1551
|
Hecker L. Mechanisms and consequences of oxidative stress in lung disease: therapeutic implications for an aging populace. Am J Physiol Lung Cell Mol Physiol 2017; 314:L642-L653. [PMID: 29351446 DOI: 10.1152/ajplung.00275.2017] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The rapid expansion of the elderly population has led to the recent epidemic of age-related diseases, including increased incidence and mortality of chronic and acute lung diseases. Numerous studies have implicated aging and oxidative stress in the pathogenesis of various pulmonary diseases; however, despite recent advances in these fields, the specific contributions of aging and oxidative stress remain elusive. This review will discuss the consequences of aging on lung morphology and physiology, and how redox imbalance with aging contributes to lung disease susceptibility. Here, we focus on three lung diseases for which aging is a significant risk factor: acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF). Preclinical and clinical development for redox- and senescence-altering therapeutic strategies are discussed, as well as scientific advancements that may direct current and future therapeutic development. A deeper understanding of how aging impacts normal lung function, redox balance, and injury-repair processes will inspire the development of new therapies to prevent and/or reverse age-associated pulmonary diseases, and ultimately increase health span and longevity. This review is intended to encourage basic, clinical, and translational research that will bridge knowledge gaps at the intersection of aging, oxidative stress, and lung disease to fuel the development of more effective therapeutic strategies for lung diseases that disproportionately afflict the elderly.
Collapse
Affiliation(s)
- Louise Hecker
- Division of Pulmonary, Allergy and Critical Care and Sleep Medicine, University of Arizona , Tucson, Arizona and Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona
| |
Collapse
|
1552
|
Brighton PJ, Maruyama Y, Fishwick K, Vrljicak P, Tewary S, Fujihara R, Muter J, Lucas ES, Yamada T, Woods L, Lucciola R, Hou Lee Y, Takeda S, Ott S, Hemberger M, Quenby S, Brosens JJ. Clearance of senescent decidual cells by uterine natural killer cells in cycling human endometrium. eLife 2017; 6. [PMID: 29227245 PMCID: PMC5724991 DOI: 10.7554/elife.31274] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/02/2017] [Indexed: 12/22/2022] Open
Abstract
In cycling human endometrium, menstruation is followed by rapid estrogen-dependent growth. Upon ovulation, progesterone and rising cellular cAMP levels activate the transcription factor Forkhead box O1 (FOXO1) in endometrial stromal cells (EnSCs), leading to cell cycle exit and differentiation into decidual cells that control embryo implantation. Here we show that FOXO1 also causes acute senescence of a subpopulation of decidualizing EnSCs in an IL-8 dependent manner. Selective depletion or enrichment of this subpopulation revealed that decidual senescence drives the transient inflammatory response associated with endometrial receptivity. Further, senescent cells prevent differentiation of endometrial mesenchymal stem cells in decidualizing cultures. As the cycle progresses, IL-15 activated uterine natural killer (uNK) cells selectively target and clear senescent decidual cells through granule exocytosis. Our findings reveal that acute decidual senescence governs endometrial rejuvenation and remodeling at embryo implantation, and suggest a critical role for uNK cells in maintaining homeostasis in cycling endometrium.
Collapse
Affiliation(s)
- Paul J Brighton
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Yojiro Maruyama
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Katherine Fishwick
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Pavle Vrljicak
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Tommy's National Centre for Miscarriage Research, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Shreeya Tewary
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Tommy's National Centre for Miscarriage Research, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Risa Fujihara
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Joanne Muter
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Emma S Lucas
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Tommy's National Centre for Miscarriage Research, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Taihei Yamada
- Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Laura Woods
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom.,Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Raffaella Lucciola
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Yie Hou Lee
- Obstetrics & Gynaecology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore.,KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Sascha Ott
- Tommy's National Centre for Miscarriage Research, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Myriam Hemberger
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom.,Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Siobhan Quenby
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Jan Joris Brosens
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
1553
|
Anti-TNF-α treatment modulates SASP and SASP-related microRNAs in endothelial cells and in circulating angiogenic cells. Oncotarget 2017; 7:11945-58. [PMID: 26943583 PMCID: PMC4914260 DOI: 10.18632/oncotarget.7858] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/14/2016] [Indexed: 12/27/2022] Open
Abstract
Endothelial cell senescence is characterized by acquisition of senescence-associated secretory phenotype (SASP), able to promote inflammaging and cancer progression. Emerging evidence suggest that preventing SASP development could help to slow the rate of aging and the progression of age-related diseases, including cancer. Aim of this study was to evaluate whether and how adalimumab, a monoclonal antibody directed against tumor necrosis factor-α (TNF-α), a major SASP component, can prevent the SASP. A three-pronged approach has been adopted to assess the if adalimumab is able to: i) modulate a panel of classic and novel senescence- and SASP-associated markers (interleukin [IL]-6, senescence associated-β-galactosidase, p16/Ink4a, plasminogen activator inhibitor 1, endothelial nitric oxide synthase, miR-146a-5p/Irak1 and miR-126-3p/Spred1) in human umbilical vein endothelial cells (HUVECs); ii) reduce the paracrine effects of senescent HUVECs' secretome on MCF-7 breast cancer cells, through wound healing and mammosphere assay; and iii) exert significant decrease of miR-146a-5p and increase of miR-126-3p in circulating angiogenic cells (CACs) from psoriasis patients receiving adalimumab in monotherapy.TNF-α blockade associated with adalimumab induced significant reduction in released IL-6 and significant increase in eNOS and miR-126-3p expression levels in long-term HUVEC cultures.A significant reduction in miR-146a-5p expression levels both in long-term HUVEC cultures and in CACs isolated from psoriasis patients was also evident. Interestingly, conditioned medium from senescent HUVECs treated with adalimumab was less consistent than medium from untreated cells in inducing migration- and mammosphere- promoting effects on MCF-7 cells.Our findings suggest that adalimumab can induce epigenetic modifications in cells undergoing senescence, thus contributing to the attenuation of SASP tumor-promoting effects.
Collapse
|
1554
|
Abbadie C, Pluquet O, Pourtier A. Epithelial cell senescence: an adaptive response to pre-carcinogenic stresses? Cell Mol Life Sci 2017; 74:4471-4509. [PMID: 28707011 PMCID: PMC11107641 DOI: 10.1007/s00018-017-2587-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 01/01/2023]
Abstract
Senescence is a cell state occurring in vitro and in vivo after successive replication cycles and/or upon exposition to various stressors. It is characterized by a strong cell cycle arrest associated with several molecular, metabolic and morphologic changes. The accumulation of senescent cells in tissues and organs with time plays a role in organismal aging and in several age-associated disorders and pathologies. Moreover, several therapeutic interventions are able to prematurely induce senescence. It is, therefore, tremendously important to characterize in-depth, the mechanisms by which senescence is induced, as well as the precise properties of senescent cells. For historical reasons, senescence is often studied with fibroblast models. Other cell types, however, much more relevant regarding the structure and function of vital organs and/or regarding pathologies, are regrettably often neglected. In this article, we will clarify what is known on senescence of epithelial cells and highlight what distinguishes it from, and what makes it like, replicative senescence of fibroblasts taken as a standard.
Collapse
Affiliation(s)
- Corinne Abbadie
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, 59000, Lille, France.
| | - Olivier Pluquet
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, 59000, Lille, France
| | - Albin Pourtier
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, 59000, Lille, France
| |
Collapse
|
1555
|
Ademowo OS, Dias HKI, Burton DGA, Griffiths HR. Lipid (per) oxidation in mitochondria: an emerging target in the ageing process? Biogerontology 2017; 18:859-879. [PMID: 28540446 PMCID: PMC5684309 DOI: 10.1007/s10522-017-9710-z] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022]
Abstract
Lipids are essential for physiological processes such as maintaining membrane integrity, providing a source of energy and acting as signalling molecules to control processes including cell proliferation, metabolism, inflammation and apoptosis. Disruption of lipid homeostasis can promote pathological changes that contribute towards biological ageing and age-related diseases. Several age-related diseases have been associated with altered lipid metabolism and an elevation in highly damaging lipid peroxidation products; the latter has been ascribed, at least in part, to mitochondrial dysfunction and elevated ROS formation. In addition, senescent cells, which are known to contribute significantly to age-related pathologies, are also associated with impaired mitochondrial function and changes in lipid metabolism. Therapeutic targeting of dysfunctional mitochondrial and pathological lipid metabolism is an emerging strategy for alleviating their negative impact during ageing and the progression to age-related diseases. Such therapies could include the use of drugs that prevent mitochondrial uncoupling, inhibit inflammatory lipid synthesis, modulate lipid transport or storage, reduce mitochondrial oxidative stress and eliminate senescent cells from tissues. In this review, we provide an overview of lipid structure and function, with emphasis on mitochondrial lipids and their potential for therapeutic targeting during ageing and age-related disease.
Collapse
Affiliation(s)
- O S Ademowo
- Life & Health Sciences, Aston University, Birmingham, UK
| | - H K I Dias
- Life & Health Sciences, Aston University, Birmingham, UK
| | - D G A Burton
- Life & Health Sciences, Aston University, Birmingham, UK
| | - H R Griffiths
- Life & Health Sciences, Aston University, Birmingham, UK.
- Health and Medical Sciences, University of Surrey, Guildford, UK.
| |
Collapse
|
1556
|
Schosserer M, Grillari J, Breitenbach M. The Dual Role of Cellular Senescence in Developing Tumors and Their Response to Cancer Therapy. Front Oncol 2017; 7:278. [PMID: 29218300 PMCID: PMC5703792 DOI: 10.3389/fonc.2017.00278] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/06/2017] [Indexed: 12/11/2022] Open
Abstract
Cellular senescence describes an irreversible growth arrest characterized by distinct morphology, gene expression pattern, and secretory phenotype. The final or intermediate stages of senescence can be reached by different genetic mechanisms and in answer to different external and internal stresses. It has been maintained in the literature but never proven by clearcut experiments that the induction of senescence serves the evolutionary purpose of protecting the individual from development and growth of cancers. This hypothesis was recently scrutinized by new experiments and found to be partly true, but part of the gene activities now known to happen in senescence are also needed for cancer growth, leading to the view that senescence is a double-edged sword in cancer development. In current cancer therapy, cellular senescence is, on the one hand, intended to occur in tumor cells, as thereby the therapeutic outcome is improved, but might, on the other hand, also be induced unintentionally in non-tumor cells, causing inflammation, secondary tumors, and cancer relapse. Importantly, organismic aging leads to accumulation of senescent cells in tissues and organs of aged individuals. Senescent cells can occur transiently, e.g., during embryogenesis or during wound healing, with beneficial effects on tissue homeostasis and regeneration or accumulate chronically in tissues, which detrimentally affects the microenvironment by de- or transdifferentiation of senescent cells and their neighboring stromal cells, loss of tissue specific functionality, and induction of the senescence-associated secretory phenotype, an increased secretory profile consisting of pro-inflammatory and tissue remodeling factors. These factors shape their surroundings toward a pro-carcinogenic microenvironment, which fuels the development of aging-associated cancers together with the accumulation of mutations over time. We are presenting an overview of well-documented stress situations and signals, which induce senescence. Among them, oncogene-induced senescence and stress-induced premature senescence are prominent. New findings about the role of senescence in tumor biology are critically reviewed with respect to new suggestions for cancer therapy leveraging genetic and pharmacological methods to prevent senescence or to selectively kill senescent cells in tumors.
