401
|
Yousefzadeh MJ, Melos KI, Angelini L, Burd CE, Robbins PD, Niedernhofer LJ. Mouse Models of Accelerated Cellular Senescence. Methods Mol Biol 2019; 1896:203-230. [PMID: 30474850 DOI: 10.1007/978-1-4939-8931-7_17] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Senescent cells accumulate in multiple tissues as virtually all vertebrate organisms age. Senescence is a highly conserved response to many forms of cellular stress intended to block the propagation of damaged cells. Senescent cells have been demonstrated to play a causal role in aging via their senescence-associated secretory phenotype and by impeding tissue regeneration. Depletion of senescent cells either through genetic or pharmacologic methods has been demonstrated to extend murine lifespan and delay the onset of age-related diseases. Measuring the burden and location of senescent cells in vivo remains challenging, as there is no marker unique to senescent cells. Here, we describe multiple methods to detect the presence and extent of cellular senescence in preclinical models, with a special emphasis on murine models of accelerated aging that exhibit a more rapid onset of cellular senescence.
Collapse
Affiliation(s)
- Matthew J Yousefzadeh
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Kendra I Melos
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Luise Angelini
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Christin E Burd
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
402
|
Colpani O, Spinetti G. MicroRNAs orchestrating senescence of endothelial and vascular smooth muscle cells. ACTA ACUST UNITED AC 2019; 1:H75-H81. [PMID: 32923957 PMCID: PMC7439843 DOI: 10.1530/vb-19-0017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/12/2019] [Indexed: 12/20/2022]
Abstract
During organism aging, the process of cellular senescence is triggered by critical stressors such as DNA damage, oncogenes, oxidative stress, and telomere erosion, and vascular cells are not exempted. Senescent cells stop proliferating but remain metabolically active producing pro-inflammatory signals in the environment collectively named senescence-associated secretory phenotype (SASP) that contribute to the amplification of the response to the neighbor and distant cells. Although the shift toward senescence is protective against tumors and needed during wound healing, the accumulation of senescent cells during aging due to an impairment of the immune system deputed to their clearance, can predispose to diseases of the cardiovascular system such as atherosclerosis. In this short review, we describe the main features of senescence of endothelial and smooth muscle cells and focus on the role non-coding RNAs of the microRNAs class in controlling this process. Finally, we discuss the potential of new strategies based on senescence removal in counteracting vascular disease burden.
Collapse
|
403
|
Cottage CT, Peterson N, Kearley J, Berlin A, Xiong X, Huntley A, Zhao W, Brown C, Migneault A, Zerrouki K, Criner G, Kolbeck R, Connor J, Lemaire R. Targeting p16-induced senescence prevents cigarette smoke-induced emphysema by promoting IGF1/Akt1 signaling in mice. Commun Biol 2019; 2:307. [PMID: 31428695 PMCID: PMC6689060 DOI: 10.1038/s42003-019-0532-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 06/27/2019] [Indexed: 12/16/2022] Open
Abstract
Senescence is a mechanism associated with aging that alters tissue regeneration by depleting the stem cell pool. Chronic obstructive pulmonary disease (COPD) displays hallmarks of senescence, including a diminished stem cell population. DNA damage from cigarette smoke (CS) induces senescence via the p16 pathway. This study evaluated the contribution of p16 to CS-associated lung pathologies. p16 expression was prominent in human COPD lungs compared with normal subjects. CS induces impaired pulmonary function, emphysema, and increased alveolar epithelial cell (AECII) senescence in wild-type mice, whereas CS-exposed p16-/- mice exhibit normal pulmonary function, reduced emphysema, diminished AECII senescence, and increased pro-growth IGF1 signaling, suggesting that improved lung function in p16-/- mice was due to increased alveolar progenitor cell proliferation. In conclusion, our study suggests that targeting senescence may facilitate alveolar regeneration in COPD emphysema by promoting IGF1 proliferative signaling.
Collapse
Affiliation(s)
- Christopher T. Cottage
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Norman Peterson
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Jennifer Kearley
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Aaron Berlin
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Ximing Xiong
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Anna Huntley
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Weiguang Zhao
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Charles Brown
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Annik Migneault
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Kamelia Zerrouki
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | | | - Roland Kolbeck
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Jane Connor
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| | - Raphael Lemaire
- Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878 United States
| |
Collapse
|
404
|
Trott DW, Fadel PJ. Inflammation as a mediator of arterial ageing. Exp Physiol 2019; 104:1455-1471. [PMID: 31325339 DOI: 10.1113/ep087499] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022]
Abstract
NEW FINDINGS What is the topic of this review? This review summarizes and synthesizes what is known about the contribution of inflammation to age-related arterial dysfunction. What advances does it highlight? This review details observational evidence for the relationship of age-related inflammation and arterial dysfunction, insight from autoimmune inflammatory diseases and their effects on arterial function, interventional evidence linking inflammation and age-related arterial dysfunction, insight into age-related arterial inflammation from preclinical models and interventions to ameliorate age-related inflammation and arterial dysfunction. ABSTRACT Advanced age is a primary risk factor for cardiovascular disease, the leading cause of death in the industrialized world. Two major components of arterial ageing are stiffening of the large arteries and impaired endothelium-dependent dilatation in multiple vascular beds. These two alterations are major contributors to the development of overt cardiovascular disease. Increasing inflammation with advanced age is likely to play a role in this arterial dysfunction. The purpose of this review is to synthesize what is known about inflammation and its relationship to age-related arterial dysfunction. This review discusses both the initial observational evidence for the relationship of age-related inflammation and arterial dysfunction and the evidence that inflammatory autoimmune diseases are associated with a premature arterial ageing phenotype. We next discuss interventional and mechanistic evidence linking inflammation and age-related arterial dysfunction in older adults. We also attempt to summarize the relevant evidence from preclinical models. Lastly, we discuss interventions in both humans and animals that have been shown to ameliorate age-related arterial inflammation and dysfunction. The available evidence provides a strong basis for the role of inflammation in both large artery stiffening and impairment of endothelium-dependent dilatation; however, the specific inflammatory mediators, the initiating factors and the relative importance of the endothelium, smooth muscle cells, perivascular adipose tissue and immune cells in arterial inflammation are not well understood. With the expansion of the ageing population, ameliorating age-related arterial inflammation represents an important potential strategy for preserving vascular health in the elderly.
