstitial fibrosis by dynamic regulation of cell senescence and apoptosis, which provides a therapeutic potential strategy for organ preservation and tissue repair in kidney transplantation.
Collapse
Affiliation(s)
- Ming Ma
- Department of Urology, West China Hospital, Sichuan University
- Organ Transplantation Center, West China Hospital, Sichuan University
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University
| | - Jun Zeng
- Department of Urology, West China Hospital, Sichuan University
- Organ Transplantation Center, West China Hospital, Sichuan University
| | - Mengli Zhu
- Core Facilities of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Li
- Department of Urology, West China Hospital, Sichuan University
- Organ Transplantation Center, West China Hospital, Sichuan University
| | - Tao Lin
- Department of Urology, West China Hospital, Sichuan University
- Organ Transplantation Center, West China Hospital, Sichuan University
| | - Hao Yang
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University
| | - Xin Wei
- Department of Urology, West China Hospital, Sichuan University
| | - Turun Song
- Department of Urology, West China Hospital, Sichuan University
- Organ Transplantation Center, West China Hospital, Sichuan University
| |
Collapse
|
3
|
Hejazian SM, Hejazian SS, Mostafavi SM, Hosseiniyan SM, Montazersaheb S, Ardalan M, Zununi Vahed S, Barzegari A. Targeting cellular senescence in kidney diseases and aging: A focus on mesenchymal stem cells and their paracrine factors. Cell Commun Signal 2024; 22:609. [PMID: 39696575 DOI: 10.1186/s12964-024-01968-1] [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: 09/15/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Cellular senescence is a phenomenon distinguished by the halting of cellular division, typically triggered by DNA injury or numerous stress-inducing factors. Cellular senescence is implicated in various pathological and physiological processes and is a hallmark of aging. The presence of accumulated senescent cells, whether transiently (acute senescence) or persistently (chronic senescence) plays a dual role in various conditions such as natural kidney aging and different kidney disorders. Elevations in senescent cells and senescence-associated secretory phenotype (SASP) levels correlate with decreased kidney function, kidney ailments, and age-related conditions. Strategies involving senotherapeutic agents like senolytics, senomorphics, and senoinflammation have been devised to specifically target senescent cells. Mesenchymal stem cells (MSCs) and their secreted factors may also offer alternative approaches for anti-senescence interventions. The MSC-derived secretome compromises significant therapeutic benefits in kidney diseases by facilitating tissue repair via anti-inflammatory, anti-fibrosis, anti-apoptotic, and pro-angiogenesis effects, thereby improving kidney function and mitigating disease progression. Moreover, by promoting the clearance of senescent cells or modulating their secretory profiles, MSCs could potentially reverse some age-related declines in kidney function.This review article intends to shed light on the present discoveries concerning the role of cellular senescence in kidney aging and diseases. Furthermore, it outlines the role of senotherapeutics utilized to alleviate kidney damage and aging. It also highlights the possible impact of MSCs secretome on mitigating kidney injury and prolonging lifespan across various models of kidney diseases as a novel senotherapy.
Collapse
Affiliation(s)
| | - Seyyed Sina Hejazian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyedeh Mina Mostafavi
- Ayatollah Taleghani Hospital, Research Development Unit, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Kirchner VA, Badshah JS, Kyun Hong S, Martinez O, Pruett TL, Niedernhofer LJ. Effect of Cellular Senescence in Disease Progression and Transplantation: Immune Cells and Solid Organs. Transplantation 2024; 108:1509-1523. [PMID: 37953486 PMCID: PMC11089077 DOI: 10.1097/tp.0000000000004838] [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: 11/14/2023]
Abstract
Aging of the world population significantly impacts healthcare globally and specifically, the field of transplantation. Together with end-organ dysfunction and prolonged immunosuppression, age increases the frequency of comorbid chronic diseases in transplant candidates and recipients, contributing to inferior outcomes. Although the frequency of death increases with age, limited use of organs from older deceased donors reflects the concerns about organ durability and inadequate function. Cellular senescence (CS) is a hallmark of aging, which occurs in response to a myriad of cellular stressors, leading to activation of signaling cascades that stably arrest cell cycle progression to prevent tumorigenesis. In aging and chronic conditions, senescent cells accumulate as the immune system's ability to clear them wanes, which is causally implicated in the progression of chronic diseases, immune dysfunction, organ damage, decreased regenerative capacity, and aging itself. The intimate interplay between senescent cells, their proinflammatory secretome, and immune cells results in a positive feedback loop, propagating chronic sterile inflammation and the spread of CS. Hence, senescent cells in organs from older donors trigger the recipient's alloimmune response, resulting in the increased risk of graft loss. Eliminating senescent cells or attenuating their inflammatory phenotype is a novel, potential therapeutic target to improve transplant outcomes and expand utilization of organs from older donors. This review focuses on the current knowledge about the impact of CS on circulating immune cells in the context of organ damage and disease progression, discusses the impact of CS on abdominal solid organs that are commonly transplanted, and reviews emerging therapies that target CS.
Collapse
Affiliation(s)
- Varvara A. Kirchner
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA
| | - Joshua S. Badshah
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA
| | - Suk Kyun Hong
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Olivia Martinez
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA
| | - Timothy L. Pruett
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN
| |
Collapse
|
5
|
Cremen S, Santiago RM, Robinson MW, Gallagher TK. Biomarkers of biological aging in recipients of solid organ transplantation and clinical outcomes: A scoping review. Transpl Immunol 2023; 79:101851. [PMID: 37182719 DOI: 10.1016/j.trim.2023.101851] [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: 11/21/2022] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Biological aging is the accumulation of cellular and molecular damage within an individual over time. The biological age of a donor organ is known to influence clinical outcomes of solid organ transplantation, including delayed graft function and frequency of rejection episodes. While much research has focused on the biological age of donor organs, the recipient's biological age may also influence transplantation outcomes. The aim of this scoping review was to identify and provide an overview of the existing evidence regarding biological aging in solid organ transplant recipients and the impact on patient outcomes post-transplant. METHODS Literature searches were carried out on PubMed, Web of Science, Google Scholar, Embase and TRIP using the phrases 'solid organ transplant', 'cell senescence', 'cell aging' and 'outcomes', using boolean 'and/or' phrases and MeSH terms. Duplicates were removed and abstracts were reviewed by two independent reviewers. Full papers were then screened for inclusion by two reviewers. Data extraction was carried out using a standardised proforma agreed on prior to starting. RESULTS 32 studies, including data on a total of 7760 patients, were identified for inclusion in this review; 23 relating to kidney transplant recipients, three to liver transplant, five to lung transplant and one to heart transplantation. A wide range of biomarkers of biological aging have been assessed in kidney transplant recipients, whereas studies of liver, lung and heart transplant have predominantly assessed recipient telomere length. The most robust associations with clinical outcomes are observed in kidney transplant recipients, possibly influenced by the larger number of studies and the use of a wider range of biomarkers of biological aging. In kidney transplant recipients reduced thymic function and accumulation of terminally differentiated T cell populations was associated with reduced risk of acute rejection but increased risk of infection and mortality. CONCLUSION Studies to date on biological aging in transplant recipients have been heavily biased to kidney transplant recipients. The results from these studies suggest recipient biological age can influence clinical outcomes and future research is needed to prioritise robust biomarkers of biological aging in transplant recipients.
Collapse
Affiliation(s)
- S Cremen
- Department of Hepatobiliary and Transplant Surgery, St Vincent's University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland
| | - R M Santiago
- Department of Biology, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Kildare, Ireland
| | - M W Robinson
- Department of Biology, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Kildare, Ireland.
| | - T K Gallagher
- Department of Hepatobiliary and Transplant Surgery, St Vincent's University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
6
|
Valentijn FA, Knoppert SN, Marquez-Exposito L, Rodrigues-Diez RR, Pissas G, Tang J, Tejedor-Santamaria L, Broekhuizen R, Samarakoon R, Eleftheriadis T, Goldschmeding R, Nguyen TQ, Ruiz-Ortega M, Falke LL. Cellular communication network 2 (connective tissue growth factor) aggravates acute DNA damage and subsequent DNA damage response-senescence-fibrosis following kidney ischemia reperfusion injury. Kidney Int 2022; 102:1305-1319. [PMID: 35921911 DOI: 10.1016/j.kint.2022.06.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/17/2022] [Accepted: 06/23/2022] [Indexed: 01/12/2023]
Abstract
Chronic allograft dysfunction with progressive fibrosis of unknown cause remains a major issue after kidney transplantation, characterized by ischemia-reperfusion injury (IRI). One hypothesis to account for this is that spontaneous progressive tubulointerstitial fibrosis following IRI is driven by cellular senescence evolving from a prolonged, unresolved DNA damage response (DDR). Since cellular communication network factor 2 ((CCN2), formerly called connective tissue growth factor), an established mediator of kidney fibrosis, is also involved in senescence-associated pathways, we investigated the relation between CCN2 and cellular senescence following kidney transplantation. Tubular CCN2 overexpression was found to be associated with DDR, loss of kidney function and tubulointerstitial fibrosis in both the early and the late phase in human kidney allograft biopsies. Consistently, CCN2 deficient mice developed reduced senescence and tubulointerstitial fibrosis in the late phase; six weeks after experimental IRI. Moreover, tubular DDR markers and plasma urea were less elevated in CCN2 knockout than in wild-type mice. Finally, CCN2 administration or overexpression in epithelial cells induced upregulation of tubular senescence-associated genes including p21, while silencing of CCN2 alleviated DDR induced by anoxia-reoxygenation injury in cultured proximal tubule epithelial cells. Thus, our observations indicate that inhibition of CCN2 can mitigate IRI-induced acute kidney injury, DNA damage, and the subsequent DDR-senescence-fibrosis sequence. Hence, targeting CCN2 might help to protect the kidney from transplantation-associated post-IRI chronic kidney dysfunction.
Collapse
Affiliation(s)
- Floris A Valentijn
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Sebastiaan N Knoppert
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Laura Marquez-Exposito
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria -Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain
| | - Raúl R Rodrigues-Diez
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria -Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain
| | - Georgios Pissas
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Jiaqi Tang
- Center for Cell Biology and Cancer Research, Albany Medical Center, Albany, New York, USA
| | - Lucia Tejedor-Santamaria
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria -Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain
| | - Roel Broekhuizen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rohan Samarakoon
- Center for Cell Biology and Cancer Research, Albany Medical Center, Albany, New York, USA
| | | | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria -Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain
| | - Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
7
|
O’Sullivan ED, Mylonas KJ, Bell R, Carvalho C, Baird DP, Cairns C, Gallagher KM, Campbell R, Docherty M, Laird A, Henderson NC, Chandra T, Kirschner K, Conway B, Dihazi GH, Zeisberg M, Hughes J, Denby L, Dihazi H, Ferenbach DA. Single-cell analysis of senescent epithelia reveals targetable mechanisms promoting fibrosis. JCI Insight 2022; 7:e154124. [PMID: 36509292 PMCID: PMC9746814 DOI: 10.1172/jci.insight.154124] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/05/2022] [Indexed: 11/22/2022] Open
Abstract
Progressive fibrosis and maladaptive organ repair result in significant morbidity and millions of premature deaths annually. Senescent cells accumulate with aging and after injury and are implicated in organ fibrosis, but the mechanisms by which senescence influences repair are poorly understood. Using 2 murine models of injury and repair, we show that obstructive injury generated senescent epithelia, which persisted after resolution of the original injury, promoted ongoing fibrosis, and impeded adaptive repair. Depletion of senescent cells with ABT-263 reduced fibrosis in reversed ureteric obstruction and after renal ischemia/reperfusion injury. We validated these findings in humans, showing that senescence and fibrosis persisted after relieved renal obstruction. We next characterized senescent epithelia in murine renal injury using single-cell RNA-Seq. We extended our classification to human kidney and liver disease and identified conserved profibrotic proteins, which we validated in vitro and in human disease. We demonstrated that increased levels of protein disulfide isomerase family A member 3 (PDIA3) augmented TGF-β-mediated fibroblast activation. Inhibition of PDIA3 in vivo significantly reduced kidney fibrosis during ongoing renal injury and as such represented a new potential therapeutic pathway. Analysis of the signaling pathways of senescent epithelia connected senescence to organ fibrosis, permitting rational design of antifibrotic therapies.