Collapse
Affiliation(s)
- Markus Schosserer
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Johannes Grillari
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria.,Evercyte GmbH, Vienna, Austria
| | - Michael Breitenbach
- Department of Cell Biology, Division of Genetics, University of Salzburg, Salzburg, Austria
| |
Collapse
|
1557
|
Hall BM, Balan V, Gleiberman AS, Strom E, Krasnov P, Virtuoso LP, Rydkina E, Vujcic S, Balan K, Gitlin I, Leonova K, Polinsky A, Chernova OB, Gudkov AV. Aging of mice is associated with p16(Ink4a)- and β-galactosidase-positive macrophage accumulation that can be induced in young mice by senescent cells. Aging (Albany NY) 2017; 8:1294-315. [PMID: 27391570 PMCID: PMC4993332 DOI: 10.18632/aging.100991] [Citation(s) in RCA: 261] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/28/2016] [Indexed: 12/12/2022]
Abstract
Senescent cells (SCs) have been considered a source of age-related chronic sterile systemic inflammation and a target for anti-aging therapies. To understand mechanisms controlling the amount of SCs, we analyzed the phenomenon of rapid clearance of human senescent fibroblasts implanted into SCID mice, which can be overcome when SCs were embedded into alginate beads preventing them from immunocyte attack. To identify putative SC killers, we analyzed the content of cell populations in lavage and capsules formed around the SC-containing beads. One of the major cell types attracted by secretory factors of SCs was a subpopulation of macrophages characterized by p16(Ink4a) gene expression and β-galactosidase activity at pH6.0 (β-gal(pH6)), thus resembling SCs. Consistently, mice with p16(Ink4a) promoter-driven luciferase, developed bright luminescence of their peritoneal cavity within two weeks following implantation of SCs embedded in alginate beads. p16(Ink4a)/β-gal(pH6)-expressing cells had surface biomarkers of macrophages F4/80 and were sensitive to liposomal clodronate used for the selective killing of cells capable of phagocytosis. At the same time, clodronate failed to kill bona fide SCs generated in vitro by genotoxic stress. Old mice with elevated proportion of p16(Ink4a)/β-gal(pH6)-positive cells in their tissues demonstrated reduction of both following systemic clodronate treatment, indicating that a significant proportion of cells previously considered to be SCs are actually a subclass of macrophages. These observations point at a significant role of p16(Ink4a)/β-gal(pH6)-positive macrophages in aging, which previously was attributed solely to SCs. They require re-interpretation of the mechanisms underlying rejuvenating effects following eradication of p16(Ink4a)/β-gal(pH6)-positive cells and reconsideration of potential cellular target for anti-aging treatment.
Collapse
Affiliation(s)
| | - Vitaly Balan
- Everon Biosciences, Inc., Buffalo, NY 14203, USA
| | | | | | | | | | | | | | - Karina Balan
- Everon Biosciences, Inc., Buffalo, NY 14203, USA
| | - Ilya Gitlin
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Katerina Leonova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | - Andrei V Gudkov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
1558
|
Zhu Y, Doornebal EJ, Pirtskhalava T, Giorgadze N, Wentworth M, Fuhrmann-Stroissnigg H, Niedernhofer LJ, Robbins PD, Tchkonia T, Kirkland JL. New agents that target senescent cells: the flavone, fisetin, and the BCL-X L inhibitors, A1331852 and A1155463. Aging (Albany NY) 2017; 9:955-963. [PMID: 28273655 PMCID: PMC5391241 DOI: 10.18632/aging.101202] [Citation(s) in RCA: 489] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/07/2017] [Indexed: 12/19/2022]
Abstract
Senescent cells accumulate with aging and at sites of pathology in multiple chronic diseases. Senolytics are drugs that selectively promote apoptosis of senescent cells by temporarily disabling the pro-survival pathways that enable senescent cells to resist the pro-apoptotic, pro-inflammatory factors that they themselves secrete. Reducing senescent cell burden by genetic approaches or by administering senolytics delays or alleviates multiple age- and disease-related adverse phenotypes in preclinical models. Reported senolytics include dasatinib, quercetin, navitoclax (ABT263), and piperlongumine. Here we report that fisetin, a naturally-occurring flavone with low toxicity, and A1331852 and A1155463, selective BCL-XL inhibitors that may have less hematological toxicity than the less specific BCL-2 family inhibitor navitoclax, are senolytic. Fisetin selectively induces apoptosis in senescent but not proliferating human umbilical vein endothelial cells (HUVECs). It is not senolytic in senescent IMR90 cells, a human lung fibroblast strain, or primary human preadipocytes. A1331852 and A1155463 are senolytic in HUVECs and IMR90 cells, but not preadipocytes. These agents may be better candidates for eventual translation into clinical interventions than some existing senolytics, such as navitoclax, which is associated with hematological toxicity.
Collapse
Affiliation(s)
- Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Ewald J Doornebal
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA.,Faculty of Science and Engineering, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Tamar Pirtskhalava
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Nino Giorgadze
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Mark Wentworth
- Office of Research Regulatory Support, Mayo Clinic, Rochester, MN 55905, USA
| | - Heike Fuhrmann-Stroissnigg
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Laura J Niedernhofer
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Paul D Robbins
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
1559
|
Schafer MJ, Miller JD, LeBrasseur NK. Cellular senescence: Implications for metabolic disease. Mol Cell Endocrinol 2017; 455:93-102. [PMID: 27591120 PMCID: PMC5857952 DOI: 10.1016/j.mce.2016.08.047] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/03/2016] [Accepted: 08/29/2016] [Indexed: 12/19/2022]
Abstract
The growing burden of obesity- and aging-related diseases has hastened the search for governing biological processes. Cellular senescence is a stress-induced state of stable growth arrest strongly associated with aging that is aberrantly activated by obesity. The transition of a cell to a senescent state is demarcated by an array of phenotypic markers, and leveraging their context-dependent presentation is essential for determining the influence of senescent cells on tissue pathogenesis. Biomarkers of senescent cells have been identified in tissues that contribute to metabolic disease, including fat, liver, skeletal muscle, pancreata, and cardiovascular tissue, suggesting that pharmacological and behavioral interventions that alter their abundance and/or behavior may be a novel therapeutic strategy. However, contradictory findings with regard to a protective versus deleterious role of senescent cells in certain contexts emphasize the need for additional studies to uncover the complex interplay that defines multi-organ disease processes associated with obesity and aging.
Collapse
Affiliation(s)
- Marissa J Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jordan D Miller
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA; Department of Surgery, Mayo Clinic, Rochester, MN, 55905, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
1560
|
Why do we age? Insights into biology and evolution of ageing. Biogerontology 2017; 18:855-857. [PMID: 29086101 DOI: 10.1007/s10522-017-9734-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 10/26/2017] [Indexed: 12/30/2022]
|
1561
|
Mora AL, Rojas M, Pardo A, Selman M. Emerging therapies for idiopathic pulmonary fibrosis, a progressive age-related disease. Nat Rev Drug Discov 2017; 16:810. [PMID: 29081515 DOI: 10.1038/nrd.2017.225] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
1562
|
|
1563
|
Mavrogonatou E, Pratsinis H, Papadopoulou A, Karamanos NK, Kletsas D. Extracellular matrix alterations in senescent cells and their significance in tissue homeostasis. Matrix Biol 2017; 75-76:27-42. [PMID: 29066153 DOI: 10.1016/j.matbio.2017.10.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/13/2017] [Accepted: 10/14/2017] [Indexed: 12/16/2022]
Abstract
Normal cells after a defined number of successive divisions or after exposure to genotoxic stresses are becoming senescent, characterized by a permanent growth arrest. In addition, they secrete increased levels of pro-inflammatory and catabolic mediators, collectively termed "senescence-associated secretory phenotype". Furthermore, senescent cells exhibit an altered expression and organization of many extracellular matrix components, leading to specific remodeling of their microenvironment. In this review we present the current knowledge on extracellular matrix alterations associated with cellular senescence and critically discuss certain characteristic examples, highlighting the ambiguous role of senescent cells in the homeostasis of various tissues under both normal and pathologic conditions.
Collapse
Affiliation(s)
- Eleni Mavrogonatou
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Harris Pratsinis
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Adamantia Papadopoulou
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece.
| |
Collapse
|
1564
|
Lujambio A. To clear, or not to clear (senescent cells)? That is the question. Bioessays 2017; 38 Suppl 1:S56-64. [PMID: 27417123 DOI: 10.1002/bies.201670910] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/30/2015] [Accepted: 12/09/2015] [Indexed: 12/22/2022]
Abstract
Cellular senescence is an anti-proliferative program that restricts the propagation of cells subjected to different kinds of stress. Cellular senescence was initially described as a cell-autonomous tumor suppressor mechanism that triggers an irreversible cell cycle arrest that prevents the proliferation of damaged cells at risk of neoplastic transformation. However, discoveries during the last decade have established that senescent cells can also impact the surrounding tissue microenvironment and the neighboring cells in a non-cell-autonomous manner. These non-cell-autonomous activities are, in part, mediated by the selective secretion of extracellular matrix degrading enzymes, cytokines, chemokines and immune modulators, which collectively constitute the senescence-associated secretory phenotype. One of the key functions of the senescence-associated secretory phenotype is to attract immune cells, which in turn can orchestrate the elimination of senescent cells. Interestingly, the clearance of senescent cells seems to be critical to dictate the net effects of cellular senescence. As a general rule, the successful elimination of senescent cells takes place in processes that are considered beneficial, such as tumor suppression, tissue remodeling and embryonic development, while the chronic accumulation of senescent cells leads to more detrimental consequences, namely, cancer and aging. Nevertheless, exceptions to this rule may exist. Now that cellular senescence is in the spotlight for both anti-cancer and anti-aging therapies, understanding the precise underpinnings of senescent cell removal will be essential to exploit cellular senescence to its full potential.