Collapse
Affiliation(s)
- Daniel W Trott
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| | - Paul J Fadel
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
405
|
Schafer MJ, Mazula DL, Brown AK, White TA, Atkinson E, Pearsall VM, Aversa Z, Verzosa GC, Smith LA, Matveyenko A, Miller JD, LeBrasseur NK. Late-life time-restricted feeding and exercise differentially alter healthspan in obesity. Aging Cell 2019; 18:e12966. [PMID: 31111669 PMCID: PMC6612646 DOI: 10.1111/acel.12966] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/09/2019] [Accepted: 04/18/2019] [Indexed: 01/03/2023] Open
Abstract
Aging and obesity increase multimorbidity and disability risk, and determining interventions for reversing healthspan decline is a critical public health priority. Exercise and time‐restricted feeding (TRF) benefit multiple health parameters when initiated in early life, but their efficacy and safety when initiated at older ages are uncertain. Here, we tested the effects of exercise versus TRF in diet‐induced obese, aged mice from 20 to 24 months of age. We characterized healthspan across key domains: body composition, physical, metabolic, and cardiovascular function, activity of daily living (ADL) behavior, and pathology. We demonstrate that both exercise and TRF improved aspects of body composition. Exercise uniquely benefited physical function, and TRF uniquely benefited metabolism, ADL behavior, and circulating indicators of liver pathology. No adverse outcomes were observed in exercised mice, but in contrast, lean mass and cardiovascular maladaptations were observed following TRF. Through a composite index of benefits and risks, we conclude the net healthspan benefits afforded by exercise are more favorable than those of TRF. Extrapolating to obese older adults, exercise is a safe and effective option for healthspan improvement, but additional comprehensive studies are warranted before recommending TRF.
Collapse
Affiliation(s)
- Marissa J. Schafer
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
- Department of Physical Medicine and Rehabilitation Mayo Clinic Rochester Minnesota
| | - Daniel L. Mazula
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
| | - Ashley K. Brown
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
| | - Thomas A. White
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
| | - Elizabeth Atkinson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research Mayo Clinic Rochester Minnesota
| | | | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
- Department of Physical Medicine and Rehabilitation Mayo Clinic Rochester Minnesota
| | | | | | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering Mayo Clinic Rochester Minnesota
| | - Jordan D. Miller
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
- Department of Surgery Mayo Clinic Rochester Minnesota
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
- Department of Physical Medicine and Rehabilitation Mayo Clinic Rochester Minnesota
| |
Collapse
|
406
|
Farr JN, Rowsey JL, Eckhardt BA, Thicke BS, Fraser DG, Tchkonia T, Kirkland JL, Monroe DG, Khosla S. Independent Roles of Estrogen Deficiency and Cellular Senescence in the Pathogenesis of Osteoporosis: Evidence in Young Adult Mice and Older Humans. J Bone Miner Res 2019; 34:1407-1418. [PMID: 30913313 PMCID: PMC6697189 DOI: 10.1002/jbmr.3729] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/27/2019] [Accepted: 03/15/2019] [Indexed: 11/10/2022]
Abstract
Estrogen deficiency is a seminal mechanism in the pathogenesis of osteoporosis. Mounting evidence, however, establishes that cellular senescence, a fundamental mechanism that drives multiple age-related diseases, also causes osteoporosis. Recently, we systematically identified an accumulation of senescent cells, characterized by increased p16Ink4a and p21Cip1 levels and development of a senescence-associated secretory phenotype (SASP), in mouse bone/marrow and human bone with aging. We then demonstrated that elimination of senescent cells prevented age-related bone loss using multiple approaches, eg, treating old mice expressing a "suicide" transgene, INK-ATTAC, with AP20187 to induce apoptosis of p16Ink4a -senescent cells or periodically treating old wild-type mice with "senolytics," ie, drugs that eliminate senescent cells. Here, we investigate a possible role for estrogen in the regulation of cellular senescence using multiple approaches. First, sex steroid deficiency 2 months after ovariectomy (OVX, n = 15) or orchidectomy (ORCH, n = 15) versus sham surgery (SHAM, n = 15/sex) in young adult (4-month-old) wild-type mice did not alter senescence biomarkers or induce a SASP in bone. Next, in elderly postmenopausal women, 3 weeks of estrogen therapy (n = 10; 74 ± 5 years) compared with no treatment (n = 10; 78 ± 5 years) did not alter senescence biomarkers or the SASP in human bone biopsies. Finally, young adult (4-month-old) female INK-ATTAC mice were randomized (n = 17/group) to SHAM+Vehicle, OVX+Vehicle, or OVX+AP20187 for 2 months. As anticipated, OVX+Vehicle caused significant trabecular/cortical bone loss compared with SHAM+Vehicle. However, treatment with AP20187, which eliminates senescent cells in INK-ATTAC mice, did not rescue the OVX-induced bone loss or alter senescence biomarkers. Collectively, our data establish independent roles of estrogen deficiency and cellular senescence in the pathogenesis of osteoporosis, which has important implications for testing novel senolytics for skeletal efficacy, as these drugs will need to be evaluated in preclinical models of aging as opposed to the current FDA model of prevention of OVX-induced bone loss. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Joshua N Farr
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jennifer L Rowsey
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Brittany A Eckhardt
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Brianne S Thicke
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Daniel G Fraser
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - David G Monroe
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
407
|
Wang MJ, Chen J, Chen F, Liu Q, Sun Y, Yan C, Yang T, Bao Y, Hu YP. Rejuvenating Strategies of Tissue-specific Stem Cells for Healthy Aging. Aging Dis 2019; 10:871-882. [PMID: 31440391 PMCID: PMC6675530 DOI: 10.14336/ad.2018.1119] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022] Open
Abstract
Although aging is a physiological process, it has raised interest in the science of aging and rejuvenation because of the increasing burden on the rapidly aging global population. With advanced age, there is a decline in homeostatic maintenance and regenerative responsiveness to the injury of various tissues, thereby contributing to the incidence of age-related diseases. The primary cause of the functional declines that occur along with aging is considered to be the exhaustion of stem cell functions in their corresponding tissues. Age-related changes in the systemic environment, the niche, and stem cells contribute to this loss. Thus, the reversal of stem cell aging at the cellular level might lead to the rejuvenation of the animal at an organismic level and the prevention of aging, which would be critical for developing new therapies for age-related dysfunction and diseases. Here, we will explore the effects of aging on stem cells in different tissues. The focus of this discussion is on pro-youth interventions that target intrinsic stem cell properties, environmental niche component, systemic factors, and senescent cellular clearance, which are promising for developing strategies related to the reversal of aged stem cell function and optimizing tissue repair processes.