Collapse
Affiliation(s)
- Eoin D. O’Sullivan
- Centre for Inflammation Research, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
| | - Katie J. Mylonas
- Centre for Inflammation Research, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Rachel Bell
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Cyril Carvalho
- Centre for Inflammation Research, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - David P. Baird
- Centre for Inflammation Research, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Carolynn Cairns
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Kevin M. Gallagher
- Department of Urology, Western General Hospital, Edinburgh, United Kingdom
| | - Ross Campbell
- Centre for Inflammation Research, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Marie Docherty
- Centre for Inflammation Research, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Alexander Laird
- Department of Urology, Western General Hospital, Edinburgh, United Kingdom
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil C. Henderson
- Centre for Inflammation Research, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Tamir Chandra
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Kristina Kirschner
- The Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Bryan Conway
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Jeremy Hughes
- Centre for Inflammation Research, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Laura Denby
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
| | - Hassan Dihazi
- Clinic for Nephrology and Rheumatology, and
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Göttingen, Germany
| | - David A. Ferenbach
- Centre for Inflammation Research, Queen’s Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
8
|
Liao CM, Wulfmeyer VC, Chen R, Erlangga Z, Sinning J, von Mässenhausen A, Sörensen-Zender I, Beer K, von Vietinghoff S, Haller H, Linkermann A, Melk A, Schmitt R. Induction of ferroptosis selectively eliminates senescent tubular cells. Am J Transplant 2022; 22:2158-2168. [PMID: 35607817 DOI: 10.1111/ajt.17102] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/27/2022] [Accepted: 05/22/2022] [Indexed: 01/25/2023]
Abstract
The accumulation of senescent cells is an important contributor to kidney aging, chronic renal disease, and poor outcome after kidney transplantation. Approaches to eliminate senescent cells with senolytic compounds have been proposed as novel strategies to improve marginal organs. While most existing senolytics induce senescent cell clearance by apoptosis, we observed that ferroptosis, an iron-catalyzed subtype of regulated necrosis, might serve as an alternative way to ablate senescent cells. We found that murine kidney tubular epithelial cells became sensitized to ferroptosis when turning senescent. This was linked to increased expression of pro-ferroptotic lipoxygenase-5 and reduced expression of anti-ferroptotic glutathione peroxidase 4 (GPX4). In tissue slice cultures from aged kidneys low dose application of the ferroptosis-inducer RSL3 selectively eliminated senescent cells while leaving healthy tubular cells unaffected. Similar results were seen in a transplantation model, in which RSL3 reduced the senescent cell burden of aged donor kidneys and caused a reduction of damage and inflammatory cell infiltration during the early post-transplantation period. In summary, these data reveal an increased susceptibility of senescent tubular cells to ferroptosis with the potential to be exploited for selective reduction of renal senescence in aged kidney transplants.
Collapse
Affiliation(s)
- Chieh M Liao
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Vera C Wulfmeyer
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Rongjun Chen
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Medical School Hannover, Hannover, Germany
| | - Zulrahman Erlangga
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Medical School Hannover, Hannover, Germany
| | - Julius Sinning
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Anne von Mässenhausen
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische University of Dresden, Dresden, Germany
| | - Inga Sörensen-Zender
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Kristina Beer
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische University of Dresden, Dresden, Germany
| | - Sibylle von Vietinghoff
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany.,Nephrology Section, First Medical Clinic, University Clinic and Rheinische Friedrich-Wilhelms Universität Bonn, Bonn, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische University of Dresden, Dresden, Germany
| | - Anette Melk
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Medical School Hannover, Hannover, Germany
| | - Roland Schmitt
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| |
Collapse
|
9
|
Hara S, Yamaguchi Y, Zoshima T, Mizushima I, Yamada K, Inoue R, Muto H, Mizutomi K, Hirata M, Araki H, Miyazaki R, Kawano M. Central fibrous area in the glomerular vascular pole consists of fibrous collagens and is associated with advanced age: a cross-sectional study. BMC Nephrol 2022; 23:204. [PMID: 35690711 PMCID: PMC9188109 DOI: 10.1186/s12882-022-02835-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/18/2022] [Indexed: 11/23/2022] Open
Abstract
Background For the optimal management of patients with both allograft kidneys and native kidney diseases, the recognition of the histological features associated with older age is important. This is because most pathological findings are non-specific. Central fibrous areas (CFAs) have recently been proposed to be age-related. However, the components of CFAs and whether CFAs are observed in various kidney diseases remain undetermined. This cross-sectional study was undertaken to clarify the histological features, epidemiology, and clinicopathological features of CFAs. Methods One hundred and one consecutive kidney needle biopsy specimens were retrospectively collected from seven facilities in the Hokuriku region and diagnosed at the Kanazawa University Hospital in 2015. First, the components of CFAs were analyzed using normal histostaining, immunostaining, and electron microscopy. Second, the patients were divided into two groups (CFA [+] or CFA [−]) according to the presence of CFA in the obtained samples. Clinical and histological features were compared between the two groups, and factors associated with CFA formation were determined using univariate and multivariate analyses. The number of CFAs per specimen was counted in the CFA (+) group. Third, the presence of myofibroblasts in CFA was examined by immunostaining. Results CFAs were observed in 56 of 101 patients (55.4%) with various kidney diseases. CFAs consist of fibrillar collagens (collagen I and III) in addition to non-fibrillar collagens (collagen IV and VI), as confirmed by electron microscopy. Clinically, the CFA (+) group was older and had a significantly higher prevalence of hypertension and hyperlipidemia than the CFA (−) group. Histologically, elastofibrosis of the interlobular artery, arteriolar hyalinosis, and membranous nephropathy were significantly more evident in the CFA (+) group than in the CFA (−) group. Multivariate analysis revealed that older age was the sole factor associated with CFA formation. Finally, 27 of 58 (46.6%) CFA-containing glomeruli in 26 cases included alpha-smooth muscle actin-positive cells in or adjacent to the CFA. Conclusions CFAs consist of fibrous collagens in addition to matrix collagens. CFA formation is associated with older age and was observed in various kidney diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02835-2.
Collapse
Affiliation(s)
- Satoshi Hara
- Department of Rheumatology, Kanazawa University Graduate School of Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan.,Medical Education Research Center, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Yutaka Yamaguchi
- Yamaguchi's Pathology Laboratory, 1-31-20Minoridai, Matsudo, Chiba, Japan
| | - Takeshi Zoshima
- Department of Rheumatology, Kanazawa University Graduate School of Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Ichiro Mizushima
- Department of Rheumatology, Kanazawa University Graduate School of Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Kazunori Yamada
- Department of Rheumatology, Kanazawa University Graduate School of Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Ryo Inoue
- Department of Internal Medicine, Saiseikai Kanazawa Hospital, Ni13-6 Akatsuchimachi, Kanazawa, Ishikawa, Japan
| | - Hisao Muto
- Department of Internal Medicine, Hokuriku Central Hospital, 123 Nodera, Oyabe, Toyama, Japan
| | - Kazuaki Mizutomi
- Department of Internal Medicine, Kaga Medical Center, Ri 36 Sakumimachi, Kaga, Ishikawa, Japan
| | - Masayoshi Hirata
- Department of Internal Medicine, Takaoka City Hospital, 4-1 Takaramachi, Takaoka, Toyama, Japan
| | - Hideo Araki
- Department of Nephrology and Rheumatology, Fukui Prefecture Hospital, 2-8-1 Yotsui, Fukui, Fukui, Japan
| | - Ryoichi Miyazaki
- Department of Internal Medicine, Fujita Memorial Hospital, 4-15-7 Houei, Fukui, Fukui, Japan
| | - Mitsuhiro Kawano
- Department of Rheumatology, Kanazawa University Graduate School of Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan.
| |
Collapse
|
10
|
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: 0.3] [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
|
11
|
Chan J, Eide IA, Tannæs TM, Waldum-Grevbo B, Jenssen T, Svensson M. Marine n-3 Polyunsaturated Fatty Acids and Cellular Senescence Markers in Incident Kidney Transplant Recipients: The Omega-3 Fatty Acids in Renal Transplantation (ORENTRA) Randomized Clinical Trial. Kidney Med 2021; 3:1041-1049. [PMID: 34939013 PMCID: PMC8664741 DOI: 10.1016/j.xkme.2021.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Rationale & Objective Deterioration of kidney graft function is associated with accelerated cellular senescence. Marine n-3 polyunsaturated fatty acids (PUFAs) have favorable properties that may counteract cellular senescence development and damage caused by the senescence-associated secretory phenotype (SASP) secretome. Our objective was to investigate the potential effects of marine n-3 PUFA supplementation on the SASP secretome in kidney transplant recipients. Study Design Exploratory substudy of the Omega-3 Fatty Acids in Renal Transplantation trial. Setting & Participants Adult kidney transplant recipients with a functional kidney graft (defined as having an estimated glomerular filtration rate of >30 mL/min/1.73 m2) 8 weeks after engraftment were included in this study conducted in Norway. Analytical Approach The intervention consisted of 2.6 g of a marine n-3 PUFA or olive oil (placebo) daily for 44 weeks. The outcome was a predefined panel of SASP components in the plasma and urine. Results A total of 132 patients were enrolled in the Omega-3 Fatty Acids in Renal Transplantation trial, and 66 patients were allocated to receive either the study drug or placebo. The intervention with the marine n-3 PUFA was associated with reduced plasma levels of granulocyte colony-stimulating factor, interleukin 1α, macrophage inflammatory protein 1α, matrix metalloproteinase (MMP)-1, and MMP-13 compared with the intervention in the control group. Limitations Post hoc analysis. Conclusions The results suggest that marine n-3 PUFA supplementation has mitigating effects on the plasma SASP components granulocyte colony-stimulating factor, interleukin 1α, macrophage inflammatory protein 1α, MMP-1, and MMP-13 in kidney transplant recipients. Future studies with kidney transplant recipients in maintenance phase, combined with an evaluation of cellular senescence markers in kidney transplant biopsies, are needed to further elucidate the potential antisenescent effect of marine n-3 PUFAs. This trial is registered as NCT01744067.