Collapse
Affiliation(s)
- Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
1565
|
Vidal-Bralo L, Lopez-Golan Y, Mera-Varela A, Rego-Perez I, Horvath S, Zhang Y, Del Real Á, Zhai G, Blanco FJ, Riancho JA, Gomez-Reino JJ, Gonzalez A. Specific premature epigenetic aging of cartilage in osteoarthritis. Aging (Albany NY) 2017; 8:2222-2231. [PMID: 27689435 PMCID: PMC5076459 DOI: 10.18632/aging.101053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/14/2016] [Indexed: 12/18/2022]
Abstract
Osteoarthritis (OA) is a disease affecting multiple tissues of the joints in the elderly, but most notably articular cartilage. Premature biological aging has been described in this tissue and in blood cells, suggesting a systemic component of premature aging in the pathogenesis of OA. Here, we have explored epigenetic aging in OA at the local (cartilage and bone) and systemic (blood) levels. Two DNA methylation age-measures (DmAM) were used: the multi-tissue age estimator for cartilage and bone; and a blood-specific biomarker for blood. Differences in DmAM between OA patients and controls showed an accelerated aging of 3.7 years in articular cartilage (95% CI = 1.1 to 6.3, P = 0.008) of OA patients. By contrast, no difference in epigenetic aging was observed in bone (0.04 years; 95% CI = -1.8 to 1.9, P = 0.3) and in blood (-0.6 years; 95% CI = -1.5 to 0.3, P = 0.2) between OA patients and controls. Therefore, premature epigenetic aging according to DNA methylation changes was specific of OA cartilage, adding further evidence and insight on premature aging of cartilage as a component of OA pathogenesis that reflects damage and vulnerability.
Collapse
Affiliation(s)
- Laura Vidal-Bralo
- Laboratorio Investigacion 10 and Rheumatology Unit, Instituto Investigacion Sanitaria, Hospital Clinico Universitario de Santiago, Travesia Choupana, sn. 15706- Santiago de Compostela, Spain
| | - Yolanda Lopez-Golan
- Laboratorio Investigacion 10 and Rheumatology Unit, Instituto Investigacion Sanitaria, Hospital Clinico Universitario de Santiago, Travesia Choupana, sn. 15706- Santiago de Compostela, Spain
| | - Antonio Mera-Varela
- Laboratorio Investigacion 10 and Rheumatology Unit, Instituto Investigacion Sanitaria, Hospital Clinico Universitario de Santiago, Travesia Choupana, sn. 15706- Santiago de Compostela, Spain
| | - Ignacio Rego-Perez
- Grupo de Reumatología, Instituto de Investigación Biomédica de A Coruña. Complexo Hospitalario Universitario de A Coruña, Universidade da Coruña. As Xubias, sn. 15006- A Coruña, Spain
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yuhua Zhang
- Discipline of Genetics, Faculty of Medicine, Memorial University of Newfoundland, A1B- St. John's, NL, Canada
| | - Álvaro Del Real
- Department of Internal Medicine, Hospital U. M. Valdecilla-IDIVAL, University of Cantabria, Cardenal Herrera Oria, 39011, Santander, Spain
| | - Guangju Zhai
- Discipline of Genetics, Faculty of Medicine, Memorial University of Newfoundland, A1B- St. John's, NL, Canada
| | - Francisco J Blanco
- Grupo de Reumatología, Instituto de Investigación Biomédica de A Coruña. Complexo Hospitalario Universitario de A Coruña, Universidade da Coruña. As Xubias, sn. 15006- A Coruña, Spain
| | - Jose A Riancho
- Department of Internal Medicine, Hospital U. M. Valdecilla-IDIVAL, University of Cantabria, Cardenal Herrera Oria, 39011, Santander, Spain
| | - Juan J Gomez-Reino
- Laboratorio Investigacion 10 and Rheumatology Unit, Instituto Investigacion Sanitaria, Hospital Clinico Universitario de Santiago, Travesia Choupana, sn. 15706- Santiago de Compostela, Spain
| | - Antonio Gonzalez
- Laboratorio Investigacion 10 and Rheumatology Unit, Instituto Investigacion Sanitaria, Hospital Clinico Universitario de Santiago, Travesia Choupana, sn. 15706- Santiago de Compostela, Spain
| |
Collapse
|
1566
|
Anderson R, Richardson GD, Passos JF. Mechanisms driving the ageing heart. Exp Gerontol 2017; 109:5-15. [PMID: 29054534 DOI: 10.1016/j.exger.2017.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/16/2017] [Indexed: 01/07/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. One of the main risk factors for CVD is age, however the biological processes that occur in the heart during ageing are poorly understood. It is therefore important to understand the fundamental mechanisms driving heart ageing to enable the development of preventions and treatments targeting these processes. Cellular senescence is often described as the irreversible cell-cycle arrest which occurs in somatic cells. Emerging evidence suggests that cellular senescence plays a key role in heart ageing, however the cell-types involved and the underlying mechanisms are not yet elucidated. In this review we discuss the current understanding of how mechanisms known to contribute to senescence impact on heart ageing and CVD. Finally, we evaluate recent data suggesting that targeting senescent cells may be a viable therapy to counteract the ageing of the heart.
Collapse
Affiliation(s)
- Rhys Anderson
- The Randall Division, King's College London, London, UK; Ageing Research Laboratories, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK; Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Gavin D Richardson
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - João F Passos
- Ageing Research Laboratories, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK; Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
1567
|
Mora AL, Rojas M, Pardo A, Selman M. Emerging therapies for idiopathic pulmonary fibrosis, a progressive age-related disease. Nat Rev Drug Discov 2017; 16:755-772. [DOI: 10.1038/nrd.2017.170] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
1568
|
|
1569
|
Deschênes M, Chabot B. The emerging role of alternative splicing in senescence and aging. Aging Cell 2017; 16:918-933. [PMID: 28703423 PMCID: PMC5595669 DOI: 10.1111/acel.12646] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 12/22/2022] Open
Abstract
Deregulation of precursor mRNA splicing is associated with many illnesses and has been linked to age-related chronic diseases. Here we review recent progress documenting how defects in the machinery that performs intron removal and controls splice site selection contribute to cellular senescence and organismal aging. We discuss the functional association linking p53, IGF-1, SIRT1, and ING-1 splice variants with senescence and aging, and review a selection of splicing defects occurring in accelerated aging (progeria), vascular aging, and Alzheimer's disease. Overall, it is becoming increasingly clear that changes in the activity of splicing factors and in the production of key splice variants can impact cellular senescence and the aging phenotype.
Collapse
Affiliation(s)
- Mathieu Deschênes
- Department of Microbiology and Infectious DiseasesFaculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecJ1E 4K8Canada
| | - Benoit Chabot
- Department of Microbiology and Infectious DiseasesFaculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecJ1E 4K8Canada
| |
Collapse
|
1570
|
Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J, van Deursen JM. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov 2017; 16:718-735. [PMID: 28729727 PMCID: PMC5942225 DOI: 10.1038/nrd.2017.116] [Citation(s) in RCA: 843] [Impact Index Per Article: 105.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronological age represents the single greatest risk factor for human disease. One plausible explanation for this correlation is that mechanisms that drive ageing might also promote age-related diseases. Cellular senescence, which is a permanent state of cell cycle arrest induced by cellular stress, has recently emerged as a fundamental ageing mechanism that also contributes to diseases of late life, including cancer, atherosclerosis and osteoarthritis. Therapeutic strategies that safely interfere with the detrimental effects of cellular senescence, such as the selective elimination of senescent cells (SNCs) or the disruption of the SNC secretome, are gaining significant attention, with several programmes now nearing human clinical studies.
Collapse
Affiliation(s)
| | | | - Darren J Baker
- Departments of Biochemistry and Molecular Biology, Mayo Clinic
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st St. SW, Rochester, Minnesota 55905, USA
| | - Remi-Martin Laberge
- Unity Biotechnology, 3280 Bayshore Boulevard Suite 100, Brisbane, California 94005, USA
| | - Dan Marquess
- Unity Biotechnology, 3280 Bayshore Boulevard Suite 100, Brisbane, California 94005, USA
| | - Jamie Dananberg
- Unity Biotechnology, 3280 Bayshore Boulevard Suite 100, Brisbane, California 94005, USA
| | - Jan M van Deursen
- Departments of Biochemistry and Molecular Biology, Mayo Clinic
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st St. SW, Rochester, Minnesota 55905, USA
| |
Collapse
|
1571
|
Rossman MJ, Kaplon RE, Hill SD, McNamara MN, Santos-Parker JR, Pierce GL, Seals DR, Donato AJ. Endothelial cell senescence with aging in healthy humans: prevention by habitual exercise and relation to vascular endothelial function. Am J Physiol Heart Circ Physiol 2017; 313:H890-H895. [PMID: 28971843 DOI: 10.1152/ajpheart.00416.2017] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/22/2017] [Accepted: 09/22/2017] [Indexed: 01/27/2023]
Abstract
Cellular senescence is emerging as a key mechanism of age-related vascular endothelial dysfunction, but evidence in healthy humans is lacking. Moreover, the influence of lifestyle factors such as habitual exercise on endothelial cell (EC) senescence is unknown. We tested the hypothesis that EC senescence increases with sedentary, but not physically active, aging and is associated with vascular endothelial dysfunction. Protein expression (quantitative immunofluorescence) of p53, a transcription factor related to increased cellular senescence, and the cyclin-dependent kinase inhibitors p21 and p16 were 116%, 119%, and 128% greater (all P < 0.05), respectively, in ECs obtained from antecubital veins of older sedentary (60 ± 1 yr, n = 12) versus young sedentary (22 ± 1 yr, n = 9) adults. These age-related differences were not present (all P > 0.05) in venous ECs from older exercising adults (57 ± 1 yr, n = 13). Furthermore, venous EC protein levels of p53 (r = -0.49, P = 0.003), p21 (r = -0.38, P = 0.03), and p16 (r = -0.58, P = 0.002) were inversely associated with vascular endothelial function (brachial artery flow-mediated dilation). Similarly, protein expression of p53 and p21 was 26% and 23% higher (both P < 0.05), respectively, in ECs sampled from brachial arteries of healthy older sedentary (63 ± 1 yr, n = 18) versus young sedentary (25 ± 1 yr, n = 9) adults; age-related changes in arterial EC p53 and p21 expression were not observed (P > 0.05) in older habitually exercising adults (59 ± 1 yr, n = 14). These data indicate that EC senescence is associated with sedentary aging and is linked to endothelial dysfunction. Moreover, these data suggest that prevention of EC senescence may be one mechanism by which aerobic exercise protects against endothelial dysfunction with age.NEW & NOTEWORTHY Our study provides novel evidence in humans of increased endothelial cell senescence with sedentary aging, which is associated with impaired vascular endothelial function. Furthermore, our data suggest an absence of age-related increases in endothelial cell senescence in older exercising adults, which is linked with preserved vascular endothelial function.