Collapse
Affiliation(s)
- Min-Jun Wang
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Jiajia Chen
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Fei Chen
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Qinggui Liu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yu Sun
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Chen Yan
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Tao Yang
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yiwen Bao
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China.,2Department of Diagnostic radiology, University of Hong Kong, Hong Kong 999077, China
| | - Yi-Ping Hu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
408
|
Grootaert MOJ, Moulis M, Roth L, Martinet W, Vindis C, Bennett MR, De Meyer GRY. Vascular smooth muscle cell death, autophagy and senescence in atherosclerosis. Cardiovasc Res 2019; 114:622-634. [PMID: 29360955 DOI: 10.1093/cvr/cvy007] [Citation(s) in RCA: 411] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 01/17/2018] [Indexed: 12/14/2022] Open
Abstract
In the present review, we describe the causes and consequences of loss of vascular smooth muscle cells (VSMCs) or their function in advanced atherosclerotic plaques and discuss possible mechanisms such as cell death or senescence, and induction of autophagy to promote cell survival. We also highlight the potential use of pharmacological modulators of these processes to limit plaque progression and/or improve plaque stability. VSMCs play a pivotal role in atherogenesis. Loss of VSMCs via initiation of cell death leads to fibrous cap thinning and promotes necrotic core formation and calcification. VSMC apoptosis is induced by pro-inflammatory cytokines, oxidized low density lipoprotein, high levels of nitric oxide and mechanical injury. Apoptotic VSMCs are characterized by a thickened basal lamina surrounding the cytoplasmic remnants of the VSMC. Inefficient clearance of apoptotic VSMCs results in secondary necrosis and subsequent inflammation. A critical determinant in the VSMC stress response and phenotypic switching is autophagy, which is activated by various stimuli, including reactive oxygen and lipid species, cytokines, growth factors and metabolic stress. Successful autophagy stimulates VSMC survival, whereas reduced autophagy promotes age-related changes in the vasculature. Recently, an interesting link between autophagy and VSMC senescence has been uncovered. Defective VSMC autophagy accelerates not only the development of stress-induced premature senescence but also atherogenesis, albeit without worsening plaque stability. VSMC senescence in atherosclerosis is likely a result of replicative senescence and/or stress-induced premature senescence in response to DNA damaging and/or oxidative stress-inducing stimuli. The finding that VSMC senescence can promote atherosclerosis further illustrates that normal, adequate VSMC function is crucial in protecting the vessel wall against atherosclerosis.
Collapse
Affiliation(s)
- Mandy O J Grootaert
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Box 110, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Manon Moulis
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases and University Paul Sabatier, F-31342 Toulouse, France
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Cécile Vindis
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases and University Paul Sabatier, F-31342 Toulouse, France
| | - Martin R Bennett
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Box 110, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| |
Collapse
|
409
|
Durham AL, Speer MY, Scatena M, Giachelli CM, Shanahan CM. Role of smooth muscle cells in vascular calcification: implications in atherosclerosis and arterial stiffness. Cardiovasc Res 2019. [PMID: 29514202 PMCID: PMC5852633 DOI: 10.1093/cvr/cvy010] [Citation(s) in RCA: 726] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Vascular calcification is associated with a significant increase in all-cause mortality and atherosclerotic plaque rupture. Calcification has been determined to be an active process driven in part by vascular smooth muscle cell (VSMC) transdifferentiation within the vascular wall. Historically, VSMC phenotype switching has been viewed as binary, with the cells able to adopt a physiological contractile phenotype or an alternate ‘synthetic’ phenotype in response to injury. More recent work, including lineage tracing has however revealed that VSMCs are able to adopt a number of phenotypes, including calcific (osteogenic, chondrocytic, and osteoclastic), adipogenic, and macrophagic phenotypes. Whilst the mechanisms that drive VSMC differentiation are still being elucidated it is becoming clear that medial calcification may differ in several ways from the intimal calcification seen in atherosclerotic lesions, including risk factors and specific drivers for VSMC phenotype changes and calcification. This article aims to compare and contrast the role of VSMCs in driving calcification in both atherosclerosis and in the vessel media focusing on the major drivers of calcification, including aging, uraemia, mechanical stress, oxidative stress, and inflammation. The review also discusses novel findings that have also brought attention to specific pro- and anti-calcifying proteins, extracellular vesicles, mitochondrial dysfunction, and a uraemic milieu as major determinants of vascular calcification.
Collapse
Affiliation(s)
- Andrew L Durham
- Division of Cardiology, James Black Centre, Kings College London, Denmark Hill, London, SE5 9NU, UK
| | - Mei Y Speer
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Marta Scatena
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Cecilia M Giachelli
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Catherine M Shanahan
- Division of Cardiology, James Black Centre, Kings College London, Denmark Hill, London, SE5 9NU, UK
| |
Collapse
|
410
|
McCarthy CG, Wenceslau CF, Webb RC, Joe B. Novel Contributors and Mechanisms of Cellular Senescence in Hypertension-Associated Premature Vascular Aging. Am J Hypertens 2019; 32:709-719. [PMID: 30982879 DOI: 10.1093/ajh/hpz052] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/25/2019] [Accepted: 04/10/2019] [Indexed: 01/08/2023] Open
Abstract
Hypertension has been described as a condition of premature vascular aging, relative to actual chronological age. In fact, many factors that contribute to the deterioration of vascular function as we age are accelerated in hypertension. Nonetheless, the precise mechanisms that underlie the aged phenotype of arteries from hypertensive patients and animals remain elusive. Cellular senescence is an age-related physiologic process in which cells undergo irreversible growth arrest. Although controlled senescence negatively regulates cell proliferation and promotes tissue regeneration, uncontrolled senescence can contribute to disease pathogenesis by presenting the senescence-associated secretory phenotype, in which molecules such as proinflammatory cytokines, matrix metalloproteases, and reactive oxygen species are released into tissue microenvironments. This review will address and critically evaluate the current literature on the role of cellular senescence in hypertension, with particular emphasis on cells types that mediate and modulate vascular function and structure.
Collapse
Affiliation(s)
- Cameron G McCarthy
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Camilla F Wenceslau
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, Georgia, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
411
|
Knoppert SN, Valentijn FA, Nguyen TQ, Goldschmeding R, Falke LL. Cellular Senescence and the Kidney: Potential Therapeutic Targets and Tools. Front Pharmacol 2019; 10:770. [PMID: 31354486 PMCID: PMC6639430 DOI: 10.3389/fphar.2019.00770] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasing health burden (affecting approximately 13.4% of the population). Currently, no curative treatment options are available and treatment is focused on limiting the disease progression. The accumulation of senescent cells has been implicated in the development of kidney fibrosis by limiting tissue rejuvenation and through the secretion of pro-fibrotic and pro-inflammatory mediators termed as the senescence-associated secretory phenotype. The clearance of senescent cells in aging models results in improved kidney function, which shows promise for the options of targeting senescent cells in CKD. There are several approaches for the development of “senotherapies”, the most rigorous of which is the elimination of senescent cells by the so-called senolytic drugs either newly developed or repurposed for off-target effects in terms of selectively inducing apoptosis in senescent cells. Several chemotherapeutics and checkpoint inhibitors currently used in daily oncological practice show senolytic properties. However, the applicability of such senolytic compounds for the treatment of renal diseases has hardly been investigated. A serious concern is that systemic side effects will limit the use of senolytics for kidney fibrosis. Specifically targeting senescent cells and/or targeted drug delivery to the kidney might circumvent these side effects. In this review, we discuss the connection between CKD and senescence, the pharmacological options for targeting senescent cells, and the means to specifically target the kidney.