Collapse
Affiliation(s)
- Joe Chan
- Department of Renal Medicine, Akershus University Hospital, Lørenskog.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo
| | - Ivar A Eide
- Department of Renal Medicine, Akershus University Hospital, Lørenskog.,Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo
| | - Tone M Tannæs
- Department of Clinical Molecular Biology (EpiGen), Division of Medicine, Akershus University Hospital and University of Oslo, Lørenskog
| | - Bård Waldum-Grevbo
- Department of Nephrology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Trond Jenssen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo.,Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo
| | - My Svensson
- Department of Renal Medicine, Akershus University Hospital, Lørenskog.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo
| |
Collapse
|
12
|
Picerno A, Stasi A, Franzin R, Curci C, di Bari I, Gesualdo L, Sallustio F. Why stem/progenitor cells lose their regenerative potential. World J Stem Cells 2021; 13:1714-1732. [PMID: 34909119 PMCID: PMC8641024 DOI: 10.4252/wjsc.v13.i11.1714] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/26/2021] [Accepted: 10/31/2021] [Indexed: 02/06/2023] Open
Abstract
Nowadays, it is clear that adult stem cells, also called as tissue stem cells, play a central role to repair and maintain the tissue in which they reside by their self-renewal ability and capacity of differentiating into distinct and specialized cells. As stem cells age, their renewal ability declines and their capacity to maintain organ homeostasis and regeneration is impaired. From a molecular perspective, these changes in stem cells properties can be due to several types of cell intrinsic injury and DNA aberrant alteration (i.e epigenomic profile) as well as changes in the tissue microenviroment, both into the niche and by systemic circulating factors. Strikingly, it has been suggested that aging-induced deterioration of stem cell functions may play a key role in the pathophysiology of the various aging-associated disorders. Therefore, understanding how resident stem cell age and affects near and distant tissues is fundamental. Here, we examine the current knowledge about aging mechanisms in several kinds of adult stem cells under physiological and pathological conditions and the principal aging-related changes in number, function and phenotype that determine the loss of tissue renewal properties. Furthermore, we examine the possible cell rejuvenation strategies. Stem cell rejuvenation may reverse the aging phenotype and the discovery of effective methods for inducing and differentiating pluripotent stem cells for cell replacement therapies could open up new possibilities for treating age-related diseases.
Collapse
Affiliation(s)
- Angela Picerno
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Bari 70124, Italy
| | - Alessandra Stasi
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Bari 70124, Italy
| | - Rossana Franzin
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Bari 70124, Italy
| | - Claudia Curci
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Bari 70124, Italy
| | - Ighli di Bari
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Bari 70124, Italy
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Bari 70124, Italy
| | - Fabio Sallustio
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Bari 70124, Italy
| |
Collapse
|
13
|
Senescence and senolytics in cardiovascular disease: Promise and potential pitfalls. Mech Ageing Dev 2021; 198:111540. [PMID: 34237321 PMCID: PMC8387860 DOI: 10.1016/j.mad.2021.111540] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 02/08/2023]
Abstract
Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the cause of death in 40 % of individuals over 65 years old. Ageing is associated with an increased prevalence of atherosclerosis, coronary artery stenosis and subsequent myocardial infarction, thoracic aortic aneurysm, valvular heart disease and heart failure. An accumulation of senescence and increased inflammation, caused by the senescence-associated secretory phenotype, have been implicated in the aetiology and progression of these age-associated diseases. Recently it has been demonstrated that compounds targeting components of anti-apoptotic pathways expressed by senescent cells can preferentially induce senescence cells to apoptosis and have been termed senolytics. In this review, we discuss the evidence demonstrating that senescence contributes to cardiovascular disease, with a particular focus on studies that indicate the promise of senotherapy. Based on these data we suggest novel indications for senolytics as a treatment of cardiovascular diseases which have yet to be studied in the context of senotherapy. Finally, while the potential benefits are encouraging, several complications may result from senolytic treatment. We, therefore, consider these challenges in the context of the cardiovascular system.
Collapse
|
14
|
Pils V, Terlecki-Zaniewicz L, Schosserer M, Grillari J, Lämmermann I. The role of lipid-based signalling in wound healing and senescence. Mech Ageing Dev 2021; 198:111527. [PMID: 34174292 DOI: 10.1016/j.mad.2021.111527] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/28/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
Lipid-based signalling modulates several cellular processes and intercellular communication during wound healing and tissue regeneration. Bioactive lipids include but are not limited to the diverse group of eicosanoids, phospholipids, and extracellular vesicles and mediate the attraction of immune cells, initiation of inflammatory responses, and their resolution. In aged individuals, wound healing and tissue regeneration are greatly impaired, resulting in a delayed healing process and non-healing wounds. Senescent cells accumulate with age in vivo, preferably at sites implicated in age-associated pathologies and their elimination was shown to alleviate many age-associated diseases and disorders. In contrast to these findings, the transient presence of senescent cells in the process of wound healing exerts beneficial effects and limits fibrosis. Hence, clearance of senescent cells during wound healing was repeatedly shown to delay wound closure in vivo. Recent findings established a dysregulated synthesis of eicosanoids, phospholipids and extracellular vesicles as part of the senescent phenotype. This intriguing connection between cellular senescence, lipid-based signalling, and the process of wound healing and tissue regeneration prompts us to compile the current knowledge in this review and propose future directions for investigation.
Collapse
Affiliation(s)
- Vera Pils
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Lucia Terlecki-Zaniewicz
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Markus Schosserer
- Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence - SKINMAGINE, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz and Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Ingo Lämmermann
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.
| |
Collapse
|
15
|
Marquez-Exposito L, Tejedor-Santamaria L, Santos-Sanchez L, Valentijn FA, Cantero-Navarro E, Rayego-Mateos S, Rodrigues-Diez RR, Tejera-Muñoz A, Marchant V, Sanz AB, Ortiz A, Goldschmeding R, Ruiz-Ortega M. Acute Kidney Injury is Aggravated in Aged Mice by the Exacerbation of Proinflammatory Processes. Front Pharmacol 2021; 12:662020. [PMID: 34239439 PMCID: PMC8258347 DOI: 10.3389/fphar.2021.662020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Acute kidney injury (AKI) is more frequent in elderly patients. Mechanisms contributing to AKI (tubular cell death, inflammatory cell infiltration, impaired mitochondrial function, and prolonged cell-cycle arrest) have been linked to cellular senescence, a process implicated in regeneration failure and progression to fibrosis. However, the molecular and pathological basis of the age-related increase in AKI incidence is not completely understood. To explore these mechanisms, experimental AKI was induced by folic acid (FA) administration in young (3-months-old) and old (1-year-old) mice, and kidneys were evaluated in the early phase of AKI, at 48 h. Tubular damage score, KIM-1 expression, the recruitment of infiltrating immune cells (mainly neutrophils and macrophages) and proinflammatory gene expression were higher in AKI kidneys of old than of young mice. Tubular cell death in FA-AKI involves several pathways, such as regulated necrosis and apoptosis. Ferroptosis and necroptosis cell-death pathways were upregulated in old AKI kidneys. In contrast, caspase-3 activation was only found in young but not in old mice. Moreover, the antiapoptotic factor BCL-xL was significantly overexpressed in old, injured kidneys, suggesting an age-related apoptosis suppression. AKI kidneys displayed evidence of cellular senescence, such as increased levels of cyclin dependent kinase inhibitors p16ink4a and p21cip1, and of the DNA damage response marker γH2AX. Furthermore, p21cip1 mRNA expression and nuclear staining for p21cip1 and γH2AX were higher in old than in young FA-AKI mice, as well as the expression of senescence-associated secretory phenotype (SASP) components (Il-6, Tgfb1, Ctgf, and Serpine1). Interestingly, some infiltrating immune cells were p21 or γH2AX positive, suggesting that molecular senescence in the immune cells (“immunosenescence”) are involved in the increased severity of AKI in old mice. In contrast, expression of renal protective factors was dramatically downregulated in old AKI mice, including the antiaging factor Klotho and the mitochondrial biogenesis driver PGC-1α. In conclusion, aging resulted in more severe AKI after the exposure to toxic compounds. This increased toxicity may be related to magnification of proinflammatory-related pathways in older mice, including a switch to a proinflammatory cell death (necroptosis) instead of apoptosis, and overactivation of cellular senescence of resident renal cells and infiltrating inflammatory cells.
Collapse
Affiliation(s)
- Laura Marquez-Exposito
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Lucia Tejedor-Santamaria
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Laura Santos-Sanchez
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Floris A Valentijn
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Elena Cantero-Navarro
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Raul R Rodrigues-Diez
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Antonio Tejera-Muñoz
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Ana B Sanz
- Red de Investigación Renal (REDinREN), Madrid, Spain.,Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Alberto Ortiz
- Red de Investigación Renal (REDinREN), Madrid, Spain.,Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| |
Collapse
|
16
|
Wang WJ, Chen XM, Cai GY. Cellular senescence and the senescence-associated secretory phenotype: Potential therapeutic targets for renal fibrosis. Exp Gerontol 2021; 151:111403. [PMID: 33984448 DOI: 10.1016/j.exger.2021.111403] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/25/2022]
Abstract
Renal fibrosis plays a crucial role in the progression of chronic kidney disease and end-stage renal disease. However, because the aetiology of this pathological process is complex and remains unclear, there is still no effective treatment. Cellular senescence and the senescence-associated secretory phenotype (SASP) have been reported to lead to renal fibrosis. This review first discusses the relationships among cellular senescence, the SASP and renal fibrosis. Then, the key role of the SASP in irreversible renal fibrosis, including fibroblast activation and abnormal extracellular matrix accumulation, is discussed, with the results of studies having indicated that inhibiting cellular senescence and the SASP might be a potential preventive and therapeutic strategy for renal fibrosis. Finally, we summarize promising therapeutic strategies revealed by existing research on senescent cells and the SASP, including emerging interventions targeting the SASP, caloric restriction and mimetics, and novel regeneration therapies with stem cells.
Collapse
Affiliation(s)
- Wen-Juan Wang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiang-Mei Chen
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| | - Guang-Yan Cai
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| |
Collapse
|
17
|
Role and mechanism of TXNIP in ageing-related renal fibrosis. Mech Ageing Dev 2021; 196:111475. [PMID: 33781783 DOI: 10.1016/j.mad.2021.111475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 02/27/2021] [Accepted: 03/22/2021] [Indexed: 11/21/2022]
Abstract
Kidney ageing, which is always accompanied by renal fibrosis, drives the progression of renal fibrosis. Thioredoxin-interacting protein (TXNIP) is an endogenous suppressor of the reactive oxygen species-scavenging protein thioredoxin, which has been implicated in the ageing of some organs and is involved in renal fibrosis. However, the expression of TXNIP in ageing kidneys has not been examined, and the relationship between TXNIP and ageing-related renal fibrosis is unclear. We found that TXNIP expression was upregulated in aged mouse kidneys, and this upregulation was accompanied by ageing-related renal fibrosis phenotypes. We demonstrated that the ageing biomarkers were downregulated in TXNIP-knockout mice, and these effects resulted in the alleviation of renal fibrosis and impairments in kidney function. TXNIP overexpression in tubular cells upregulated senescence markers, promoted a profibrotic response and activated STAT3 signalling, and these parameters were inhibited by the silencing of TXNIP. Similarly, the TXNIP-mediated profibrotic response was significantly suppressed by a STAT3 inhibitor. By coimmunoprecipitation, we verified that TXNIP directly bound to STAT3, which suggested that TXNIP exacerbates renal tubular epithelial fibrosis by activating the STAT3 pathway. In summary, TXNIP plays an important role in age-related renal fibrosis and might be a therapeutic target for preventing ageing-associated renal fibrosis.