Collapse
Affiliation(s)
- Matthew J Rossman
- Integrative Physiology, University of Colorado Boulder, Boulder, Colorado;
| | - Rachelle E Kaplon
- Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Sierra D Hill
- Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Molly N McNamara
- Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | | - Gary L Pierce
- Health and Human Physiology, University of Iowa, Iowa City, Iowa; and
| | - Douglas R Seals
- Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | |
Collapse
|
1572
|
Malavolta M, Costarelli L, Giacconi R, Basso A, Piacenza F, Pierpaoli E, Provinciali M, Ogo OA, Ford D. Changes in Zn homeostasis during long term culture of primary endothelial cells and effects of Zn on endothelial cell senescence. Exp Gerontol 2017; 99:35-45. [PMID: 28918363 DOI: 10.1016/j.exger.2017.09.006] [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: 06/23/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
Abstract
Endothelial cell senescence and Zn nutritional status influence cardiovascular disease. The influence of Zn appears dichotomous, hence it is imperative to understand the relationship with cellular senescence to improve knowledge about the molecular and cellular basis of the disease. Here we aimed to determine: 1) the impact of chronic exposure to a moderately high dose of Zn on senescence of endothelial cells; 2) the changes in Zn homeostasis during the lifespan of primary cultured endothelial cells; and 3) the susceptibility of proliferating and senescent endothelial cells to cell death after short term exposure to increasing doses of Zn and of the Zn chelator TPEN. Chronic exposure to Zn accelerated senescence and untreated cells at later passages, where doubling time had increased, displayed relocation of labile Zn and altered expression of genes involved in the response to Zn toxicity, including SLC30A1, SLC39A6, SLC30A5, SLC30A10 and metallothioneins, indicating that senescent cells have altered zinc homeostasis. Most Zn-dependent genes that were expressed differently between early and late passages were correlated with changes in the expression of anti-apoptotic genes. Short-term treatment with a high dose of Zn leads to cell death, but only in the population of cells at both earlier and later passages that had already entered senescence. In contrast, Zn depletion led to death of cells at earlier but not later passages, which suggests that there are sub-populations of senescent cells that are resistant to Zn depletion. This resistant senescent cell population may accumulate under conditions of Zn deficiency and contribute to vascular pathology.
Collapse
Affiliation(s)
- Marco Malavolta
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy.
| | - Laura Costarelli
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Robertina Giacconi
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Andrea Basso
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Elisa Pierpaoli
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, Scientific and Technological Pole, Italian National Institute of Health and Science on Aging (INRCA), Ancona, Italy
| | - Ogo A Ogo
- Institute for Cell and Molecular Biosciences, University of Newcastle, Newcastle upon Tyne, UK
| | - Dianne Ford
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| |
Collapse
|
1573
|
Stout MB, Justice JN, Nicklas BJ, Kirkland JL. Physiological Aging: Links Among Adipose Tissue Dysfunction, Diabetes, and Frailty. Physiology (Bethesda) 2017; 32:9-19. [PMID: 27927801 DOI: 10.1152/physiol.00012.2016] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Advancing age is associated with progressive declines in physiological function that lead to overt chronic disease, frailty, and eventual mortality. Importantly, age-related physiological changes occur in cellularity, insulin-responsiveness, secretory profiles, and inflammatory status of adipose tissue, leading to adipose tissue dysfunction. Although the mechanisms underlying adipose tissue dysfunction are multifactorial, the consequences result in secretion of proinflammatory cytokines and chemokines, immune cell infiltration, an accumulation of senescent cells, and an increase in senescence-associated secretory phenotype (SASP). These processes synergistically promote chronic sterile inflammation, insulin resistance, and lipid redistribution away from subcutaneous adipose tissue. Without intervention, these effects contribute to age-related systemic metabolic dysfunction, physical limitations, and frailty. Thus adipose tissue dysfunction may be a fundamental contributor to the elevated risk of chronic disease, disability, and adverse health outcomes with advancing age.
Collapse
Affiliation(s)
- Michael B Stout
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jamie N Justice
- Department of Internal Medicine-Geriatrics, Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina; and
| | - Barbara J Nicklas
- Department of Internal Medicine-Geriatrics, Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina; and
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
1574
|
Fuhrmann-Stroissnigg H, Ling YY, Zhao J, McGowan SJ, Zhu Y, Brooks RW, Grassi D, Gregg SQ, Stripay JL, Dorronsoro A, Corbo L, Tang P, Bukata C, Ring N, Giacca M, Li X, Tchkonia T, Kirkland JL, Niedernhofer LJ, Robbins PD. Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun 2017; 8:422. [PMID: 28871086 PMCID: PMC5583353 DOI: 10.1038/s41467-017-00314-z] [Citation(s) in RCA: 474] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/21/2017] [Indexed: 01/07/2023] Open
Abstract
Aging is the main risk factor for many chronic degenerative diseases and cancer. Increased senescent cell burden in various tissues is a major contributor to aging and age-related diseases. Recently, a new class of drugs termed senolytics were demonstrated to extending healthspan, reducing frailty and improving stem cell function in multiple murine models of aging. To identify novel and more optimal senotherapeutic drugs and combinations, we established a senescence associated β-galactosidase assay as a screening platform to rapidly identify drugs that specifically affect senescent cells. We used primary Ercc1 -/- murine embryonic fibroblasts with reduced DNA repair capacity, which senesce rapidly if grown at atmospheric oxygen. This platform was used to screen a small library of compounds that regulate autophagy, identifying two inhibitors of the HSP90 chaperone family as having significant senolytic activity in mouse and human cells. Treatment of Ercc1 -/∆ mice, a mouse model of a human progeroid syndrome, with the HSP90 inhibitor 17-DMAG extended healthspan, delayed the onset of several age-related symptoms and reduced p16INK4a expression. These results demonstrate the utility of our screening platform to identify senotherapeutic agents as well as identified HSP90 inhibitors as a promising new class of senolytic drugs.The accumulation of senescent cells is thought to contribute to the age-associated decline in tissue function. Here, the authors identify HSP90 inhibitors as a new class of senolytic compounds in an in vitro screening and show that administration of a HSP90 inhibitor reduces age-related symptoms in progeroid mice.
Collapse
Affiliation(s)
| | - Yuan Yuan Ling
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Jing Zhao
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Sara J McGowan
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Yi Zhu
- University of Pittsburgh School of Medicine, Pittsburgh, 15261, PA, USA
| | - Robert W Brooks
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Diego Grassi
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Siobhan Q Gregg
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, 55905, MN, USA
| | - Jennifer L Stripay
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, 55905, MN, USA
| | - Akaitz Dorronsoro
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Lana Corbo
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Priscilla Tang
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Christina Bukata
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Nadja Ring
- International Centre for Genetic Engineering and Biotechnology, Trieste, 34100, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology, Trieste, 34100, Italy
| | - Xuesen Li
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Tamara Tchkonia
- University of Pittsburgh School of Medicine, Pittsburgh, 15261, PA, USA
| | - James L Kirkland
- University of Pittsburgh School of Medicine, Pittsburgh, 15261, PA, USA
| | - Laura J Niedernhofer
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA
| | - Paul D Robbins
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, 33458, FL, USA.
| |
Collapse
|
1575
|
Kirkland JL, Tchkonia T, Zhu Y, Niedernhofer LJ, Robbins PD. The Clinical Potential of Senolytic Drugs. J Am Geriatr Soc 2017; 65:2297-2301. [PMID: 28869295 DOI: 10.1111/jgs.14969] [Citation(s) in RCA: 385] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Senolytic drugs are agents that selectively induce apoptosis of senescent cells. These cells accumulate in many tissues with aging and at sites of pathology in multiple chronic diseases. In studies in animals, targeting senescent cells using genetic or pharmacological approaches delays, prevents, or alleviates multiple age-related phenotypes, chronic diseases, geriatric syndromes, and loss of physiological resilience. Among the chronic conditions successfully treated by depleting senescent cells in preclinical studies are frailty, cardiac dysfunction, vascular hyporeactivity and calcification, diabetes mellitus, liver steatosis, osteoporosis, vertebral disk degeneration, pulmonary fibrosis, and radiation-induced damage. Senolytic agents are being tested in proof-of-concept clinical trials. To do so, new clinical trial paradigms for testing senolytics and other agents that target fundamental aging mechanisms are being developed, because use of long-term endpoints such as lifespan or healthspan is not feasible. These strategies include testing effects on multimorbidity, accelerated aging-like conditions, diseases with localized accumulation of senescent cells, potentially fatal diseases associated with senescent cell accumulation, age-related loss of physiological resilience, and frailty. If senolytics or other interventions that target fundamental aging processes prove to be effective and safe in clinical trials, they could transform geriatric medicine by enabling prevention or treatment of multiple diseases and functional deficits in parallel, instead of one at a time.
Collapse
Affiliation(s)
- James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Laura J Niedernhofer
- Department of Molecular Medicine and the Center on Aging, Scripps Research Institute, Jupiter, Florida
| | - Paul D Robbins
- Department of Molecular Medicine and the Center on Aging, Scripps Research Institute, Jupiter, Florida
| |
Collapse
|
1576
|
Parisi MM, Grun LK, Lavandoski P, Alves LB, Bristot IJ, Mattiello R, Mottin CC, Klamt F, Jones MH, Padoin AV, Guma FCR, Barbé-Tuana FM. Immunosenescence Induced by Plasma from Individuals with Obesity Caused Cell Signaling Dysfunction and Inflammation. Obesity (Silver Spring) 2017; 25:1523-1531. [PMID: 28707376 DOI: 10.1002/oby.21888] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 04/06/2017] [Accepted: 04/25/2017] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To evaluate the consequences of plasma from individuals with obesity on parameters associated with immunosenescence in unrelated healthy peripheral blood mononuclear cells (PBMC). METHODS Freshly isolated PBMC were incubated in media supplemented with 10% of plasma from individuals with obesity or control subjects for the first 4 hours of 24 to 120 hours of culture. RESULTS Plasma from individuals with obesity modulated the phenotype of healthy PBMC, leading to a higher rate of apoptosis, lower amounts of phospho-γH2AX and -p53, and mitochondrial dysfunction. After 120 hours, there was a higher secretion of inflammatory cytokines IL-1β and IL-8. CD8+ T lymphocytes presented decreased expression of CD28, which is associated with the immunosenescent phenotype. CD14+ macrophages showed increased expression of CD80 and CD206, suggesting a modulation in the activation of macrophages. CONCLUSIONS These results demonstrate that chronic systemic inflammation observed in obesity induces dysfunctional features in PBMC that are consistent with premature immunosenescence.