Collapse
Affiliation(s)
- Sebastian N Knoppert
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Floris A Valentijn
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Internal Medicine, Diakonessenhuis, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
412
|
Soto-Gamez A, Quax WJ, Demaria M. Regulation of Survival Networks in Senescent Cells: From Mechanisms to Interventions. J Mol Biol 2019; 431:2629-2643. [DOI: 10.1016/j.jmb.2019.05.036] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 01/10/2023]
|
413
|
Chapman J, Fielder E, Passos JF. Mitochondrial dysfunction and cell senescence: deciphering a complex relationship. FEBS Lett 2019; 593:1566-1579. [PMID: 31211858 DOI: 10.1002/1873-3468.13498] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/31/2022]
Abstract
Cellular senescence and mitochondrial dysfunction have both been defined as classical hallmarks of the ageing process. Here, we review the intricate relationship between the two. In the context of ageing, it is now well regarded that cellular senescence is a key driver in both ageing and the onset of a number of age-related pathologies. Emerging evidence has pinpointed mitochondria as one of the key modulators in the development of the senescence phenotype, particularly the pro-inflammatory senescence associated secretory phenotype (SASP). This review focuses on the contribution of homeostatic mechanisms, as well as of reactive oxygen species and mitochondrial metabolites in the senescence programme. Furthermore, we discuss emerging pathways and mitochondrial-mediated mechanisms that may be influencing the SASP and, subsequently, explore how these may be exploited to open up new therapeutic avenues.
Collapse
Affiliation(s)
- James Chapman
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Edward Fielder
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - João F Passos
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Department of Physiology and Biochemical Engineering, Mayo Clinic, Rochester, NY, USA
| |
Collapse
|
414
|
Lipotoxicity, aging, and muscle contractility: does fiber type matter? GeroScience 2019; 41:297-308. [PMID: 31227962 DOI: 10.1007/s11357-019-00077-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/05/2019] [Indexed: 12/18/2022] Open
Abstract
Sarcopenia is a universal characteristic of the aging process and is often accompanied by increases in whole-body adiposity. These changes in body composition have important clinical implications, given that loss of muscle and gain of fat mass are both significantly and independently associated with declining physical performance as well as an increased risk for disability, hospitalizations, and mortality in older individuals. This increased fat mass is not exclusively stored in adipose depots but may become deposited in non-adipose tissues, such as skeletal muscle, when the oxidative capacity of the adipose tissue itself is exceeded. The redistributed adipose tissue is thought to exert detrimental local effects on the muscle environment given the close proximity. Thus, sarcopenia observed with aging may be better defined in the context of loss of muscle quality rather than loss of muscle quantity per se. In this perspective, we briefly review the age-related physiological changes in cellularity, secretory profiles, and inflammatory status of adipose tissue which drive lipotoxicity (spillover) of skeletal muscle and then provide evidence of how this may affect specific fiber type contractility. We focus on biological contributors (cellular machinery) to contractility for which there is some evidence of vulnerability to lipid stress distinguishing between fiber types.
Collapse
|
415
|
Masaldan S, Belaidi AA, Ayton S, Bush AI. Cellular Senescence and Iron Dyshomeostasis in Alzheimer's Disease. Pharmaceuticals (Basel) 2019; 12:E93. [PMID: 31248150 PMCID: PMC6630536 DOI: 10.3390/ph12020093] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023] Open
Abstract
Iron dyshomeostasis is a feature of Alzheimer's disease (AD). The impact of iron on AD is attributed to its interactions with the central proteins of AD pathology (amyloid precursor protein and tau) and/or through the iron-mediated generation of prooxidant molecules (e.g., hydroxyl radicals). However, the source of iron accumulation in pathologically relevant regions of the brain and its contribution to AD remains unclear. One likely contributor to iron accumulation is the age-associated increase in tissue-resident senescent cells that drive inflammation and contribute to various pathologies associated with advanced age. Iron accumulation predisposes ageing tissue to oxidative stress that can lead to cellular dysfunction and to iron-dependent cell death modalities (e.g., ferroptosis). Further, elevated brain iron is associated with the progression of AD and cognitive decline. Elevated brain iron presents a feature of AD that may be modified pharmacologically to mitigate the effects of age/senescence-associated iron dyshomeostasis and improve disease outcome.
Collapse
Affiliation(s)
- Shashank Masaldan
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - Abdel Ali Belaidi
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| |
Collapse
|
416
|
Dai L, Qureshi AR, Witasp A, Lindholm B, Stenvinkel P. Early Vascular Ageing and Cellular Senescence in Chronic Kidney Disease. Comput Struct Biotechnol J 2019; 17:721-729. [PMID: 31303976 PMCID: PMC6603301 DOI: 10.1016/j.csbj.2019.06.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/08/2019] [Accepted: 06/11/2019] [Indexed: 01/08/2023] Open
Abstract
Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by progressive vascular disease, systemic inflammation, muscle wasting and frailty. The predominant early vascular ageing (EVA) process mediated by medial vascular calcification (VC) results in a marked discrepancy between chronological and biological vascular age in CKD. Though the exact underlying mechanisms of VC and EVA are not fully elucidated, accumulating evidence indicates that cellular senescence - and subsequent chronic inflammation through the senescence-associated secretary phenotype (SASP) - plays a fundamental role in its initiation and progression. In this review, we discuss the pathophysiological links between senescence and the EVA process in CKD, with focus on cellular senescence and media VC, and potential anti-ageing therapeutic strategies of senolytic drugs targeting cellular senescence and EVA in CKD.