Collapse
|
18
|
Abstract
Interstitial fibrosis with tubule atrophy (IF/TA) is the response to virtually any sustained kidney injury and correlates inversely with kidney function and allograft survival. IF/TA is driven by various pathways that include hypoxia, renin-angiotensin-aldosterone system, transforming growth factor (TGF)-β signaling, cellular rejection, inflammation and others. In this review we will focus on key pathways in the progress of renal fibrosis, diagnosis and therapy of allograft fibrosis. This review discusses the role and origin of myofibroblasts as matrix producing cells and therapeutic targets in renal fibrosis with a particular focus on renal allografts. We summarize current trends to use multi-omic approaches to identify new biomarkers for IF/TA detection and to predict allograft survival. Furthermore, we review current imaging strategies that might help to identify and follow-up IF/TA complementary or as alternative to invasive biopsies. We further discuss current clinical trials and therapeutic strategies to treat kidney fibrosis.Supplemental Visual Abstract; http://links.lww.com/TP/C141.
Collapse
|
19
|
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: 60] [Impact Index Per Article: 12.0] [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
|
20
|
Santin Y, Lluel P, Rischmann P, Gamé X, Mialet-Perez J, Parini A. Cellular Senescence in Renal and Urinary Tract Disorders. Cells 2020; 9:cells9112420. [PMID: 33167349 PMCID: PMC7694377 DOI: 10.3390/cells9112420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
Cellular senescence is a state of cell cycle arrest induced by repetitive cell mitoses or different stresses, which is implicated in various physiological or pathological processes. The beneficial or adverse effects of senescent cells depend on their transitory or persistent state. Transient senescence has major beneficial roles promoting successful post-injury repair and inhibiting malignant transformation. On the other hand, persistent accumulation of senescent cells has been associated with chronic diseases and age-related illnesses like renal/urinary tract disorders. The deleterious effects of persistent senescent cells have been related, in part, to their senescence-associated secretory phenotype (SASP) characterized by the release of a variety of factors responsible for chronic inflammation, extracellular matrix adverse remodeling, and fibrosis. Recently, an increase in senescent cell burden has been reported in renal, prostate, and bladder disorders. In this review, we will summarize the molecular mechanisms of senescence and their implication in renal and urinary tract diseases. We will also discuss the differential impacts of transient versus persistent status of cellular senescence, as well as the therapeutic potential of senescent cell targeting in these diseases.
Collapse
Affiliation(s)
- Yohan Santin
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1048—I2MC, 31432 Toulouse, France; (Y.S.); (J.M.-P.)
| | - Philippe Lluel
- Urosphere SAS, Rue des Satellites, 31400 Toulouse, France;
| | - Pascal Rischmann
- Department of Urology, Kidney Transplantation and Andrology, Toulouse Rangueil University Hospital, 31432 Toulouse, France; (P.R.); (X.G.)
| | - Xavier Gamé
- Department of Urology, Kidney Transplantation and Andrology, Toulouse Rangueil University Hospital, 31432 Toulouse, France; (P.R.); (X.G.)
| | - Jeanne Mialet-Perez
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1048—I2MC, 31432 Toulouse, France; (Y.S.); (J.M.-P.)
| | - Angelo Parini
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1048—I2MC, 31432 Toulouse, France; (Y.S.); (J.M.-P.)
- Correspondence: ; Tel.: +33-561325601
| |
Collapse
|
21
|
Complement component C5a induces aberrant epigenetic modifications in renal tubular epithelial cells accelerating senescence by Wnt4/βcatenin signaling after ischemia/reperfusion injury. Aging (Albany NY) 2020; 11:4382-4406. [PMID: 31284268 PMCID: PMC6660044 DOI: 10.18632/aging.102059] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/24/2019] [Indexed: 12/30/2022]
Abstract
Epigenetic mechanisms, such as DNA methylation, affect tubular maladaptive response after Acute Kidney Injury (AKI) and accelerate renal aging. Upon ischemia/reperfusion (I/R) injury, Complement activation leads to C5a release that mediates damage; however, little is known about the effect of C5a-C5a Receptor (C5aR) interaction in Renal Tubular Epithelial Cells (RTEC). Through a whole-genome DNA methylation analysis in cultured RTEC, we found that C5a induced aberrant methylation, particularly in regions involved in cell cycle control, DNA damage and Wnt signaling. The most represented genes were BCL9, CYP1B1 and CDK6. C5a stimulation of RTEC led to up-regulation of SA-β Gal and cell cycle arrest markers such as p53 and p21. C5a increased also IL-6, MCP-1 and CTGF gene expression, consistent with SASP development. In accordance, in a swine model of renal I/R injury, we found the increased expression of Wnt4 and βcatenin correlating with SA-β Gal, p21, p16 and IL-6 positivity. Administration of Complement Inhibitor (C1-Inh), antagonized SASP by reducing SA-β Gal, p21, p16, IL-6 and abrogating Wnt4/βcatenin activation. Thus, C5a affects the DNA methylation of genes involved in tubular senescence. Targeting epigenetic programs and Complement may offer novels strategies to protect tubular cells from accelerated aging and to counteract progression to Chronic Kidney Disease
Collapse
|
22
|
Sumida K, Han Z, Dashputre AA, Potukuchi PK, Kovesdy CP. Association between Nrf2 and CDKN2A expression in patients with end-stage renal disease: a pilot study. Aging (Albany NY) 2020; 12:16357-16367. [PMID: 32661200 PMCID: PMC7485736 DOI: 10.18632/aging.103685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/29/2020] [Indexed: 04/12/2023]
Abstract
Patients with end-stage renal disease (ESRD) display phenotypic features of premature biological aging, characterized by disproportionately high morbidity and mortality at a younger age. Nuclear factor erythroid 2-related factor 2 (Nrf2) activity, a master regulator of antioxidative responses, declines with age and is implicated in the pathogenesis of age-related disorders; however, little is known about the association between Nrf2 and premature biological aging in ESRD patients. In a cross-sectional pilot cohort of 34 ESRD patients receiving maintenance hemodialysis, we measured the expression of Nrf2 and cyclin-dependent kinase inhibitor 2A (CDKN2A, or p16INK4a, a biomarker of biological aging) genes in whole blood and examined the association of Nrf2 with CDKN2A expression, using Spearman's rank correlation and multivariable linear regression models with adjustment for potential confounders. There was a significant negative correlation between Nrf2 and CDKN2A expression (rho=-0.51, P=0.002); while no significant correlation was found between Nrf2 expression and chronological age (rho=-0.02, P=0.91). After multivariable adjustment, Nrf2 expression remained significantly and negatively associated with CDKN2A expression (β coefficient=-1.51, P=0.01), independent of chronological age, gender, race, and diabetes status. These findings suggest a potential contribution of Nrf2 dysfunction to the development of premature biological aging and its related morbidities in ESRD patients.
Collapse
Affiliation(s)
- Keiichi Sumida
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zhongji Han
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ankur A. Dashputre
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Institute for Health Outcomes and Policy, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Praveen K. Potukuchi
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Csaba P. Kovesdy
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Nephrology Section, Memphis VA Medical Center, Memphis, TN 38104, USA
| |
Collapse
|
23
|
Franzin R, Stasi A, Fiorentino M, Stallone G, Cantaluppi V, Gesualdo L, Castellano G. Inflammaging and Complement System: A Link Between Acute Kidney Injury and Chronic Graft Damage. Front Immunol 2020; 11:734. [PMID: 32457738 PMCID: PMC7221190 DOI: 10.3389/fimmu.2020.00734] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The aberrant activation of complement system in several kidney diseases suggests that this pillar of innate immunity has a critical role in the pathophysiology of renal damage of different etiologies. A growing body of experimental evidence indicates that complement activation contributes to the pathogenesis of acute kidney injury (AKI) such as delayed graft function (DGF) in transplant patients. AKI is characterized by the rapid loss of the kidney's excretory function and is a complex syndrome currently lacking a specific medical treatment to arrest or attenuate progression in chronic kidney disease (CKD). Recent evidence suggests that independently from the initial trigger (i.e., sepsis or ischemia/reperfusions injury), an episode of AKI is strongly associated with an increased risk of subsequent CKD. The AKI-to-CKD transition may involve a wide range of mechanisms including scar-forming myofibroblasts generated from different sources, microvascular rarefaction, mitochondrial dysfunction, or cell cycle arrest by the involvement of epigenetic, gene, and protein alterations leading to common final signaling pathways [i.e., transforming growth factor beta (TGF-β), p16 ink4a , Wnt/β-catenin pathway] involved in renal aging. Research in recent years has revealed that several stressors or complications such as rejection after renal transplantation can lead to accelerated renal aging with detrimental effects with the establishment of chronic proinflammatory cellular phenotypes within the kidney. Despite a greater understanding of these mechanisms, the role of complement system in the context of the AKI-to-CKD transition and renal inflammaging is still poorly explored. The purpose of this review is to summarize recent findings describing the role of complement in AKI-to-CKD transition. We will also address how and when complement inhibitors might be used to prevent AKI and CKD progression, therefore improving graft function.
Collapse
Affiliation(s)
- Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
- Department Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Marco Fiorentino
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vincenzo Cantaluppi
- Department Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
24
|
Kidney allograft fibrosis: what we learned from latest translational research studies. J Nephrol 2020; 33:1201-1211. [PMID: 32193834 DOI: 10.1007/s40620-020-00726-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023]
Abstract
To add new molecular and pathogenetic insights into the biological machinery associated to kidney allograft fibrosis is a major research target in nephrology and organ transplant translational medicine. Interstitial fibrosis associated to tubular atrophy (IF/TA) is, in fact, an inevitable and progressive process that occurs in almost every type of chronic allograft injury (particularly in grafts from expanded criteria donors) characterized by profound remodeling and excessive production/deposition of fibrillar extracellular matrix (ECM) with a great clinical impact. IF/TA is detectable in more than 50% of kidney allografts at 2 years. However, although well studied, the complete cellular/biological network associated with IF/TA is only partially evaluated. In the last few years, then, thanks to the introduction of new biomolecular technologies, inflammation in scarred/fibrotic parenchyma areas (recently acknowledged by the BANFF classification) has been recognized as a pivotal element able to accelerate the onset and development of the allograft chronic damage. Therefore, in this review, we focused on some new pathogenetic elements involved in graft fibrosis (including epithelial/endothelial to mesenchymal transition, oxidative stress, activation of Wnt and Hedgehog signaling pathways, fatty acids oxidation and cellular senescence) that, in our opinion, could become in future good candidates as potential biomarkers and therapeutic targets.