Collapse
Affiliation(s)
- Mariana Migliorini Parisi
- Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program of Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lucas Kich Grun
- Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program of Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Patrícia Lavandoski
- Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Letícia Biscaino Alves
- Center of Obesity and Metabolic Syndrome, Hospital São Lucas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ivi Juliana Bristot
- Postgraduate Program of Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Cellular Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rita Mattiello
- Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cláudio Corá Mottin
- Center of Obesity and Metabolic Syndrome, Hospital São Lucas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fábio Klamt
- Postgraduate Program of Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Cellular Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marcus Herbert Jones
- Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Alexandre Vontobel Padoin
- Center of Obesity and Metabolic Syndrome, Hospital São Lucas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fátima Costa Rodrigues Guma
- Laboratory of Biochemistry and Cellular Biology of Lipids, Department of Biochemistry, ICBS/Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Florencia María Barbé-Tuana
- Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program of Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
1577
|
Pathways from senescence to melanoma: focus on MITF sumoylation. Oncogene 2017; 36:6659-6667. [PMID: 28825724 DOI: 10.1038/onc.2017.292] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/09/2017] [Accepted: 07/11/2017] [Indexed: 12/11/2022]
Abstract
Cutaneous melanoma is a deadly skin cancer that originates from melanocytes. The development of cutaneous melanoma involves a complex interaction between environmental factors, mainly ultraviolet radiation from sunlight, and genetic alterations. Melanoma can also occur from a pre-existing nevus, a benign lesion formed from melanocytes harboring oncogenic mutations that trigger proliferative arrest and senescence entry. Senescence is a potent barrier against tumor progression. As such, the acquisition of mutations that suppress senescence and promote cell division is mandatory for cancer development. This topic appears central to melanoma development because, in humans, several somatic and germline mutations are related to the control of cellular senescence and proliferative activity. Consequently, primary melanoma can be viewed as a paradigm of senescence evasion. In support of this notion, a sumoylation-defective germline mutation in microphthalmia-associated transcription factor (MITF), a master regulator of melanocyte homeostasis, is associated with the development of melanoma. Interestingly, this MITF variant has also been recently reported to negatively impact the program of senescence. This article reviews the genetic alterations that have been shown to be involved in melanoma and that alter the process of senescence to favor melanoma development. Then, the transcription factor MITF and its sumoylation-defective mutant are described. How sumoylation misregulation can change MITF activity and impact the process of senescence is discussed. Finally, the contribution of such information to the development of anti-malignant melanoma strategies is evaluated.
Collapse
|
1578
|
Targeting cellular senescence prevents age-related bone loss in mice. Nat Med 2017; 23:1072-1079. [PMID: 28825716 PMCID: PMC5657592 DOI: 10.1038/nm.4385] [Citation(s) in RCA: 860] [Impact Index Per Article: 107.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/13/2017] [Indexed: 11/08/2022]
Abstract
Aging is associated with increased cellular senescence, which is hypothesized to drive the eventual development of multiple comorbidities. Here we investigate a role for senescent cells in age-related bone loss through multiple approaches. In particular, we used either genetic (i.e., the INK-ATTAC 'suicide' transgene encoding an inducible caspase 8 expressed specifically in senescent cells) or pharmacological (i.e., 'senolytic' compounds) means to eliminate senescent cells. We also inhibited the production of the proinflammatory secretome of senescent cells using a JAK inhibitor (JAKi). In aged (20- to 22-month-old) mice with established bone loss, activation of the INK-ATTAC caspase 8 in senescent cells or treatment with senolytics or the JAKi for 2-4 months resulted in higher bone mass and strength and better bone microarchitecture than in vehicle-treated mice. The beneficial effects of targeting senescent cells were due to lower bone resorption with either maintained (trabecular) or higher (cortical) bone formation as compared to vehicle-treated mice. In vitro studies demonstrated that senescent-cell conditioned medium impaired osteoblast mineralization and enhanced osteoclast-progenitor survival, leading to increased osteoclastogenesis. Collectively, these data establish a causal role for senescent cells in bone loss with aging, and demonstrate that targeting these cells has both anti-resorptive and anabolic effects on bone. Given that eliminating senescent cells and/or inhibiting their proinflammatory secretome also improves cardiovascular function, enhances insulin sensitivity, and reduces frailty, targeting this fundamental mechanism to prevent age-related bone loss suggests a novel treatment strategy not only for osteoporosis, but also for multiple age-related comorbidities.
Collapse
|
1579
|
Ngo K, Patil P, McGowan SJ, Niedernhofer LJ, Robbins PD, Kang J, Sowa G, Vo N. Senescent intervertebral disc cells exhibit perturbed matrix homeostasis phenotype. Mech Ageing Dev 2017; 166:16-23. [PMID: 28830687 DOI: 10.1016/j.mad.2017.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/07/2017] [Accepted: 08/14/2017] [Indexed: 12/19/2022]
Abstract
Aging greatly increases the risk for intervertebral disc degeneration (IDD) as a result of proteoglycan loss due to reduced synthesis and enhanced degradation of the disc matrix proteoglycan (PG). How disc matrix PG homeostasis becomes perturbed with age is not known. The goal of this study is to determine whether cellular senescence is a source of this perturbation. We demonstrated that disc cellular senescence is dramatically increased in the DNA repair-deficient Ercc1-/Δ mouse model of human progeria. In these accelerated aging mice, increased disc cellular senescence is closely associated with the rapid loss of disc PG. We also directly examine PG homeostasis in oxidative damage-induced senescent human cells using an in vitro cell culture model system. Senescence of human disc cells treated with hydrogen peroxide was confirmed by growth arrest, senescence-associated β-galactosidase activity, γH2AX foci, and acquisition of senescence-associated secretory phenotype. Senescent human disc cells also exhibited perturbed matrix PG homeostasis as evidenced by their decreased capacity to synthesize new matrix PG and enhanced degradation of aggrecan, a major matrix PG. of the disc. Our in vivo and in vitro findings altogether suggest that disc cellular senescence is an important driver of PG matrix homeostatic perturbation and PG loss.
Collapse
Affiliation(s)
- Kevin Ngo
- Department of Orthopaedic Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Prashanti Patil
- Department of Orthopaedic Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Sara J McGowan
- Department of Molecular Medicine and the TSRI Center on Aging, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Laura J Niedernhofer
- Department of Molecular Medicine and the TSRI Center on Aging, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Paul D Robbins
- Department of Molecular Medicine and the TSRI Center on Aging, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - James Kang
- Department of Orthopaedic Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Orthopaedic Surgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Gwendolyn Sowa
- Department of Orthopaedic Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Physical Medicine and Rehabilitation, University of Pittsburgh, 3471 5th Ave., Pittsburgh, PA 15213, USA
| | - Nam Vo
- Department of Orthopaedic Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| |
Collapse
|
1580
|
Xu M, Bradley EW, Weivoda MM, Hwang SM, Pirtskhalava T, Decklever T, Curran GL, Ogrodnik M, Jurk D, Johnson KO, Lowe V, Tchkonia T, Westendorf JJ, Kirkland JL. Transplanted Senescent Cells Induce an Osteoarthritis-Like Condition in Mice. J Gerontol A Biol Sci Med Sci 2017; 72:780-785. [PMID: 27516624 PMCID: PMC5861939 DOI: 10.1093/gerona/glw154] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/18/2016] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is the leading form of arthritis in the elderly, causing pain, disability, and immobility. OA has been associated with accumulation of senescent cells in or near joints. However, evidence for a causal link between OA and cellular senescence is lacking. Here, we present a novel senescent cell transplantation model involving injection of small numbers of senescent or nonsenescent cells from the ear cartilage of luciferase-expressing mice into the knee joint area of wild-type mice. By using bioluminescence and 18FDG PET imaging, we could track the injected cells in vivo for more than 10 days. Transplanting senescent cells into the knee region caused leg pain, impaired mobility, and radiographic and histological changes suggestive of OA. Transplanting nonsenescent cells had less of these effects. Thus, senescent cells can induce an OA-like state and targeting senescent cells could be a promising strategy for treating OA.
Collapse
Affiliation(s)
- Ming Xu
- Robert and Arlene Kogod Center on Aging
| | | | | | | | | | | | | | - Mikolaj Ogrodnik
- Robert and Arlene Kogod Center on Aging.,Newcastle University Institute for Ageing and Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Diana Jurk
- Newcastle University Institute for Ageing and Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | | | - Val Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | - Jennifer J Westendorf
- Department of Orthopedic Surgery.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
1581
|
Biogenesis of Pro-senescent Microparticles by Endothelial Colony Forming Cells from Premature Neonates is driven by SIRT1-Dependent Epigenetic Regulation of MKK6. Sci Rep 2017; 7:8277. [PMID: 28811647 PMCID: PMC5557933 DOI: 10.1038/s41598-017-08883-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/19/2017] [Indexed: 12/18/2022] Open
Abstract
Senescent cells may exert detrimental effect on microenvironment through the secretion of soluble factors and the release of extracellular vesicles, such as microparticles, key actors in ageing and cardiovascular diseases. We previously reported that sirtuin-1 (SIRT1) deficiency drives accelerated senescence and dysfunction of endothelial colony-forming cells (ECFC) in PT neonates. Because preterm birth (PT) increases the risk for cardiovascular diseases during neonatal period as well as at adulthood, we hypothesized that SIRT1 deficiency could control the biogenesis of microparticles as part of a senescence–associated secretory phenotype (SASP) of PT-ECFC and investigated the related molecular mechanisms. Compared to control ECFC, PT-ECFC displayed a SASP associated with increased release of endothelial microparticles (EMP), mediating a paracrine induction of senescence in naïve endothelial cells. SIRT1 level inversely correlated with EMP release and drives PT-ECFC vesiculation. Global transcriptomic analysis revealed changes in stress response pathways, specifically the MAPK pathway. We delineate a new epigenetic mechanism by which SIRT1 deficiency regulates MKK6/p38MAPK/Hsp27 pathway to promote EMP biogenesis in senescent ECFC. These findings deepen our understanding of the role of ECFC senescence in the disruption of endothelial homeostasis and provide potential new targets towards the control of cardiovascular risk in individuals born preterm.