Collapse
Affiliation(s)
| | | | | | | | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| |
Collapse
|
417
|
|
418
|
Abstract
Replicative capacity of somatic cells is limited. It indicates that aging also develops at the cellular level, and this is described as "cellular senescence". Senescent cells become flattened, enlarged, and irreversibly lose capacity for proliferation. Lack of specific and conclusive markers for cellular senescence makes it difficult to comprehensively define and understand this biological process especially in vivo. Molecules including p53, p21, p16Ink4a, p38MAPK, and γH2AX, telomere attrition, enhanced signals for SA-β-gal, etc. are widely used to detect senescent cells, but these are indirect indicators of cellular senescence, and biological markers reflecting direct evidence need to be established. Genetic profiles are altered in senescent cells, letting these cells secrete pro-inflammatory molecules. Aging or age-related disorders including heart failure and atherosclerotic diseases link with an accumulation of cells undergoing cellular senescence in cardiovascular systems including heart and vessels. Senescent cells become pathogenic in most cases by mediating chronic sterile inflammation and tissue remodeling. A recent conceptual as well as technical breakthrough in this research area is "senolysis", meaning the specific elimination of senescent cells. Genetic as well as pharmacological models with senolysis contributed to reverse aging phenotypes and ameliorated pathologies in age-related disorders without enhancing the risk of tumorigenesis, and opened a new avenue for aging research. Several compounds are identified as senolytics, and some are already tested in clinical settings. It was recently reported that senolysis reverses aging phenotype in cardiovascular disorders. Generating therapies targeting suppression or elimination of senescent cells would inhibit the progression of undesirable aspects of aging, and become promising therapies for cardiac diseases.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| |
Collapse
|
419
|
Muñoz DP, Yannone SM, Daemen A, Sun Y, Vakar-Lopez F, Kawahara M, Freund AM, Rodier F, Wu JD, Desprez PY, Raulet DH, Nelson PS, van ’t Veer LJ, Campisi J, Coppé JP. Targetable mechanisms driving immunoevasion of persistent senescent cells link chemotherapy-resistant cancer to aging. JCI Insight 2019; 5:124716. [PMID: 31184599 PMCID: PMC6675550 DOI: 10.1172/jci.insight.124716] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 06/05/2019] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence is a tumor suppressive mechanism that can paradoxically contribute to aging pathologies. Despite evidence of immune clearance in mouse models, it is not known how senescent cells (SnCs) persist and accumulate with age or in tumors in individuals. Here, we identify cooperative mechanisms that orchestrate the immunoevasion and persistence of normal and cancer human SnCs through extracellular targeting of natural killer receptor signaling. Damaged SnCs avoid immune recognition through MMPs-dependent shedding of NKG2D-ligands reinforced via paracrine suppression of NKG2D receptor-mediated immunosurveillance. These coordinated immunoediting processes are evident in residual, drug-resistant tumors from cohorts of >700 prostate and breast cancer patients treated with senescence-inducing genotoxic chemotherapies. Unlike in mice, these reversible senescence-subversion mechanisms are independent of p53/p16 and exacerbated in oncogenic RAS-induced senescence. Critically, the p16INK4A tumor suppressor can disengage the senescence growth arrest from the damage-associated immune senescence program, which is manifest in benign nevi lesions where indolent SnCs accumulate over time and preserve a non-pro-inflammatory tissue microenvironment maintaining NKG2D-mediated immunosurveillance. Our study shows how subpopulations of SnCs elude immunosurveillance, and reveals secretome-targeted therapeutic strategies to selectively eliminate -and restore the clearance of- the detrimental SnCs that actively persist after chemotherapy and accumulate at sites of aging pathologies.
Collapse
Affiliation(s)
- Denise P. Muñoz
- Swim Across America National Laboratory, Children’s Hospital Oakland Research Institute, UCSF Benioff Children’s Hospital Oakland, Oakland, California, USA
| | - Steven M. Yannone
- Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, California, USA
| | - Anneleen Daemen
- Helen Diller Family Comprehensive Cancer Center, Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Yu Sun
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Funda Vakar-Lopez
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Misako Kawahara
- Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, California, USA
- Helen Diller Family Comprehensive Cancer Center, Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Adam M. Freund
- Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, California, USA
- Buck Institute for Research on Aging, Novato, California, USA
| | - Francis Rodier
- Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, California, USA
| | - Jennifer D. Wu
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Pierre-Yves Desprez
- Buck Institute for Research on Aging, Novato, California, USA
- Research Institute, California Pacific Medical Center, San Francisco, California, USA
| | - David H. Raulet
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Laura J. van ’t Veer
- Helen Diller Family Comprehensive Cancer Center, Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Judith Campisi
- Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, California, USA
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jean-Philippe Coppé
- Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, California, USA
- Helen Diller Family Comprehensive Cancer Center, Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
420
|
Walaszczyk A, Dookun E, Redgrave R, Tual‐Chalot S, Victorelli S, Spyridopoulos I, Owens A, Arthur HM, Passos JF, Richardson GD. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell 2019; 18:e12945. [PMID: 30920115 PMCID: PMC6516151 DOI: 10.1111/acel.12945] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/13/2019] [Accepted: 02/16/2019] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease is the leading cause of death in individuals over 60 years old. Aging is associated with an increased prevalence of coronary artery disease and a poorer prognosis following acute myocardial infarction (MI). With age, senescent cells accumulate in tissues, including the heart, and contribute to age‐related pathologies. However, the role of senescence in recovery following MI has not been investigated. In this study, we demonstrate that treatment of aged mice with the senolytic drug, navitoclax, eliminates senescent cardiomyocytes and attenuates profibrotic protein expression in aged mice. Importantly, clearance of senescent cells improved myocardial remodelling and diastolic function as well as overall survival following MI. These data provide proof‐of‐concept evidence that senescent cells are major contributors to impaired function and increased mortality following MI and that senolytics are a potential new therapeutic avenue for MI.