Collapse
|
25
|
Odeh A, Dronina M, Domankevich V, Shams I, Manov I. Downregulation of the inflammatory network in senescent fibroblasts and aging tissues of the long-lived and cancer-resistant subterranean wild rodent, Spalax. Aging Cell 2020; 19:e13045. [PMID: 31605433 PMCID: PMC6974727 DOI: 10.1111/acel.13045] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/26/2019] [Accepted: 08/31/2019] [Indexed: 12/11/2022] Open
Abstract
The blind mole rat (Spalax) is a wild, long‐lived rodent that has evolved mechanisms to tolerate hypoxia and resist cancer. Previously, we demonstrated high DNA repair capacity and low DNA damage in Spalax fibroblasts following genotoxic stress compared with rats. Since the acquisition of senescence‐associated secretory phenotype (SASP) is a consequence of persistent DNA damage, we investigated whether cellular senescence in Spalax is accompanied by an inflammatory response. Spalax fibroblasts undergo replicative senescence (RS) and etoposide‐induced senescence (EIS), evidenced by an increased activity of senescence‐associated beta‐galactosidase (SA‐β‐Gal), growth arrest, and overexpression of p21, p16, and p53 mRNAs. Yet, unlike mouse and human fibroblasts, RS and EIS Spalax cells showed undetectable or decreased expression of the well‐known SASP factors: interleukin‐6 (IL6), IL8, IL1α, growth‐related oncogene alpha (GROα), SerpinB2, and intercellular adhesion molecule (ICAM‐1). Apparently, due to the efficient DNA repair in Spalax, senescent cells did not accumulate the DNA damage necessary for SASP activation. Conversely, Spalax can maintain DNA integrity during replicative or moderate genotoxic stress and limit pro‐inflammatory secretion. However, exposure to the conditioned medium of breast cancer cells MDA‐MB‐231 resulted in an increase in DNA damage, activation of the nuclear factor κB (NF‐κB) through nuclear translocation, and expression of inflammatory mediators in RS Spalax cells. Evaluation of SASP in aging Spalax brain and intestine confirmed downregulation of inflammatory‐related genes. These findings suggest a natural mechanism for alleviating the inflammatory response during cellular senescence and aging in Spalax, which can prevent age‐related chronic inflammation supporting healthy aging and longevity.
Collapse
Affiliation(s)
- Amani Odeh
- Department of Evolutionary and Environmental Biology Faculty of Natural Sciences University of Haifa Haifa Israel
| | - Maria Dronina
- Institute of Evolution University of Haifa Haifa Israel
| | - Vered Domankevich
- Department of Evolutionary and Environmental Biology Faculty of Natural Sciences University of Haifa Haifa Israel
| | - Imad Shams
- Department of Evolutionary and Environmental Biology Faculty of Natural Sciences University of Haifa Haifa Israel
- Institute of Evolution University of Haifa Haifa Israel
| | - Irena Manov
- Institute of Evolution University of Haifa Haifa Israel
| |
Collapse
|
26
|
Knoppert SN, Valentijn FA, Nguyen TQ, Goldschmeding R, Falke LL. Cellular Senescence and the Kidney: Potential Therapeutic Targets and Tools. Front Pharmacol 2019; 10:770. [PMID: 31354486 PMCID: PMC6639430 DOI: 10.3389/fphar.2019.00770] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasing health burden (affecting approximately 13.4% of the population). Currently, no curative treatment options are available and treatment is focused on limiting the disease progression. The accumulation of senescent cells has been implicated in the development of kidney fibrosis by limiting tissue rejuvenation and through the secretion of pro-fibrotic and pro-inflammatory mediators termed as the senescence-associated secretory phenotype. The clearance of senescent cells in aging models results in improved kidney function, which shows promise for the options of targeting senescent cells in CKD. There are several approaches for the development of “senotherapies”, the most rigorous of which is the elimination of senescent cells by the so-called senolytic drugs either newly developed or repurposed for off-target effects in terms of selectively inducing apoptosis in senescent cells. Several chemotherapeutics and checkpoint inhibitors currently used in daily oncological practice show senolytic properties. However, the applicability of such senolytic compounds for the treatment of renal diseases has hardly been investigated. A serious concern is that systemic side effects will limit the use of senolytics for kidney fibrosis. Specifically targeting senescent cells and/or targeted drug delivery to the kidney might circumvent these side effects. In this review, we discuss the connection between CKD and senescence, the pharmacological options for targeting senescent cells, and the means to specifically target the kidney.
Collapse
Affiliation(s)
- Sebastian N Knoppert
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Floris A Valentijn
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Internal Medicine, Diakonessenhuis, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
27
|
Dai L, Qureshi AR, Witasp A, Lindholm B, Stenvinkel P. Early Vascular Ageing and Cellular Senescence in Chronic Kidney Disease. Comput Struct Biotechnol J 2019; 17:721-729. [PMID: 31303976 PMCID: PMC6603301 DOI: 10.1016/j.csbj.2019.06.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/08/2019] [Accepted: 06/11/2019] [Indexed: 01/08/2023] Open
Abstract
Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by progressive vascular disease, systemic inflammation, muscle wasting and frailty. The predominant early vascular ageing (EVA) process mediated by medial vascular calcification (VC) results in a marked discrepancy between chronological and biological vascular age in CKD. Though the exact underlying mechanisms of VC and EVA are not fully elucidated, accumulating evidence indicates that cellular senescence - and subsequent chronic inflammation through the senescence-associated secretary phenotype (SASP) - plays a fundamental role in its initiation and progression. In this review, we discuss the pathophysiological links between senescence and the EVA process in CKD, with focus on cellular senescence and media VC, and potential anti-ageing therapeutic strategies of senolytic drugs targeting cellular senescence and EVA in CKD.
Collapse
Affiliation(s)
| | | | | | | | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| |
Collapse
|
28
|
Wei SY, Pan SY, Li B, Chen YM, Lin SL. Rejuvenation: Turning back the clock of aging kidney. J Formos Med Assoc 2019; 119:898-906. [PMID: 31202499 DOI: 10.1016/j.jfma.2019.05.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/14/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
Aging is inevitable in life. It is defined as impaired adaptive capacity to environmental or internal stresses with growing rates of disease and death. Aging is also an important risk factor for various kidney diseases such as acute kidney injury and chronic kidney disease. Patients older than 65 years have nearly 28% risk of failing recovery of kidney function when suffering from acute kidney injury. It is reported that more than a third of population aged 65 years and older have chronic kidney disease in Taiwan, and the occurrence of multiple age-related disorders is predicted to increase in parallel. Renal aging is a complex, multifactorial process characterized by many anatomical and functional changes. Several factors are involved in renal aging, such as loss of telomeres, cell cycle arrest, chronic inflammation, activation of renin-angiotensin system, decreased klotho expression, and development of tertiary lymphoid tissues. These changes can also be observed in many other different types of renal injury. Recent studies suggested that young blood may rejuvenate aged organs, including the kidneys. In order to develop new therapeutic strategies for renal aging, the mechanisms underlying renal aging and by which young blood can halt or reverse aging process warrants further study.
Collapse
Affiliation(s)
- Shi-Yao Wei
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Szu-Yu Pan
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Bing Li
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Yung-Ming Chen
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuei-Liong Lin
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan; Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
29
|
Docherty MH, O'Sullivan ED, Bonventre JV, Ferenbach DA. Cellular Senescence in the Kidney. J Am Soc Nephrol 2019; 30:726-736. [PMID: 31000567 DOI: 10.1681/asn.2018121251] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Senescent cells have undergone permanent growth arrest, adopt an altered secretory phenotype, and accumulate in the kidney and other organs with ageing and injury. Senescence has diverse physiologic roles and experimental studies support its importance in nephrogenesis, successful tissue repair, and in opposing malignant transformation. However, recent murine studies have shown that depletion of chronically senescent cells extends healthy lifespan and delays age-associated disease-implicating senescence and the senescence-associated secretory phenotype as drivers of organ dysfunction. Great interest is therefore focused on the manipulation of senescence as a novel therapeutic target in kidney disease. In this review, we examine current knowledge and areas of ongoing uncertainty regarding senescence in the human kidney and experimental models. We summarize evidence supporting the role of senescence in normal kidney development and homeostasis but also senescence-induced maladaptive repair, renal fibrosis, and transplant failure. Recent studies using senescent cell manipulation and depletion as novel therapies to treat renal disease are discussed, and we explore unanswered questions for future research.
Collapse
Affiliation(s)
| | - Eoin D O'Sullivan
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; and
| | - Joseph V Bonventre
- Renal Division and Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - David A Ferenbach
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK; .,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; and
| |
Collapse
|
30
|
Johansen KL. Metrics of Aging in Transplantation. CURRENT TRANSPLANTATION REPORTS 2019. [DOI: 10.1007/s40472-019-0229-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
31
|
McGuinness D, Mohammed S, Monaghan L, Wilson PA, Kingsmore DB, Shapter O, Stevenson KS, Coley SM, Devey L, Kirkpatrick RB, Shiels PG. A molecular signature for delayed graft function. Aging Cell 2018; 17:e12825. [PMID: 30094915 PMCID: PMC6156499 DOI: 10.1111/acel.12825] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/08/2018] [Accepted: 07/02/2018] [Indexed: 01/01/2023] Open
Abstract
Chronic kidney disease and associated comorbidities (diabetes, cardiovascular diseases) manifest with an accelerated ageing phenotype, leading ultimately to organ failure and renal replacement therapy. This process can be modulated by epigenetic and environmental factors which promote loss of physiological function and resilience to stress earlier, linking biological age with adverse outcomes post-transplantation including delayed graft function (DGF). The molecular features underpinning this have yet to be fully elucidated. We have determined a molecular signature for loss of resilience and impaired physiological function, via a synchronous genome, transcriptome and proteome snapshot, using human renal allografts as a source of healthy tissue as an in vivo model of ageing in humans. This comprises 42 specific transcripts, related through IFNγ signalling, which in allografts displaying clinically impaired physiological function (DGF) exhibited a greater magnitude of change in transcriptional amplitude and elevated expression of noncoding RNAs and pseudogenes, consistent with increased allostatic load. This was accompanied by increased DNA methylation within the promoter and intragenic regions of the DGF panel in preperfusion allografts with immediate graft function. Pathway analysis indicated that an inability to sufficiently resolve inflammatory responses was enabled by decreased resilience to stress and resulted in impaired physiological function in biologically older allografts. Cross-comparison with publically available data sets for renal pathologies identified significant transcriptional commonality for over 20 DGF transcripts. Our data are clinically relevant and important, as they provide a clear molecular signature for the burden of "wear and tear" within the kidney and thus age-related physiological capability and resilience.