Collapse
|
1582
|
Jin J, Tao J, Gu X, Yu Z, Wang R, Zuo G, Li Q, Lv X, Miao D. P16 INK4a Deletion Ameliorated Renal Tubulointerstitial Injury in a Stress-induced Premature Senescence Model of Bmi-1 Deficiency. Sci Rep 2017; 7:7502. [PMID: 28790310 PMCID: PMC5548892 DOI: 10.1038/s41598-017-06868-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 06/20/2017] [Indexed: 12/16/2022] Open
Abstract
To determine whether p16 INK4a deletion ameliorated renal tubulointerstitial injury by inhibiting a senescence-associated secretory phenotype (SASP) in Bmi-1-deficient (Bmi-1 -/-) mice, renal phenotypes were compared among 5-week-old Bmi-1 and p16 INK4a double-knockout, and Bmi-1 -/- and wild-type mice. Fifth-passage renal interstitial fibroblasts (RIFs) from the three groups were analyzed for senescence and proliferation. The effect of Bmi-1 deficiency on epithelial-to-mesenchymal transition (EMT) was examined in Bmi-1-knockdown human renal proximal tubular epithelial (HK2) cells, which were treated with concentrated conditioned medium (CM) from the fifth-passage renal interstitial fibroblasts (RIFs) of above three group mice or with exogenous TGF-β1. Our results demonstrated that p16 INK4a deletion largely rescued renal aging phenotypes caused by Bmi-1 deficiency, including impaired renal structure and function, decreased proliferation, increased apoptosis, senescence and SASP, DNA damage, NF-κB and TGF-β1/Smad signal activation, inflammatory cell infiltration, and tubulointerstitial fibrosis and tubular atrophy. P16 INK4a deletion also promoted proliferation, reduced senescence and SASP of RIFs and subsequently inhibited EMT of Bmi-1-knockdown HK2 cells. TGF-β1 further induced the EMT of Bmi-1-knockdown HK2 cells. Thus, p16 INK4a positive senescent cells would be a therapeutic target for preventing renal tubulointerstitial injury.
Collapse
MESH Headings
- Acute Kidney Injury/genetics
- Acute Kidney Injury/metabolism
- Acute Kidney Injury/pathology
- Acute Kidney Injury/prevention & control
- Animals
- Cell Line, Transformed
- Cell Proliferation
- Cellular Senescence
- Coculture Techniques
- Culture Media, Conditioned/pharmacology
- Cyclin-Dependent Kinase Inhibitor p16/deficiency
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Epithelial-Mesenchymal Transition/genetics
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Gene Expression Regulation
- Humans
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Mice
- Mice, Knockout
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/metabolism
- Nephritis, Interstitial/pathology
- Nephritis, Interstitial/prevention & control
- Polycomb Repressive Complex 1/antagonists & inhibitors
- Polycomb Repressive Complex 1/deficiency
- Polycomb Repressive Complex 1/genetics
- Polycomb Repressive Complex 1/metabolism
- Proto-Oncogene Proteins/deficiency
- Proto-Oncogene Proteins/genetics
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Smad Proteins/genetics
- Smad Proteins/metabolism
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/pharmacology
Collapse
Affiliation(s)
- Jianliang Jin
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianguo Tao
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xin Gu
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Zhenzhen Yu
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Rong Wang
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Guoping Zuo
- Laboratory Centre for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Qing Li
- Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, Jiangsu, 210029, China
| | - Xianhui Lv
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Dengshun Miao
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
1583
|
Lehmann M, Korfei M, Mutze K, Klee S, Skronska-Wasek W, Alsafadi HN, Ota C, Costa R, Schiller HB, Lindner M, Wagner DE, Günther A, Königshoff M. Senolytic drugs target alveolar epithelial cell function and attenuate experimental lung fibrosis ex vivo. Eur Respir J 2017; 50:50/2/1602367. [PMID: 28775044 PMCID: PMC5593348 DOI: 10.1183/13993003.02367-2016] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/21/2017] [Indexed: 12/21/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with poor prognosis and limited therapeutic options. The incidence of IPF increases with age, and ageing-related mechanisms such as cellular senescence have been proposed as pathogenic drivers. The lung alveolar epithelium represents a major site of tissue injury in IPF and senescence of this cell population is probably detrimental to lung repair. However, the potential pathomechanisms of alveolar epithelial cell senescence and the impact of senolytic drugs on senescent lung cells and fibrosis remain unknown. Here we demonstrate that lung epithelial cells exhibit increased P16 and P21 expression as well as senescence-associated β-galactosidase activity in experimental and human lung fibrosis tissue and primary cells. Primary fibrotic mouse alveolar epithelial type (AT)II cells secreted increased amounts of senescence-associated secretory phenotype (SASP) factors in vitro, as analysed using quantitative PCR, mass spectrometry and ELISA. Importantly, pharmacological clearance of senescent cells by induction of apoptosis in fibrotic ATII cells or ex vivo three-dimensional lung tissue cultures reduced SASP factors and extracellular matrix markers, while increasing alveolar epithelial markers. These data indicate that alveolar epithelial cell senescence contributes to lung fibrosis development and that senolytic drugs may be a viable therapeutic option for IPF. Alveolar epithelial cell senescence occurs in IPF and senolytic treatment attenuates experimental lung fibrosishttp://ow.ly/nFlz30bsmNm
Collapse
Affiliation(s)
- Mareike Lehmann
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Martina Korfei
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-Universität Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Kathrin Mutze
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Stephan Klee
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Wioletta Skronska-Wasek
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Hani N Alsafadi
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Chiharu Ota
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Rita Costa
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Herbert B Schiller
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Michael Lindner
- Center for Thoracic Surgery, Asklepios Biobank for Lung Diseases, Comprehensive Pneumology Center, Asklepios Clinic Munich-Gauting, Munich, Germany
| | - Darcy E Wagner
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Andreas Günther
- Dept of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-Universität Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany.,Agaplesion Lung Clinic Waldhof Elgershausen, Greifenstein, Germany.,European IPF Network and European IPF Registry
| | - Melanie Königshoff
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München and University Hospital of the Ludwig Maximilians Universität, Member of the German Center for Lung Research (DZL), Munich, Germany .,Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, CO, USA
| |
Collapse
|
1584
|
Robbins PD, Niedernhofer LJ. Advances in Therapeutic Approaches to Extend Healthspan: a perspective from the 2 nd Scripps Symposium on the Biology of Aging. Aging Cell 2017; 16:610-614. [PMID: 28585366 PMCID: PMC5506446 DOI: 10.1111/acel.12620] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2017] [Indexed: 12/18/2022] Open
Abstract
The 2nd Scripps Florida Symposium on The Biology of Aging entitled ‘Advances in Therapeutic Approaches to Extend Healthspan’ was held on January 22nd–25th, 2017 at The Scripps Research Institute in Jupiter, Florida. The meeting highlighted a variety of therapeutic approaches in animal models of aging that either are or soon will be in clinic trials. For example, drugs targeting senescent cells, metformin, rapalogs, NAD precursors, young plasma, mitochondrial‐targeted free radical scavengers, stem cells, and stem cell factors all have shown significant preclinical efficacy. This perspective, based on presentations and discussions at the symposium, outlines the current and future state of development of therapeutic approaches to extend human healthspan.
Collapse
Affiliation(s)
- Paul D. Robbins
- Department of Molecular Medicine and the Center on Aging; The Scripps Research Institute; Jupiter FL 33458 USA
| | - Laura J. Niedernhofer
- Department of Molecular Medicine and the Center on Aging; The Scripps Research Institute; Jupiter FL 33458 USA
| |
Collapse
|
1585
|
He S, Sharpless NE. Senescence in Health and Disease. Cell 2017; 169:1000-1011. [PMID: 28575665 DOI: 10.1016/j.cell.2017.05.015] [Citation(s) in RCA: 1216] [Impact Index Per Article: 152.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 02/07/2023]
Abstract
Many cellular stresses activate senescence, a persistent hyporeplicative state characterized in part by expression of the p16INK4a cell-cycle inhibitor. Senescent cell production occurs throughout life and plays beneficial roles in a variety of physiological and pathological processes including embryogenesis, wound healing, host immunity, and tumor suppression. Meanwhile, the steady accumulation of senescent cells with age also has adverse consequences. These non-proliferating cells occupy key cellular niches and elaborate pro-inflammatory cytokines, contributing to aging-related diseases and morbidity. This model suggests that the abundance of senescent cells in vivo predicts "molecular," as opposed to chronologic, age and that senescent cell clearance may mitigate aging-associated pathology.
Collapse
Affiliation(s)
- Shenghui He
- Departments of Medicine and Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7295, USA; The Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7295, USA
| | - Norman E Sharpless
- Departments of Medicine and Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7295, USA; The Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7295, USA.
| |
Collapse
|
1586
|
Affiliation(s)
- Komal Saraswat
- Department of Biochemistry, University of Allahabad, Allahabad, India
| | | |
Collapse
|
1587
|
Abstract
The incidence and prevalence of cardiac diseases, which are the main cause of death worldwide, are likely to increase because of population ageing. Prevailing theories about the mechanisms of ageing feature the gradual derailment of cellular protein homeostasis (proteostasis) and loss of protein quality control as central factors. In the heart, loss of protein patency, owing to flaws in genetically-determined design or because of environmentally-induced 'wear and tear', can overwhelm protein quality control, thereby triggering derailment of proteostasis and contributing to cardiac ageing. Failure of protein quality control involves impairment of chaperones, ubiquitin-proteosomal systems, autophagy, and loss of sarcomeric and cytoskeletal proteins, all of which relate to induction of cardiomyocyte senescence. Targeting protein quality control to maintain cardiac proteostasis offers a novel therapeutic strategy to promote cardiac health and combat cardiac disease. Currently marketed drugs are available to explore this concept in the clinical setting.
Collapse
Affiliation(s)
- Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, De Boelelaan 1117, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
1588
|
Heiss C, Spyridopoulos I, Haendeler J. Interventions to slow cardiovascular aging: Dietary restriction, drugs and novel molecules. Exp Gerontol 2017; 109:108-118. [PMID: 28658611 DOI: 10.1016/j.exger.2017.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/16/2017] [Accepted: 06/21/2017] [Indexed: 02/05/2023]
Abstract
Cardiovascular aging is a highly dynamic process. Despite the fact that cardiovascular function and structure change with age, they can still be modulated even in aged humans. The most prominent approaches to improve age-dependent vascular changes include dietary restriction and pharmacologic agents interacting with signaling pathways implicated in this context. These include inhibition of TOR, glycolysis, and GH/IGF-1, activation of sirtuins, and AMPK, as well as modulators of inflammation, epigenetic pathways, and telomeres. Promising nutritional approaches include Mediterranean diet and novel dietary bioactives including flavanols, anthocyanins, and lignins. Many plant bioactives improve cardiovascular parameters implied in vascular healthy aging including endothelial function, arterial stiffness, blood pressure, cholesterol, and glycemic control. However, the mechanism of action of most bioactives is not established and it remains to be elucidated whether they act as dietary restriction mimetics or via other modes of action. Even more importantly, whether these interventions can slow or even reverses components of cardiovascular aging itself and can increase healthspan or longevity in humans needs to be determined.