Collapse
Affiliation(s)
- Anna Walaszczyk
- Cardiovascular Research Centre, Institute of Genetic Medicine Newcastle University Newcastle upon Tyne UK
| | - Emily Dookun
- Cardiovascular Research Centre, Institute of Genetic Medicine Newcastle University Newcastle upon Tyne UK
| | - Rachael Redgrave
- Cardiovascular Research Centre, Institute of Genetic Medicine Newcastle University Newcastle upon Tyne UK
| | - Simon Tual‐Chalot
- Cardiovascular Research Centre, Institute of Genetic Medicine Newcastle University Newcastle upon Tyne UK
| | - Stella Victorelli
- Institute for Cell and Molecular Biosciences Newcastle University Newcastle upon Tyne UK
- Department of Physiology and Biomedical Engineering Mayo Clinic Rochester Minnesota
| | - Ioakim Spyridopoulos
- Cardiovascular Research Centre, Institute of Genetic Medicine Newcastle University Newcastle upon Tyne UK
| | - Andrew Owens
- Cardiovascular Research Centre, Institute of Genetic Medicine Newcastle University Newcastle upon Tyne UK
| | - Helen M. Arthur
- Cardiovascular Research Centre, Institute of Genetic Medicine Newcastle University Newcastle upon Tyne UK
| | - João F. Passos
- Institute for Cell and Molecular Biosciences Newcastle University Newcastle upon Tyne UK
- Department of Physiology and Biomedical Engineering Mayo Clinic Rochester Minnesota
| | - Gavin D. Richardson
- Cardiovascular Research Centre, Institute of Genetic Medicine Newcastle University Newcastle upon Tyne UK
| |
Collapse
|
421
|
Trendelenburg A, Scheuren A, Potter P, Müller R, Bellantuono I. Geroprotectors: A role in the treatment of frailty. Mech Ageing Dev 2019; 180:11-20. [DOI: 10.1016/j.mad.2019.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 12/25/2022]
|
422
|
Lewis‐McDougall FC, Ruchaya PJ, Domenjo‐Vila E, Shin Teoh T, Prata L, Cottle BJ, Clark JE, Punjabi PP, Awad W, Torella D, Tchkonia T, Kirkland JL, Ellison‐Hughes GM. Aged-senescent cells contribute to impaired heart regeneration. Aging Cell 2019; 18:e12931. [PMID: 30854802 PMCID: PMC6516154 DOI: 10.1111/acel.12931] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/09/2019] [Accepted: 01/31/2019] [Indexed: 12/19/2022] Open
Abstract
Aging leads to increased cellular senescence and is associated with decreased potency of tissue-specific stem/progenitor cells. Here, we have done an extensive analysis of cardiac progenitor cells (CPCs) isolated from human subjects with cardiovascular disease, aged 32-86 years. In aged subjects (>70 years old), over half of CPCs are senescent (p16INK4A , SA-β-gal, DNA damage γH2AX, telomere length, senescence-associated secretory phenotype [SASP]), unable to replicate, differentiate, regenerate or restore cardiac function following transplantation into the infarcted heart. SASP factors secreted by senescent CPCs renders otherwise healthy CPCs to senescence. Elimination of senescent CPCs using senolytics abrogates the SASP and its debilitative effect in vitro. Global elimination of senescent cells in aged mice (INK-ATTAC or wild-type mice treated with D + Q senolytics) in vivo activates resident CPCs and increased the number of small Ki67-, EdU-positive cardiomyocytes. Therapeutic approaches that eliminate senescent cells may alleviate cardiac deterioration with aging and restore the regenerative capacity of the heart.
Collapse
Affiliation(s)
- Fiona C. Lewis‐McDougall
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - Prashant J. Ruchaya
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - Eva Domenjo‐Vila
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - Tze Shin Teoh
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - Larissa Prata
- Robert and Arlene Kogod Center on AgingMayo Clinic College of MedicineRochesterMinnesota
| | - Beverley J. Cottle
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - James E. Clark
- School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | | | | | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Medical and Surgical SciencesMagna Graecia UniversityCatanzaroItaly
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on AgingMayo Clinic College of MedicineRochesterMinnesota
| | - James L. Kirkland
- Robert and Arlene Kogod Center on AgingMayo Clinic College of MedicineRochesterMinnesota
| | - Georgina M. Ellison‐Hughes
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| |
Collapse
|
423
|
Cohen J, Torres C. Astrocyte senescence: Evidence and significance. Aging Cell 2019; 18:e12937. [PMID: 30815970 PMCID: PMC6516680 DOI: 10.1111/acel.12937] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/14/2019] [Accepted: 02/04/2019] [Indexed: 12/12/2022] Open
Abstract
Astrocytes participate in numerous aspects of central nervous system (CNS) physiology ranging from ion balance to metabolism, and disruption of their physiological roles can therefore be a contributor to CNS dysfunction and pathology. Cellular senescence, one of the mechanisms of aging, has been proposed as a central component of the age dependency of neurodegenerative disorders. Cumulative evidence supports an integral role of astrocytes in the initiation and progression of neurodegenerative disease and cognitive decline with aging. The loss of astrocyte function or the gain of neuroinflammatory function as a result of cellular senescence could have profound implications for the aging brain and neurodegenerative disorders, and we propose the term “astrosenescence” to describe this phenotype. This review summarizes the current evidence pertaining to astrocyte senescence from early evidence, in vitro characterization and relationship to age‐related neurodegenerative disease. We discuss the significance of targeting senescent astrocytes as a novel approach toward therapies for age‐associated neurodegenerative disease.
Collapse
Affiliation(s)
- Justin Cohen
- Department of Pathology and Laboratory Medicine Drexel University College of Medicine Philadelphia Pennsylvania
| | - Claudio Torres
- Department of Pathology and Laboratory Medicine Drexel University College of Medicine Philadelphia Pennsylvania
| |
Collapse
|
424
|
Palmer AK, Xu M, Zhu Y, Pirtskhalava T, Weivoda MM, Hachfeld CM, Prata LG, van Dijk TH, Verkade E, Casaclang‐Verzosa G, Johnson KO, Cubro H, Doornebal EJ, Ogrodnik M, Jurk D, Jensen MD, Chini EN, Miller JD, Matveyenko A, Stout MB, Schafer MJ, White TA, Hickson LJ, Demaria M, Garovic V, Grande J, Arriaga EA, Kuipers F, von Zglinicki T, LeBrasseur NK, Campisi J, Tchkonia T, Kirkland JL. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell 2019; 18:e12950. [PMID: 30907060 PMCID: PMC6516193 DOI: 10.1111/acel.12950] [Citation(s) in RCA: 434] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/16/2019] [Accepted: 03/03/2019] [Indexed: 12/18/2022] Open
Abstract
Adipose tissue inflammation and dysfunction are associated with obesity-related insulin resistance and diabetes, but mechanisms underlying this relationship are unclear. Although senescent cells accumulate in adipose tissue of obese humans and rodents, a direct pathogenic role for these cells in the development of diabetes remains to be demonstrated. Here, we show that reducing senescent cell burden in obese mice, either by activating drug-inducible "suicide" genes driven by the p16Ink4a promoter or by treatment with senolytic agents, alleviates metabolic and adipose tissue dysfunction. These senolytic interventions improved glucose tolerance, enhanced insulin sensitivity, lowered circulating inflammatory mediators, and promoted adipogenesis in obese mice. Elimination of senescent cells also prevented the migration of transplanted monocytes into intra-abdominal adipose tissue and reduced the number of macrophages in this tissue. In addition, microalbuminuria, renal podocyte function, and cardiac diastolic function improved with senolytic therapy. Our results implicate cellular senescence as a causal factor in obesity-related inflammation and metabolic derangements and show that emerging senolytic agents hold promise for treating obesity-related metabolic dysfunction and its complications.