Collapse
Affiliation(s)
- Dagmara McGuinness
- Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences; University of Glasgow; Glasgow UK
| | - Suhaib Mohammed
- Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences; University of Glasgow; Glasgow UK
| | - Laura Monaghan
- Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences; University of Glasgow; Glasgow UK
| | - Paul A. Wilson
- Computational Biology; GlaxoSmithKline Medicines Research Centre; Stevenage UK
| | - David B. Kingsmore
- Renal Transplant Unit, NHS Greater Glasgow and Clyde; South Glasgow University Hospital; Glasgow UK
| | - Oliver Shapter
- Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences; University of Glasgow; Glasgow UK
- Renal Transplant Unit, NHS Greater Glasgow and Clyde; South Glasgow University Hospital; Glasgow UK
| | - Karen S. Stevenson
- Renal Transplant Unit, NHS Greater Glasgow and Clyde; South Glasgow University Hospital; Glasgow UK
| | - Shana M. Coley
- Research Institute of Infection Immunity and Inflammation, College of Medical, Veterinary & Life Sciences; University of Glasgow; Glasgow UK
| | - Luke Devey
- Metabolic Pathways Cardio Therapy Area Unit; GlaxoSmithKline; King of Prussia Pennsylvania
| | | | - Paul G. Shiels
- Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences; University of Glasgow; Glasgow UK
| |
Collapse
|
32
|
van Willigenburg H, de Keizer PLJ, de Bruin RWF. Cellular senescence as a therapeutic target to improve renal transplantation outcome. Pharmacol Res 2018; 130:322-330. [PMID: 29471104 DOI: 10.1016/j.phrs.2018.02.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/02/2018] [Accepted: 02/12/2018] [Indexed: 01/18/2023]
Abstract
Kidney transplants from aged donors are more vulnerable to ischemic injury, suffer more from delayed graft function and have a lower graft survival compared to kidneys from younger donors. On a cellular level, aging results in an increase in cells that are in a permanent cell cycle arrest, termed senescence, which secrete a range of pro-inflammatory cytokines and growth factors. Consequently, these senescent cells negatively influence the local milieu by causing inflammaging, and by reducing the regenerative capacity of the kidney. Moreover, the oxidative damage that is inflicted by ischemia-reperfusion injury during transplantation can induce senescence and accelerate aging. In this review, we describe recent developments in the understanding of the biology of aging that have led to the development of a new class of therapeutic agents aimed at eliminating senescent cells. These compounds have already shown to be able to restore tissue homeostasis in old mice, improve kidney function and general health- and lifespan. Use of these anti-senescence compounds holds great promise to improve the quality of marginal donor kidneys as well as to remove senescent cells induced by ischemia-reperfusion injury. Altogether, senescent cell removal may increase the donor pool, relieving the growing organ shortage and improve long-term transplantation outcome.
Collapse
Affiliation(s)
- Hester van Willigenburg
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Peter L J de Keizer
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Ron W F de Bruin
- Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
33
|
Wiley CD, Schaum N, Alimirah F, Lopez-Dominguez JA, Orjalo AV, Scott G, Desprez PY, Benz C, Davalos AR, Campisi J. Small-molecule MDM2 antagonists attenuate the senescence-associated secretory phenotype. Sci Rep 2018; 8:2410. [PMID: 29402901 PMCID: PMC5799282 DOI: 10.1038/s41598-018-20000-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/10/2018] [Indexed: 01/07/2023] Open
Abstract
Processes that have been linked to aging and cancer include an inflammatory milieu driven by senescent cells. Senescent cells lose the ability to divide, essentially irreversibly, and secrete numerous proteases, cytokines and growth factors, termed the senescence-associated secretory phenotype (SASP). Senescent cells that lack p53 tumor suppressor function show an exaggerated SASP, suggesting the SASP is negatively controlled by p53. Here, we show that increased p53 activity caused by small molecule inhibitors of MDM2, which promotes p53 degradation, reduces inflammatory cytokine production by senescent cells. Upon treatment with the MDM2 inhibitors nutlin-3a or MI-63, human cells acquired a senescence-like growth arrest, but the arrest was reversible. Importantly, the inhibitors reduced expression of the signature SASP factors IL-6 and IL-1α by cells made senescent by genotoxic stimuli, and suppressed the ability of senescent fibroblasts to stimulate breast cancer cell aggressiveness. Our findings suggest that MDM2 inhibitors could reduce cancer progression in part by reducing the pro-inflammatory environment created by senescent cells.
Collapse
Affiliation(s)
- Christopher D Wiley
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Nicholas Schaum
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, School of Medicine, 1265 Welch Road, Stanford, CA, 94305, USA
| | - Fatouma Alimirah
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | | | - Arturo V Orjalo
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Gary Scott
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Pierre-Yves Desprez
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA.,California Pacific Medical Center, Research Institute, 475 Brannan Street, San Francisco, CA, 94107, USA
| | - Christopher Benz
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Albert R Davalos
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA.
| | - Judith Campisi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA. .,Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA, 94720, USA.
| |
Collapse
|
34
|
Abstract
PURPOSE OF THE REVIEW Senescent cells have the capacity to both effect and limit fibrosis. Senotherapeutics target senescent cells to improve aging conditions. Here, we review the contexts in which senescent cells mediate wound healing and fibrotic pathology and the potential utility of senotherapeutic drugs for treatment of fibrotic disease. RECENT FINDINGS Multi-action and temporal considerations influence deleterious versus beneficial actions of senescent cells. Acutely generated senescent cells can limit proliferation, and the senescence-associated secretory phenotype (SASP) contains factors that can facilitate tissue repair. Long-lived senescent cells that evade clearance or are generated outside of programmed remodeling can deplete the progenitor pool to exhaust regenerative capacity and through the SASP, stimulate continual activation, leading to disorganized tissue architecture, fibrotic damage, sterile inflammation, and induction of bystander senescence. Senescent cells contribute to fibrotic pathogenesis in multiple tissues, including the liver, kidney, and lung. Senotherapeutics may be a viable strategy for treatment of a range of fibrotic conditions.
Collapse
|
35
|
Sofue T, Kushida Y, Ozaki T, Moritoki M, Nishijima Y, Ohsaki H, Ueda N, Kakehi Y, Nishiyama A, Minamino T. Tubular Cell Senescence in the Donated Kidney Predicts Allograft Function, but Not Donor Remnant Kidney Function, in Living Donor Kidney Transplantation. Am J Nephrol 2017; 47:8-17. [PMID: 29275400 DOI: 10.1159/000485845] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/28/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND It is uncertain whether kidneys from marginal donors are suitable for live kidney transplantation. In deceased donor kidneys, tubular cell senescence affects allograft function. However, the degree of cell senescence in a living donor kidney with marginal factors has not been reported. In this study, we assessed the association of tubular cell senescence with allograft and remnant kidney function by a prospective observational clinical study. METHODS Thirty-eight living donor kidney transplantations were analyzed prospectively. Tissue sections obtained from preimplantation kidney biopsies were immunostained for p16INK4a to indicate cell senescence. Various kidney biomarkers were analyzed in urine and blood samples. RESULTS Of the 38 donors, 21 had marginal factors. Severe tubular senescence was found in living donors with overlapping marginal criteria. Tubular senescence in living donor kidneys was significantly related to donor age and lower recipient kidney function at 1 year after transplantation independently of donor age (β = -0.281; p = 0.050) but did not affect remnant kidney function after donation. Pretransplantation donor pre-estimated glomerular filtration rate and hypertension did not show a significant area under the curve (AUC) for prediction of high tubular senescence. High plasma levels of soluble αKlotho were associated with a higher predictive value for low tubular cell senescence with an AUC of 0.78 (95% CI 0.62-0.93; p < 0.01). CONCLUSIONS The nuclear p16-staining rate in donated kidney tubules is a predictor for allograft kidney function but not donor remnant kidney function. Detection of tubular cell senescence may facilitate selection of appropriate living donor candidates.
Collapse
Affiliation(s)
- Tadashi Sofue
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa, Japan
| | - Yoshio Kushida
- Department of Pathology, Kagawa University, Kagawa, Japan
| | - Taro Ozaki
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa, Japan
| | - Masahiro Moritoki
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa, Japan
| | - Yoko Nishijima
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa, Japan
| | - Hiroyuki Ohsaki
- Department of Medical Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Nobufumi Ueda
- Department of Urology, Kagawa University, Kagawa, Japan
| | | | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Tetsuo Minamino
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
36
|
Cuna V, Comai G, Cappuccilli M, Baraldi O, Capelli I, De Liberali M, Gasperoni L, Conte D, Ravaioli M, Pinna AD, La Manna G. Fifteen-Year Analysis of Deceased Kidney Donation: A Single Transplant Center Experience in a Region of Northern Italy. Med Sci Monit 2017; 23:4482-4489. [PMID: 28919594 PMCID: PMC5616149 DOI: 10.12659/msm.903513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background The rising number of patients on waiting lists for kidney transplant and the shortage of available organs has intensified efforts to increase the number of potential donors. Material/Methods This study investigated changes in clinical parameters among potential deceased donors in the 15-year period between 1999 and 2013 and their impact on transplantation procedure and outcomes. A total of 1634 potential deceased donors were examined and divided into 2 groups: 707 of them identified from 1999 to 2005 (Group A), and 927 from 2006 to 2013 (Group B). Results The comparison between the potential donors in Group A vs. Group B revealed an increase over time in donor age (54.6±17.2 vs. 58.8±16.3, p<0.001), a reduction in the percentage of standard donors (52.3% vs. 39.8%, p<0.001), a broader utilization of organs from expanded criteria donors, and a greater number of comorbidities, particularly cardiovascular disease and dyslipidemia. However, renal function parameters and the bioptic scores did not change significantly over the years. Conclusions These results suggest the usefulness of strategies to increase the number of potential donors suitable for organ donation, especially among elderly and marginal donors.