Collapse
Affiliation(s)
- Christian Heiss
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| | - Ioakim Spyridopoulos
- Institute of Genetic Medicine, Medical Faculty, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.
| | - Judith Haendeler
- Central Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, University of Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany.
| |
Collapse
|
1589
|
Lozano-Torres B, Galiana I, Rovira M, Garrido E, Chaib S, Bernardos A, Muñoz-Espín D, Serrano M, Martínez-Máñez R, Sancenón F. An OFF–ON Two-Photon Fluorescent Probe for Tracking Cell Senescence in Vivo. J Am Chem Soc 2017. [DOI: 10.1021/jacs.7b04985] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Beatriz Lozano-Torres
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain
- CIBER de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN)
- Unidad
Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe, Valencia, Spain
| | - Irene Galiana
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain
- CIBER de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN)
- Unidad
Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe, Valencia, Spain
| | - Miguel Rovira
- Tumor
Suppression Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eva Garrido
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain
- CIBER de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN)
- Unidad
Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe, Valencia, Spain
| | - Selim Chaib
- Tumor
Suppression Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Andrea Bernardos
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain
- CIBER de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN)
- Unidad
Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe, Valencia, Spain
| | - Daniel Muñoz-Espín
- Tumor
Suppression Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- CRUK
Cancer Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, U.K
| | - Manuel Serrano
- Tumor
Suppression Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ramón Martínez-Máñez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain
- CIBER de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN)
- Unidad
Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe, Valencia, Spain
| | - Félix Sancenón
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain
- CIBER de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN)
- Unidad
Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe, Valencia, Spain
| |
Collapse
|
1590
|
Baar MP, Brandt RMC, Putavet DA, Klein JDD, Derks KWJ, Bourgeois BRM, Stryeck S, Rijksen Y, van Willigenburg H, Feijtel DA, van der Pluijm I, Essers J, van Cappellen WA, van IJcken WF, Houtsmuller AB, Pothof J, de Bruin RWF, Madl T, Hoeijmakers JHJ, Campisi J, de Keizer PLJ. Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging. Cell 2017; 169:132-147.e16. [PMID: 28340339 DOI: 10.1016/j.cell.2017.02.031] [Citation(s) in RCA: 984] [Impact Index Per Article: 123.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/29/2016] [Accepted: 02/22/2017] [Indexed: 02/06/2023]
Abstract
The accumulation of irreparable cellular damage restricts healthspan after acute stress or natural aging. Senescent cells are thought to impair tissue function, and their genetic clearance can delay features of aging. Identifying how senescent cells avoid apoptosis allows for the prospective design of anti-senescence compounds to address whether homeostasis can also be restored. Here, we identify FOXO4 as a pivot in senescent cell viability. We designed a FOXO4 peptide that perturbs the FOXO4 interaction with p53. In senescent cells, this selectively causes p53 nuclear exclusion and cell-intrinsic apoptosis. Under conditions where it was well tolerated in vivo, this FOXO4 peptide neutralized doxorubicin-induced chemotoxicity. Moreover, it restored fitness, fur density, and renal function in both fast aging XpdTTD/TTD and naturally aged mice. Thus, therapeutic targeting of senescent cells is feasible under conditions where loss of health has already occurred, and in doing so tissue homeostasis can effectively be restored.
Collapse
Affiliation(s)
- Marjolein P Baar
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Diana A Putavet
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Julian D D Klein
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Kasper W J Derks
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Benjamin R M Bourgeois
- Institute of Molecular Biology & Biochemistry, Center of Molecular Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Sarah Stryeck
- Institute of Molecular Biology & Biochemistry, Center of Molecular Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Yvonne Rijksen
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Hester van Willigenburg
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Danny A Feijtel
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands; Department of Vascular Surgery, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands; Department of Vascular Surgery, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands; Department of Radiation Oncology, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Wiggert A van Cappellen
- Erasmus Optical Imaging Center and Department of Pathology, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Wilfred F van IJcken
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Adriaan B Houtsmuller
- Erasmus Optical Imaging Center and Department of Pathology, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Joris Pothof
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Ron W F de Bruin
- Department of Surgery, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Tobias Madl
- Institute of Molecular Biology & Biochemistry, Center of Molecular Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Judith Campisi
- The Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945, USA; Lawrence Berkeley National Laboratories, Berkeley, CA 94720, USA
| | - Peter L J de Keizer
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands; The Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945, USA.
| |
Collapse
|
1591
|
Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, Day CP, Burt A, Palmer A, Anstee QM, Grellscheid SN, Hoeijmakers JHJ, Barnhoorn S, Mann DA, Bird TG, Vermeij WP, Kirkland JL, Passos JF, von Zglinicki T, Jurk D. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun 2017; 8:15691. [PMID: 28608850 PMCID: PMC5474745 DOI: 10.1038/ncomms15691] [Citation(s) in RCA: 726] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 04/20/2017] [Indexed: 02/06/2023] Open
Abstract
The incidence of non-alcoholic fatty liver disease (NAFLD) increases with age. Cellular senescence refers to a state of irreversible cell-cycle arrest combined with the secretion of proinflammatory cytokines and mitochondrial dysfunction. Senescent cells contribute to age-related tissue degeneration. Here we show that the accumulation of senescent cells promotes hepatic fat accumulation and steatosis. We report a close correlation between hepatic fat accumulation and markers of hepatocyte senescence. The elimination of senescent cells by suicide gene-meditated ablation of p16Ink4a-expressing senescent cells in INK-ATTAC mice or by treatment with a combination of the senolytic drugs dasatinib and quercetin (D+Q) reduces overall hepatic steatosis. Conversely, inducing hepatocyte senescence promotes fat accumulation in vitro and in vivo. Mechanistically, we show that mitochondria in senescent cells lose the ability to metabolize fatty acids efficiently. Our study demonstrates that cellular senescence drives hepatic steatosis and elimination of senescent cells may be a novel therapeutic strategy to reduce steatosis.
Collapse
Affiliation(s)
- Mikolaj Ogrodnik
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Satomi Miwa
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Dina Tiniakos
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Pathology, Aretaieio Hospital, Medical School, National & Kapodistrian University of Athens, Athens 11528, Greece
| | - Caroline L. Wilson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Albert Lahat
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Christoper P. Day
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Liver Unit, Newcastle upon Tyne Hospitals NHS Trust, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK
| | - Alastair Burt
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- The University of Adelaide, Faculty of Health Science, North Terrace, Adelaide, South Australia 5005, Australia
| | - Allyson Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Quentin M. Anstee
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | | | - Jan H J. Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
- CECAD Forschungszentrum, Universität zu Köln, Joseph-Stelzmann-Straße 26, Köln 50931, Germany
| | - Sander Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Derek A. Mann
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Thomas G. Bird
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Wilbert P. Vermeij
- Department of Molecular Genetics, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - João F. Passos
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Thomas von Zglinicki
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Diana Jurk
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
1592
|
Salmonowicz H, Passos JF. Detecting senescence: a new method for an old pigment. Aging Cell 2017; 16:432-434. [PMID: 28185406 PMCID: PMC5418201 DOI: 10.1111/acel.12580] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2017] [Indexed: 01/23/2023] Open
Abstract
Cellular senescence is a state of irreversible cell cycle arrest induced by different types of cellular stresses. The field of senescence has made significant advances in the understanding of many of the mechanisms governing this phenomenon; however, a universal biomarker that unambiguously distinguishes senescent from proliferating cells has not been found. In this issue of Aging Cell, Evangelou and colleagues developed a sensitive method for identification of senescent cells in different types of biological material based on the detection of lipofuscin using an analogue of Sudan Black B (SBB) histochemical dye coupled with biotin, which they named GL13. The authors propose that this method is more sensitive and versatile than using SBB alone. Lipofuscin, a nondegradable oxidation product of lipids, proteins and metals, is found in senescent cells. Detection of lipofuscin using GL13 staining may be a more feasible method than others currently used for identification of senescent cells both in cell culture and tissues.
Collapse
Affiliation(s)
- Hanna Salmonowicz
- Institute for Cell and Molecular Biosciences & Newcastle University Institute for Ageing; Newcastle upon Tyne NE4 5PL UK
| | - João F. Passos
- Institute for Cell and Molecular Biosciences & Newcastle University Institute for Ageing; Newcastle upon Tyne NE4 5PL UK
| |
Collapse
|
1593
|
Cortese FAB, Aguiar S, Santostasi G. Induced Cell Turnover: A Novel Therapeutic Modality for In Situ Tissue Regeneration. Hum Gene Ther 2017; 28:703-716. [PMID: 28557533 DOI: 10.1089/hum.2016.167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Induced cell turnover (ICT) is a theoretical intervention in which the targeted ablation of damaged, diseased, and/or nonfunctional cells is coupled with replacement by partially differentiated induced pluripotent stem cells in a gradual and multiphasic manner. Tissue-specific ablation can be achieved using pro-apoptotic small molecule cocktails, peptide mimetics, and/or tissue-tropic adeno-associated virus-delivered suicide genes driven by cell type-specific promoters. Replenishment with new cells can be mediated by systemic administration of cells engineered for homing, robustness, and even enhanced function and disease resistance. Otherwise, the controlled release of cells can be achieved using implanted biodegradable scaffolds, hydrogels, and polymer matrixes. In theory, ICT would enable in situ tissue regeneration without the need for surgical transplantation of organs produced ex vivo, and addresses non-transplantable tissues (such as the vasculature, lymph nodes, and the nervous system). This article outlines several complimentary strategies for overcoming barriers to ICT in an effort to stimulate further research at this promising interface of cell therapy, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Francesco Albert Bosco Cortese
- 1 Biogerontology Research Foundation, Oxford, United Kingdom .,2 Department of Biomedical and Molecular Sciences, Queen's University School of Medicine, Queen's University, Kingston, Canada
| | - Sebastian Aguiar
- 3 Neurobiology Department, Swammerdam Institute of Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Giovanni Santostasi
- 4 Department of Neurology, Feinberg School of Medicine, Northwestern University , Chicago, Illinois
| |
Collapse
|
1594
|
Samaraweera L, Adomako A, Rodriguez-Gabin A, McDaid HM. A Novel Indication for Panobinostat as a Senolytic Drug in NSCLC and HNSCC. Sci Rep 2017; 7:1900. [PMID: 28507307 PMCID: PMC5432488 DOI: 10.1038/s41598-017-01964-1] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 04/07/2017] [Indexed: 01/07/2023] Open
Abstract
Panobinostat (pano) is an FDA-approved histone deacetylase inhibitor. There is interest in evaluating alternate dosing schedules and novel combinations of pano for the treatment of upper aerodigestive and lung malignancies; thus we evaluated it in combination with Taxol, a chemotherapeutic with activity in both diseases. Dose-dependent synergy was observed in Non-Small Cell Lung Cancer (NSCLC) and Head and Neck Squamous Cell Carcinoma (HNSCC) cell lines and was due to senescence rather than potentiation of cell death. Senescence occurred following cisplatin- or Taxol-treatment in cell lines from both cancer types and was associated with decreased histone 3 (H3) acetylation and increased Bcl-xL expression: the latter a biomarker of senescence and target of anti-senescence therapeutics, or senolytics. Since H3 acetylation and Bcl-xL expression were altered in senescence, we subsequently evaluated pano as a senolytic in chemotherapy-treated cancer cells enriched for senescent cells. Pano caused cell death at significantly higher rates compared to repeat dosing with chemotherapy. This was associated with decreased expression of Bcl-xL and increased acetylated H3, reversing the expression patterns observed in senescence. These data support evaluating pano as a post-chemotherapy senolytic with the potential to kill persistent senescent cells that accumulate during standard chemotherapy in NSCLC and HNSCC.