Collapse
|
425
|
|
426
|
Chromosomal instability and pro-inflammatory response in aging. Mech Ageing Dev 2019; 182:111118. [PMID: 31102604 DOI: 10.1016/j.mad.2019.111118] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/25/2019] [Accepted: 05/14/2019] [Indexed: 01/10/2023]
Abstract
Aging refers to the progressive deterioration of tissue and organ function over time. Increasing evidence points to the accumulation of highly damaged cell cycle-arrested cells with age (cellular senescence) as major reason for the development of certain aging-associated diseases. Recent studies have independently shown that aneuploidy, an abnormal chromosome set, occurs in senescent cells, and that the accumulation of cytoplasmic DNA driven by faulty chromosome segregation during mitosis aids in the establishment of senescence and its associated secretory phenotype known as SASP. Here we review the emerging link between chromosomal instability (CIN) and senescence in the context of aging, with emphasis on the cGAS-STING pathway activation and its role in the development of the SASP. Based on current evidence, we propose that age-associated CIN in mitotically active cells contributes to aging and its associated diseases, and we discuss the inhibition of CIN as a potential strategy to prevent the generation of aneuploid senescent cells and thereby to delay aging.
Collapse
|
427
|
Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Krüger P, Fielder E, Victorelli S, Ruswhandi RA, Giorgadze N, Pirtskhalava T, Podgorni O, Enikolopov G, Johnson KO, Xu M, Inman C, Palmer AK, Schafer M, Weigl M, Ikeno Y, Burns TC, Passos JF, von Zglinicki T, Kirkland JL, Jurk D. Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis. Cell Metab 2019; 29:1061-1077.e8. [PMID: 30612898 PMCID: PMC6509403 DOI: 10.1016/j.cmet.2018.12.008] [Citation(s) in RCA: 315] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 10/23/2018] [Accepted: 12/05/2018] [Indexed: 12/25/2022]
Abstract
Cellular senescence entails a stable cell-cycle arrest and a pro-inflammatory secretory phenotype, which contributes to aging and age-related diseases. Obesity is associated with increased senescent cell burden and neuropsychiatric disorders, including anxiety and depression. To investigate the role of senescence in obesity-related neuropsychiatric dysfunction, we used the INK-ATTAC mouse model, from which p16Ink4a-expressing senescent cells can be eliminated, and senolytic drugs dasatinib and quercetin. We found that obesity results in the accumulation of senescent glial cells in proximity to the lateral ventricle, a region in which adult neurogenesis occurs. Furthermore, senescent glial cells exhibit excessive fat deposits, a phenotype we termed "accumulation of lipids in senescence." Clearing senescent cells from high fat-fed or leptin receptor-deficient obese mice restored neurogenesis and alleviated anxiety-related behavior. Our study provides proof-of-concept evidence that senescent cells are major contributors to obesity-induced anxiety and that senolytics are a potential new therapeutic avenue for treating neuropsychiatric disorders.
Collapse
Affiliation(s)
- Mikolaj Ogrodnik
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Larissa G P Langhi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Patrick Krüger
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Edward Fielder
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Stella Victorelli
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Rifqha A Ruswhandi
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Nino Giorgadze
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Tamar Pirtskhalava
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Oleg Podgorni
- Department of Anesthesiology, Stony Brook School of Medicine, 101 Nicolls Road, Stony Brook, New York, NY 11794, USA; Center for Developmental Genetics, Stony Brook University, 100 Nicolls Road, Stony Brook, New York, NY 11794, USA
| | - Grigori Enikolopov
- Department of Anesthesiology, Stony Brook School of Medicine, 101 Nicolls Road, Stony Brook, New York, NY 11794, USA; Center for Developmental Genetics, Stony Brook University, 100 Nicolls Road, Stony Brook, New York, NY 11794, USA; Department of Nano-, Bio-, Information Technology and Cognitive Science, Moscow Institute of Physics and Technology, Moscow, Russia; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Kurt O Johnson
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Ming Xu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Christine Inman
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Marissa Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Moritz Weigl
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Yuji Ikeno
- The Barshop Institute for Longevity and Aging Studies, San Antonio, Department of Pathology, The University of Texas Health Science Center at San Antonio, Research Service, Audie L. Murphy VA Hospital (STVHCS), San Antonio, TX 78229, USA
| | - Terry C Burns
- Departments of Neurologic Surgery and Neuroscience, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - João F Passos
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Thomas von Zglinicki
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Near East University, Arts and Sciences Faculty, Molecular Biology and Genetics, Nicosia, North Cyprus POB 99138 Mersin 10, Turkey
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | - Diana Jurk
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
428
|
Abstract
Cellular senescence (CS) is one of hallmarks of aging and accumulation of senescent cells (SCs) with age contributes to tissue or organismal aging, as well as the pathophysiologies of diverse age-related diseases (ARDs). Genetic ablation of SCs in tissues lengthened health span and reduced the risk of age-related pathologies in a mouse model, suggesting a direct link between SCs, longevity, and ARDs. Therefore, senotherapeutics, medicines targeting SCs, might be an emerging strategy for the extension of health span, and prevention or treatment of ARDs. Senotherapeutics are classified as senolytics which kills SCs selectively; senomorphics which modulate functions and morphology of SCs to those of young cells, or delays the progression of young cells to SCs in tissues; and immune-system mediators of the clearance of SCs. Some senolytics and senomorphics have been proven to markedly prevent or treat ARDs in animal models. This review will present the current status of the development of senotherapeutics, in relation to aging itself and ARDs. Finally, future directions and opportunities for senotherapeutics use will discussed. This knowledge will provide information that can be used to develop novel senotherapeutics for health span and ARDs. [BMB Reports 2019; 52(1): 47-55].