Collapse
Affiliation(s)
- Vania Cuna
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) - Nephrology, Dialysis, and Transplantation Unit, University of Bologna, Bologna, Italy
| | - Giorgia Comai
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) - Nephrology, Dialysis, and Transplantation Unit, University of Bologna, Bologna, Italy
| | - Maria Cappuccilli
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) - Nephrology, Dialysis, and Transplantation Unit, University of Bologna, Bologna, Italy
| | - Olga Baraldi
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) - Nephrology, Dialysis, and Transplantation Unit, University of Bologna, Bologna, Italy
| | - Irene Capelli
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) - Nephrology, Dialysis, and Transplantation Unit, University of Bologna, Bologna, Italy
| | - Matteo De Liberali
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) - Nephrology, Dialysis, and Transplantation Unit, University of Bologna, Bologna, Italy
| | - Lorenzo Gasperoni
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) - Nephrology, Dialysis, and Transplantation Unit, University of Bologna, Bologna, Italy
| | - Diletta Conte
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) - Nephrology, Dialysis, and Transplantation Unit, Universita degli Studi di Bologna, Bologna, Italy
| | - Matteo Ravaioli
- Department of General Surgery and Transplantation, University of Bologna, Bologna, Italy
| | - Antonio D Pinna
- Department of General Surgery and Transplantation, University of Bologna, Bologna, Italy
| | - Gaetano La Manna
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) - Nephrology, Dialysis, and Transplantation Unit, University of Bologna, Bologna, Italy
| |
Collapse
|
37
|
Schmitt R, Melk A. Molecular mechanisms of renal aging. Kidney Int 2017; 92:569-579. [DOI: 10.1016/j.kint.2017.02.036] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/05/2017] [Accepted: 02/14/2017] [Indexed: 12/31/2022]
|
38
|
Shiels PG, McGuinness D, Eriksson M, Kooman JP, Stenvinkel P. The role of epigenetics in renal ageing. Nat Rev Nephrol 2017. [PMID: 28626222 DOI: 10.1038/nrneph.2017.78] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
An ability to separate natural ageing processes from processes specific to morbidities is required to understand the heterogeneity of age-related organ dysfunction. Mechanistic insight into how epigenetic factors regulate ageing throughout the life course, linked to a decline in renal function with ageing, is already proving to be of value in the analyses of clinical and epidemiological cohorts. Noncoding RNAs provide epigenetic regulatory circuits within the kidney, which reciprocally interact with DNA methylation processes, histone modification and chromatin. These interactions have been demonstrated to reflect the biological age and function of renal allografts. Epigenetic factors control gene expression and activity in response to environmental perturbations. They also have roles in highly conserved signalling pathways that modulate ageing, including the mTOR and insulin/insulin-like growth factor signalling pathways, and regulation of sirtuin activity. Nutrition, the gut microbiota, inflammation and environmental factors, including psychosocial and lifestyle stresses, provide potential mechanistic links between the epigenetic landscape of ageing and renal dysfunction. Approaches to modify the renal epigenome via nutritional intervention, targeting the methylome or targeting chromatin seem eminently feasible, although caution is merited owing to the potential for intergenerational and transgenerational effects.
Collapse
Affiliation(s)
- Paul G Shiels
- Section of Epigenetics, Institute of Cancer Sciences, Wolfson Wohl Translational Research Centre, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1QH, UK
| | - Dagmara McGuinness
- Section of Epigenetics, Institute of Cancer Sciences, Wolfson Wohl Translational Research Centre, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1QH, UK
| | - Maria Eriksson
- Department of Biosciences and Nutrition (BioNut), H2, Eriksson, Novum 141, 83 Huddinge, Sweden
| | - Jeroen P Kooman
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, PO Box 5800, 6202 AZ Maastrich, Netherlands
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Huddinge, Karolinska Institutet, SE-14157 Stockholm, Sweden
| |
Collapse
|
39
|
Susnik N, Sen P, Melk A, Schmitt R. Aging, Cellular Senescence, and Kidney Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0143-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
40
|
Senotherapy: growing old and staying young? Pflugers Arch 2017; 469:1051-1059. [PMID: 28389776 DOI: 10.1007/s00424-017-1972-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/29/2017] [Indexed: 12/16/2022]
Abstract
Cellular senescence, which has been linked to age-related diseases, occurs during normal aging or as a result of pathological cell stress. Due to their incapacity to proliferate, senescent cells cannot contribute to normal tissue maintenance and tissue repair. Instead, senescent cells disturb the microenvironment by secreting a plethora of bioactive factors that may lead to inflammation, regenerative dysfunction and tumor progression. Recent understanding of stimuli and pathways that induce and maintain cellular senescence offers the possibility to selectively eliminate senescent cells. This novel strategy, which so far has not been tested in humans, has been coined senotherapy or senolysis. In mice, senotherapy proofed to be effective in models of accelerated aging and also during normal chronological aging. Senotherapy prolonged lifespan, rejuvenated the function of bone marrow, muscle and skin progenitor cells, improved vasomotor function and slowed down atherosclerosis progression. While initial studies used genetic approaches for the killing of senescent cells, recent approaches showed similar effects with senolytic drugs. These observations open up exciting possibilities with a great potential for clinical development. However, before the integration of senotherapy into patient care can be considered, we need further research to improve our insight into the safety and efficacy of this strategy during short- and long-term use.
Collapse
|
41
|
Salvadori M, Tsalouchos A. Pre-transplant biomarkers and prediction of post-transplant outcomes in kidney transplantation. J Renal Inj Prev 2017; 6:222-230. [DOI: 10.15171/jrip.2017.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
|
42
|
Shiels PG, Stenvinkel P, Kooman JP, McGuinness D. Circulating markers of ageing and allostatic load: A slow train coming. Pract Lab Med 2017; 7:49-54. [PMID: 28856219 PMCID: PMC5574864 DOI: 10.1016/j.plabm.2016.04.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 04/15/2016] [Indexed: 12/15/2022] Open
Abstract
Dealing with the growing burden of age-related morbidities is one of the greatest challenges facing modern society. How we age across the lifecourse and how psychosocial and lifestyle factors interplay with the biology of ageing remains to be fully elucidated. Sensitive and specific biomarkers with which to interrogate the biology of the ageing process are sparse. Recent evidence suggests that non-coding RNAs are key determinants of such processes and that these can be used as potential circulatory bio-markers of ageing. They may also provide a mechanism which mediates the spread of allostatic load across the body over time, ultimately reflecting the immunological health and physiological status of tissues and organs. The interplay between exosomal microRNAs and ageing processes is still relatively unexplored, although circulating microRNAs have been linked to the regulation of a range of physiological and pathological processes and offer insight into mechanistic determinants of healthspan.
Collapse
Affiliation(s)
- Paul G. Shiels
- University of Glasgow, Institute of Cancer Sciences, Wolfson-Wohl Translational Cancer Research Centre, Glasgow, UK
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Huddinge, Sweden
| | - Jeroen P. Kooman
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Dagmara McGuinness
- University of Glasgow, Institute of Cancer Sciences, Wolfson-Wohl Translational Cancer Research Centre, Glasgow, UK
| |
Collapse
|
43
|
Identification of the activating cytotoxicity receptor NKG2D as a senescence marker in zero-hour kidney biopsies is indicative for clinical outcome. Kidney Int 2017; 91:1447-1463. [PMID: 28233611 DOI: 10.1016/j.kint.2016.12.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 12/09/2016] [Accepted: 12/15/2016] [Indexed: 11/21/2022]
Abstract
The definition of biological donor organ age rather than chronological age seems obvious for the establishment of a valid pre-transplant risk assessment. Therefore, we studied gene expression for candidate markers in 60 zero-hour kidney biopsies. Compared with 29 younger donors under age 55, 31 elderly donors age 55 and older had significant mRNA expression for immunoproteasome subunits (PSMB8, PSMB9 and PSMB10), HLA-DRB, and transcripts of the activating cytotoxicity receptor NKG2D. Gene expression was validated in an independent donor cohort consisting of 37 kidneys from donors 30 years and under (Group I), 75 kidneys from donors age 31-54 years (Group II) and 75 kidneys from donors age 55 and older (Group III). Significant gene induction was confirmed in kidneys from Group III for PSMB9 and PSMB10. Strikingly, transcripts of NKG2D had the significantly highest gene induction in Group III versus Group II and Group I. Similar results were obtained for CDKN2A, but not for telomere length. Both NKG2D and CDKN2A mRNA expression were significantly correlated with creatinine levels at 24 months after transplantation. Univariate regression analysis showed significant predictive power regarding graft function at 6 and 12 months for NKG2D and CDKN2A. However, only NKG2D remained significantly predictive in the multivariate model at 12 months. Thus, our results reveal novel candidate markers in aged renal allografts, which could be helpful in the assessment of organ quality.
Collapse
|
44
|
Murillo-Ortiz B, Martínez-Garza S, Suárez García D, Castillo Valenzuela RDC, García Regalado JF, Cano Velázquez G. Association between telomere length and CYP19 TTTA repetition polymorphism in healthy and breast cancer-diagnosed women. BREAST CANCER-TARGETS AND THERAPY 2017; 9:21-27. [PMID: 28144163 PMCID: PMC5245810 DOI: 10.2147/bctt.s125431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Introduction Several studies have reported an increase in breast cancer (BC) risk when patients are carriers of the CYP19 TTA polymorphism with ≥10 repeats; moreover, it has been reported that telomere length is associated with a higher susceptibility of developing cancer. Objective The objective of this study was to understand the relationship between CYP19 TTTA repetition polymorphism and telomere length and its effects on serum estradiol, estrone and follicle-stimulating hormone (FSH). Materials and methods A total of 180 postmenopausal healthy and 70 BC-diagnosed women were included. Telomere length was determined through real-time quantitative polymerase chain reaction, and aromatase polymorphism was analyzed through DNA; both samples were obtained from circulating leukocytes. Serum estrone, estradiol and FSH were determined by enzyme-linked immunosorbent assay. Results Patients with a BC diagnosis showed >10 repetitions more frequently, compared with that of healthy women (50% vs 23%, χ2 = 11.44, p = 0.0007). A significant difference in telomere length between healthy and BC women was observed (5,042.7 vs 2,256.7 pb, Z = 4.88, p < 0.001). CYP19 TTTA repeat polymorphism was associated with serum levels of estradiol and estrone in both groups, being higher in those with >10 repeats. Moreover, telomere length showed an inverse relationship with the number of repeats of the aromatase polymorphism in healthy women (R2 = 0.04, r = −0.24); in contrast, BC patients did not display this relationship. In addition, telomere length presented an inverse relationship with serum levels of estradiol and estrone in BC patients (p = 0.02). Conclusion Telomere length is shorter in BC patients than in healthy patients. The CYP19 TTTA repeat polymorphism is associated with serum levels of estradiol and estrone in both healthy women and BC patients, being higher in those with polymorphism carriers >10 repeats. Telomere length has an inverse correlation with the number of repeats of the aromatase polymorphism in healthy women but not in BC women. Estradiol and estrone levels in BC women have an inverse relationship with telomere length.