Collapse
Affiliation(s)
- Leleesha Samaraweera
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alfred Adomako
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alicia Rodriguez-Gabin
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Hayley M McDaid
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
1595
|
|
1596
|
Menendez JA, Alarcón T. Senescence-Inflammatory Regulation of Reparative Cellular Reprogramming in Aging and Cancer. Front Cell Dev Biol 2017; 5:49. [PMID: 28529938 PMCID: PMC5418360 DOI: 10.3389/fcell.2017.00049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/18/2017] [Indexed: 12/16/2022] Open
Abstract
The inability of adult tissues to transitorily generate cells with functional stem cell-like properties is a major obstacle to tissue self-repair. Nuclear reprogramming-like phenomena that induce a transient acquisition of epigenetic plasticity and phenotype malleability may constitute a reparative route through which human tissues respond to injury, stress, and disease. However, tissue rejuvenation should involve not only the transient epigenetic reprogramming of differentiated cells, but also the committed re-acquisition of the original or alternative committed cell fate. Chronic or unrestrained epigenetic plasticity would drive aging phenotypes by impairing the repair or the replacement of damaged cells; such uncontrolled phenomena of in vivo reprogramming might also generate cancer-like cellular states. We herein propose that the ability of senescence-associated inflammatory signaling to regulate in vivo reprogramming cycles of tissue repair outlines a threshold model of aging and cancer. The degree of senescence/inflammation-associated deviation from the homeostatic state may delineate a type of thresholding algorithm distinguishing beneficial from deleterious effects of in vivo reprogramming. First, transient activation of NF-κB-related innate immunity and senescence-associated inflammatory components (e.g., IL-6) might facilitate reparative cellular reprogramming in response to acute inflammatory events. Second, para-inflammation switches might promote long-lasting but reversible refractoriness to reparative cellular reprogramming. Third, chronic senescence-associated inflammatory signaling might lock cells in highly plastic epigenetic states disabled for reparative differentiation. The consideration of a cellular reprogramming-centered view of epigenetic plasticity as a fundamental element of a tissue's capacity to undergo successful repair, aging degeneration or malignant transformation should provide challenging stochastic insights into the current deterministic genetic paradigm for most chronic diseases, thereby increasing the spectrum of therapeutic approaches for physiological aging and cancer.
Collapse
Affiliation(s)
- Javier A Menendez
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance, Catalan Institute of OncologyGirona, Spain.,Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI)Girona, Spain.,METABOSTEMBarcelona, Spain
| | - Tomás Alarcón
- Institució Catalana de Recerca i Estudis Avançats (ICREA)Barcelona, Spain.,Computational and Mathematical Biology Research Group, Centre de Recerca MatemàticaBarcelona, Spain.,Departament de Matemàtiques, Universitat Autònoma de BarcelonaBarcelona, Spain.,Barcelona Graduate School of MathematicsBarcelona, Spain
| |
Collapse
|
1597
|
Bettio LEB, Rajendran L, Gil-Mohapel J. The effects of aging in the hippocampus and cognitive decline. Neurosci Biobehav Rev 2017; 79:66-86. [PMID: 28476525 DOI: 10.1016/j.neubiorev.2017.04.030] [Citation(s) in RCA: 404] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/15/2017] [Accepted: 04/10/2017] [Indexed: 02/06/2023]
Abstract
Aging is a natural process that is associated with cognitive decline as well as functional and social impairments. One structure of particular interest when considering aging and cognitive decline is the hippocampus, a brain region known to play an important role in learning and memory consolidation as well as in affective behaviours and mood regulation, and where both functional and structural plasticity (e.g., neurogenesis) occur well into adulthood. Neurobiological alterations seen in the aging hippocampus including increased oxidative stress and neuroinflammation, altered intracellular signalling and gene expression, as well as reduced neurogenesis and synaptic plasticity, are thought to be associated with age-related cognitive decline. Non-invasive strategies such as caloric restriction, physical exercise, and environmental enrichment have been shown to counteract many of the age-induced alterations in hippocampal signalling, structure, and function. Thus, such approaches may have therapeutic value in counteracting the deleterious effects of aging and protecting the brain against age-associated neurodegenerative processes.
Collapse
Affiliation(s)
- Luis E B Bettio
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Luckshi Rajendran
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joana Gil-Mohapel
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; UBC Island Medical program, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
1598
|
Sarcopenic obesity or obese sarcopenia: A cross talk between age-associated adipose tissue and skeletal muscle inflammation as a main mechanism of the pathogenesis. Ageing Res Rev 2017; 35:200-221. [PMID: 27702700 DOI: 10.1016/j.arr.2016.09.008] [Citation(s) in RCA: 519] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/23/2016] [Accepted: 09/26/2016] [Indexed: 02/08/2023]
Abstract
Sarcopenia, an age-associated decline in skeletal muscle mass coupled with functional deterioration, may be exacerbated by obesity leading to higher disability, frailty, morbidity and mortality rates. In the combination of sarcopenia and obesity, the state called sarcopenic obesity (SOB), some key age- and obesity-mediated factors and pathways may aggravate sarcopenia. This review will analyze the mechanisms underlying the pathogenesis of SOB. In obese adipose tissue (AT), adipocytes undergo hypertrophy, hyperplasia and activation resulted in accumulation of pro-inflammatory macrophages and other immune cells as well as dysregulated production of various adipokines that together with senescent cells and the immune cell-released cytokines and chemokines create a local pro-inflammatory status. In addition, obese AT is characterized by excessive production and disturbed capacity to store lipids, which accumulate ectopically in skeletal muscle. These intramuscular lipids and their derivatives induce mitochondrial dysfunction characterized by impaired β-oxidation capacity and increased reactive oxygen species formation providing lipotoxic environment and insulin resistance as well as enhanced secretion of some pro-inflammatory myokines capable of inducing muscle dysfunction by auto/paracrine manner. In turn, by endocrine manner, these myokines may exacerbate AT inflammation and also support chronic low grade systemic inflammation (inflammaging), overall establishing a detrimental vicious circle maintaining AT and skeletal muscle inflammation, thus triggering and supporting SOB development. Under these circumstances, we believe that AT inflammation dominates over skeletal muscle inflammation. Thus, in essence, it redirects the vector of processes from "sarcopenia→obesity" to "obesity→sarcopenia". We therefore propose that this condition be defined as "obese sarcopenia", to reflect the direction of the pathological pathway.
Collapse
|
1599
|
Sritharen Y, Enriquez-Sarano M, Schaff HV, Casaclang-Verzosa G, Miller JD. Pathophysiology of Aortic Valve Stenosis: Is It Both Fibrocalcific and Sex Specific? Physiology (Bethesda) 2017; 32:182-196. [PMID: 28404735 PMCID: PMC6148342 DOI: 10.1152/physiol.00025.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/10/2017] [Accepted: 02/10/2017] [Indexed: 12/24/2022] Open
Abstract
Our understanding of the fundamental biology and identification of efficacious therapeutic targets in aortic valve stenosis has lagged far behind the fields of atherosclerosis and heart failure. In this review, we highlight the most clinically relevant problems facing men and women with fibrocalcific aortic valve stenosis, discuss the fundamental biology underlying valve calcification and fibrosis, and identify key molecular points of intersection with sex hormone signaling.
Collapse
Affiliation(s)
- Yoginee Sritharen
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
| | | | - Hartzell V Schaff
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
| | - Grace Casaclang-Verzosa
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Jordan D Miller
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota;
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Department of Surgery, Mayo Clinic, Rochester, Minnesota; and the
- Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
1600
|
Niedernhofer LJ, Kirkland JL, Ladiges W. Molecular pathology endpoints useful for aging studies. Ageing Res Rev 2017; 35:241-249. [PMID: 27721062 DOI: 10.1016/j.arr.2016.09.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/15/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
Abstract
The first clinical trial aimed at targeting fundamental processes of aging will soon be launched (TAME: Targeting Aging with Metformin). In its wake is a robust pipeline of therapeutic interventions that have been demonstrated to extend lifespan or healthspan of preclinical models, including rapalogs, antioxidants, anti-inflammatory agents, and senolytics. This ensures that if the TAME trial is successful, numerous additional clinical trials are apt to follow. But a significant impediment to these trials remains the question of what endpoints should be measured? The design of the TAME trial very cleverly skirts around this based on the fact that there are decades of data on metformin in humans, providing unequaled clarity of what endpoints are most likely to yield a positive outcome. But for a new chemical entity, knowing what endpoints to measure remains a formidable challenge. For economy's sake, and to achieve results in a reasonable time frame, surrogate markers of lifespan and healthy aging are desperately needed. This review provides a comprehensive analysis of molecular endpoints that are currently being used as indices of age-related phenomena (e.g., morbidity, frailty, mortality) and proposes an approach for validating and prioritizing these endpoints.
Collapse
Affiliation(s)
- L J Niedernhofer
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, United States.
| | - J L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States
| | - W Ladiges
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|