Collapse
Affiliation(s)
- Blanca Murillo-Ortiz
- Institute Mexican of Social Security, Department Oncology, Unit of Research in Clinical Epidemiology
| | - Sandra Martínez-Garza
- Institute Mexican of Social Security, Department Oncology, Unit of Research in Clinical Epidemiology
| | - David Suárez García
- Institute Mexican of Social Security, Department Oncology, Unit of Research in Clinical Epidemiology
| | | | | | - Gerardo Cano Velázquez
- Institute Mexican of Social Security, Department Oncology, Unit of Research in Clinical Epidemiology
| |
Collapse
|
45
|
Sturmlechner I, Durik M, Sieben CJ, Baker DJ, van Deursen JM. Cellular senescence in renal ageing and disease. Nat Rev Nephrol 2016; 13:77-89. [DOI: 10.1038/nrneph.2016.183] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Wood WA, Krishnamurthy J, Mitin N, Torrice C, Parker JS, Snavely AC, Shea TC, Serody JS, Sharpless NE. Chemotherapy and Stem Cell Transplantation Increase p16 INK4a Expression, a Biomarker of T-cell Aging. EBioMedicine 2016; 11:227-238. [PMID: 27591832 PMCID: PMC5049997 DOI: 10.1016/j.ebiom.2016.08.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 08/16/2016] [Accepted: 08/19/2016] [Indexed: 12/13/2022] Open
Abstract
The expression of markers of cellular senescence increases exponentially in multiple tissues with aging. Age-related physiological changes may contribute to adverse outcomes in cancer survivors. To investigate the impact of high dose chemotherapy and stem cell transplantation on senescence markers in vivo, we collected blood and clinical data from a cohort of 63 patients undergoing hematopoietic cell transplantation. The expression of p16INK4a, a well-established senescence marker, was determined in T-cells before and 6 months after transplant. RNA sequencing was performed on paired samples from 8 patients pre- and post-cancer therapy. In patients undergoing allogeneic transplant, higher pre-transplant p16INK4a expression was associated with a greater number of prior cycles of chemotherapy received (p = 0.003), prior autologous transplantation (p = 0.01) and prior exposure to alkylating agents (p = 0.01). Transplantation was associated with a marked increase in p16INK4a expression 6 months following transplantation. Patients receiving autologous transplant experienced a larger increase in p16INK4a expression (3.1-fold increase, p = 0.002) than allogeneic transplant recipients (1.9-fold increase, p = 0.0004). RNA sequencing of T-cells pre- and post- autologous transplant or cytotoxic chemotherapy demonstrated increased expression of transcripts associated with cellular senescence and physiological aging. Cytotoxic chemotherapy, especially alkylating agents, and stem cell transplantation strongly accelerate expression of a biomarker of molecular aging in T-cells. Peripheral blood T-cell senescence, as measured by the marker p16INK4a, increases following autologous or allogeneic HSCT. RNAseq of T-cells post- auto HSCT or chemotherapy show increased expression of transcripts associated with senescence. Autologous HCT in particular induces a stronger effect on Tcell p16INK4a expression than any other environmental stimulus tested to date.
Human chronological aging is associated with increased expression of markers of cellular aging (senescence). Cancer chemotherapy can produce frailty syndromes – recipients of cancer treatment may experience physiological changes ordinarily seen in individuals of more advanced chronological age. In our study, we found that a well-known marker of cellular senescence, p16INK4a, increased in patients following autologous or allogeneic hematopoietic cell transplantation. Expression of p16INK4a was higher in patients exposed to greater amounts of chemotherapy before transplant and those exposed to specific types of chemotherapy. These findings may ultimately influence clinical decision-making for patients with diseases that are commonly treated with transplantation.
Collapse
Affiliation(s)
- William A Wood
- Department of Medicine, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Janakiraman Krishnamurthy
- Department of Medicine, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Genetics, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Natalia Mitin
- Department of Medicine, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Genetics, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Chad Torrice
- Department of Medicine, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Genetics, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Joel S Parker
- Department of Genetics, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Anna C Snavely
- Department of Medicine, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Thomas C Shea
- Department of Medicine, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jonathan S Serody
- Department of Medicine, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Norman E Sharpless
- Department of Medicine, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Genetics, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
47
|
Burd CE, Gill MS, Niedernhofer LJ, Robbins PD, Austad SN, Barzilai N, Kirkland JL. Barriers to the Preclinical Development of Therapeutics that Target Aging Mechanisms. J Gerontol A Biol Sci Med Sci 2016; 71:1388-1394. [PMID: 27535964 PMCID: PMC5055650 DOI: 10.1093/gerona/glw112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/02/2016] [Indexed: 01/08/2023] Open
Abstract
Through the progress of basic science research, fundamental mechanisms that contribute to age-related decline are being described with increasing depth and detail. Although these efforts have identified new drug targets and compounds that extend life span in model organisms, clinical trials of therapeutics that target aging processes remain scarce. Progress in aging research is hindered by barriers associated with the translation of basic science discoveries into the clinic. This report summarizes discussions held at a 2014 Geroscience Network retreat focused on identifying hurdles that currently impede the preclinical development of drugs targeting fundamental aging processes. From these discussions, it was evident that aging researchers have varied perceptions of the ideal preclinical pipeline. To forge a clear and cohesive path forward, several areas of controversy must first be resolved and new tools developed. Here, we focus on five key issues in preclinical drug development (drug discovery, lead compound development, translational preclinical biomarkers, funding, and integration between researchers and clinicians), expanding upon discussions held at the Geroscience Retreat and suggesting areas for further research. By bringing these findings to the attention of the aging research community, we hope to lay the foundation for a concerted preclinical drug development pipeline.
Collapse
Affiliation(s)
- Christin E Burd
- Department of Molecular Genetics and.,Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus
| | - Matthew S Gill
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida
| | - Laura J Niedernhofer
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida
| | - Paul D Robbins
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida
| | | | - Nir Barzilai
- Department of Medicine, Division of Endocrinology and.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
48
|
Sosa Peña MDP, Lopez-Soler R, Melendez JA. Senescence in chronic allograft nephropathy. Am J Physiol Renal Physiol 2016; 315:F880-F889. [PMID: 27306980 DOI: 10.1152/ajprenal.00195.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Despite increasing numbers of patients on dialysis, the numbers of renal transplants performed yearly have remained relatively static. During the last 50 years, there have been many advances in the pharmacology of prevention of organ rejection. However, most patients will suffer from a slow but steady decline in renal function leading to graft loss. The most common cause of long-term graft loss is chronic allograft nephropathy (CAN). Therefore, elucidating and understanding the mechanisms involved in CAN is crucial for achieving better posttransplant outcomes. It is thought that the development of epithelial to mesenchymal transition (EMT) in proximal tubules is one of the first steps towards CAN, and has been shown to be a result of cellular senescence. Cells undergoing senescence acquire a senescence associated secretory phenotype (SASP) leading to the production of interleukin-1 alpha (IL-1α), which has been implicated in several degenerative and inflammatory processes including renal disease. A central mediator in SASP activation is the production of reactive oxygen species (ROS), which are produced in response to numerous physiological and pathological stimuli. This review explores the connection between SASP and the development of EMT/CAN in an effort to suggest future directions for research leading to improved long-term graft outcomes.
Collapse
Affiliation(s)
| | - Reynold Lopez-Soler
- Albany Medical Center, Department of Surgery, Division of Transplantation, Albany, New York
| | - J Andrés Melendez
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, Albany, New York
| |
Collapse
|
49
|
Leierer J, Rudnicki M, Braniff SJ, Perco P, Koppelstaetter C, Mühlberger I, Eder S, Kerschbaum J, Schwarzer C, Schroll A, Weiss G, Schneeberger S, Wagner S, Königsrainer A, Böhmig GA, Mayer G. Metallothioneins and renal ageing. Nephrol Dial Transplant 2016; 31:1444-52. [PMID: 26908771 DOI: 10.1093/ndt/gfv451] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/16/2015] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Human lifespan is increasing continuously and about one-third of the population >70 years of age suffers from chronic kidney disease. The pathophysiology of the loss of renal function with ageing is unclear. METHODS We determined age-associated gene expression changes in zero-hour biopsies of deceased donor kidneys without laboratory signs of impaired renal function, defined as a last serum creatinine >0.96 mg/dL in females and >1.18 mg/dL in males, using microarray technology and the Significance Analysis of Microarrays routine. Expression changes of selected genes were confirmed by quantitative polymerase chain reaction and in situ hybridization and immunohistochemistry for localization of respective mRNA and protein. Functional aspects were examined in vitro. RESULTS Donors were classified into three age groups (<40, 40-59 and >59 years; Groups 1, 2 and 3, respectively). In Group 3 especially, genes encoding for metallothionein (MT) isoforms were more significantly expressed when compared with Group 1; localization studies revealed predominant staining in renal proximal tubular cells. RPTEC/TERT1 cells overexpressing MT2A were less susceptible towards cadmium chloride-induced cytotoxicity and hypoxia-induced apoptosis, both models for increased generation of reactive oxygen species. CONCLUSIONS Increased expression of MTs in the kidney with ageing might be a protective mechanism against increased oxidative stress, which is closely related to the ageing process. Our findings indicate that MTs are functionally involved in the pathophysiology of ageing-related processes.
Collapse
Affiliation(s)
- Johannes Leierer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Michael Rudnicki
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Susie-Jane Braniff
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Paul Perco
- Emergentec Biodevelopment GmbH, Vienna, Austria
| | - Christian Koppelstaetter
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | | | - Susanne Eder
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Julia Kerschbaum
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Christoph Schwarzer
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine VI, Clinical Immunology and Infectious Diseases, Medical University, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine VI, Clinical Immunology and Infectious Diseases, Medical University, Innsbruck, Austria
| | - Stefan Schneeberger
- Center of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Silvia Wagner
- Department of General, Visceral and Transplant Surgery, University of Tübingen, Tübingen, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University of Tübingen, Tübingen, Germany
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| |
Collapse
|
50
|
Zhao X, Wang Y, Wang Y, Li S, Chen P. Oxidative stress and premature senescence in corneal endothelium following penetrating keratoplasty in an animal model. BMC Ophthalmol 2016; 16:16. [PMID: 26839109 PMCID: PMC4736695 DOI: 10.1186/s12886-016-0192-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 01/27/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The purpose of this study was to address the question of how the premature senescence process may affect corneal endothelium after penetrating keratoplasty, because the quality of donor corneal endothelial cells is important for corneal transplant success. METHODS The cell senescence and induced oxidative stress in corneal endothelium were assessed using a normal-risk orthotopic mice corneal transplantation model. Senescence associated beta-galactosidase (SA-beta-Gal) staining was used to evaluate premature senescence in the endothelium of corneal allografts. Oxidative Stress and Antioxidant Defense RT(2)-PCR Arrays and in vitro experimental model using H2O2 treatment were used to investigate the possible mechanism. RESULTS SA-beta-Gal positivity was observed obviously in mice corneal endothelium of allogenic group and the levels of p16(INK4a) message and protein increased in endothelium of allogenic group compared to syngenic group. By PCR array, an oxidant-antioxidant imbalance was found in the endothelium of corneal allograft after PKP. The results from mice model were validated using human endothelium samples of corneal allograft after PKP. We also developed an in vitro experimental model using H2O2 treatment to simulate a state of oxidative stress in cultured human corneal endothelial cells (HCECs) and found that elevated ROS levels, the up-regulation of CDK inhibitors and ROS-mediated p16(INK4A) up-regulation in HCECs occur via the ASK1-p38 MAPK pathway. CONCLUSIONS Our results demonstrate the presence of oxidative stress and premature senescence in the endothelium of corneal allografts following PKP.
Collapse
Affiliation(s)
- Xiaowen Zhao
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of medical Sciences, No. 5 Yanerdao Rd, Qingdao, 266071, China
| | - Ye Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of medical Sciences, No. 5 Yanerdao Rd, Qingdao, 266071, China. .,Current affiliation: Central Laboratory of the Second Affiliated Hospital, Medical College of Qingdao University, Qingdao, 266042, China.
| | - Yao Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of medical Sciences, No. 5 Yanerdao Rd, Qingdao, 266071, China
| | - Suxia Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of medical Sciences, No. 5 Yanerdao Rd, Qingdao, 266071, China
| | - Peng Chen
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of medical Sciences, No. 5 Yanerdao Rd, Qingdao, 266071, China
| |
Collapse
|