Collapse
Affiliation(s)
- Eok-Cheon Kim
- Department of Biochemistry and Molecular Biology, Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu 42415, Korea
| | - Jae-Ryong Kim
- Department of Biochemistry and Molecular Biology, Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu 42415, Korea
| |
Collapse
|
429
|
Fulop GA, Tarantini S, Yabluchanskiy A, Molnar A, Prodan CI, Kiss T, Csipo T, Lipecz A, Balasubramanian P, Farkas E, Toth P, Sorond F, Csiszar A, Ungvari Z. Role of age-related alterations of the cerebral venous circulation in the pathogenesis of vascular cognitive impairment. Am J Physiol Heart Circ Physiol 2019; 316:H1124-H1140. [PMID: 30848677 PMCID: PMC6580383 DOI: 10.1152/ajpheart.00776.2018] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/31/2019] [Accepted: 02/18/2019] [Indexed: 02/07/2023]
Abstract
There has been an increasing appreciation of the role of vascular contributions to cognitive impairment and dementia (VCID) associated with old age. Strong preclinical and translational evidence links age-related dysfunction and structural alterations of the cerebral arteries, arterioles, and capillaries to the pathogenesis of many types of dementia in the elderly, including Alzheimer's disease. The low-pressure, low-velocity, and large-volume venous circulation of the brain also plays critical roles in the maintenance of homeostasis in the central nervous system. Despite its physiological importance, the role of age-related alterations of the brain venous circulation in the pathogenesis of vascular cognitive impairment and dementia is much less understood. This overview discusses the role of cerebral veins in the pathogenesis of VCID. Pathophysiological consequences of age-related dysregulation of the cerebral venous circulation are explored, including blood-brain barrier disruption, neuroinflammation, exacerbation of neurodegeneration, development of cerebral microhemorrhages of venous origin, altered production of cerebrospinal fluid, impaired function of the glymphatics system, dysregulation of cerebral blood flow, and ischemic neuronal dysfunction and damage. Understanding the age-related functional and phenotypic alterations of the cerebral venous circulation is critical for developing new preventive, diagnostic, and therapeutic approaches to preserve brain health in older individuals.
Collapse
Affiliation(s)
- Gabor A Fulop
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Heart and Vascular Center, Semmelweis University , Budapest , Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Andrea Molnar
- Heart and Vascular Center, Semmelweis University , Budapest , Hungary
| | - Calin I Prodan
- Veterans Affairs Medical Center , Oklahoma City, Oklahoma
- Department of Neurology, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Vascular Cognitive Impairment Program, Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Agnes Lipecz
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Eszter Farkas
- Vascular Cognitive Impairment Program, Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Cerebrovascular Laboratory, Department of Neurosurgery and Szentagothai Research Center, University of Pecs Medical School , Pecs , Hungary
| | - Farzaneh Sorond
- Department of Neurology, Northwestern University , Chicago, Illinois
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Vascular Cognitive Impairment Program, Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Vascular Cognitive Impairment Program, Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
- Semmelweis University, Department of Pulmonology , Budapest , Hungary
| |
Collapse
|
430
|
Guo D, Shen Y, Li W, Li Q, Zhao Y, Pan C, Chen B, Zhong Y, Miao Y. 6-Bromoindirubin-3'-Oxime (6BIO) Suppresses the mTOR Pathway, Promotes Autophagy, and Exerts Anti-aging Effects in Rodent Liver. Front Pharmacol 2019; 10:320. [PMID: 31057395 PMCID: PMC6477879 DOI: 10.3389/fphar.2019.00320] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/15/2019] [Indexed: 01/09/2023] Open
Abstract
Liver aging is associated with age-related histopathological and functional changes that significantly enhance the risk of numerous diseases or disorders developing in elderly populations. 6-Bromoindirubin-3'-oxime (6BIO), a potent inhibitor of glycogen synthase kinase-3 (GSK-3), has been implicated in various age-related diseases and processes, such as tumorigenesis, neurodegeneration, and diabetes. Recent studies have also revealed that 6BIO increases autophagy in yeast, mammalian cell lines, and dopaminergic neurons, which is one of the classical mechanisms strongly associated with liver aging. However, the impact or the mechanism of action of 6BIO in liver remains entirely unknown. Here, we find that 6BIO reduces oxidative stress, improves lipid metabolism, enhances autophagy, and significantly retards liver aging via modulating the GSK-3β pathway and mTOR pathway. Our findings suggest that 6BIO could be a potential agent to protect the liver in the field of anti-aging pharmacology.
Collapse
Affiliation(s)
- Donghao Guo
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yun Shen
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Li
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qinjie Li
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yu Zhao
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chenhao Pan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bi Chen
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuan Zhong
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ya Miao
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
431
|
Pignolo RJ, Samsonraj RM, Law SF, Wang H, Chandra A. Targeting Cell Senescence for the Treatment of Age-Related Bone Loss. Curr Osteoporos Rep 2019; 17:70-85. [PMID: 30806947 DOI: 10.1007/s11914-019-00504-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We review cell senescence in the context of age-related bone loss by broadly discussing aging mechanisms in bone, currently known inducers and markers of senescence, the senescence-associated secretory phenotype (SASP), and the emerging roles of senescence in bone homeostasis and pathology. RECENT FINDINGS Cellular senescence is a state of irreversible cell cycle arrest induced by insults or stressors including telomere attrition, oxidative stress, DNA damage, oncogene activation, and other intrinsic or extrinsic triggers and there is mounting evidence for the role of senescence in aging bone. Cellular aging also instigates a SASP that exerts detrimental paracrine and likely systemic effects. With aging, multiple cell types in the bone microenvironment become senescent, with osteocytes and myeloid cells as primary contributors to the SASP. Targeting undesired senescent cells may be a favorable strategy to promote bone anabolic and anti-resorptive functions in aging bone, with the possibility of improving bone quality and function with normal aging and/or disease.
Collapse
Affiliation(s)
- Robert J Pignolo
- Department of Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of Geriatric Medicine & Gerontology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA.
| | | | - Susan F Law
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Haitao Wang
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Geriatric Medicine & Gerontology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| | - Abhishek Chandra
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Geriatric Medicine & Gerontology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
432
|
Abstract
Cellular senescence refers to a process induced by various types of stress that causes irreversible cell cycle arrest and distinct cellular alterations, including profound changes in gene expression, metabolism, and chromatin organization as well as activation/reinforcement of anti-apoptotic pathways and development of a pro-inflammatory secretome or senescence-associated secretory phenotype (SASP). However, because of challenges and technical limitations in identifying and characterizing senescent cells in living organisms, only recently have some of the diverse in vivo roles of these unique cells been discovered. New findings indicate that senescent cells and their SASP can have acute beneficial functions, such as in tissue regeneration and wound healing. However, in contrast, when senescent cells accumulate in excess chronically at sites of pathology or in old tissues they drive multiple age-associated chronic diseases. Senotherapeutics that selectively eliminate senescent cells ("senolytics") or inhibit their detrimental SASP ("senomorphics") have been developed and tested in aged preclinical models. These studies have established that targeting senescence is a powerful anti-aging strategy to improve "healthspan" - i.e., the healthy period of life free of chronic disease. The roles of senescence in mediating age-related bone loss have been a recent focus of rigorous investigation. Studies in mice and humans demonstrate that with aging, at least a subset of most cell types in the bone microenvironment become senescent and develop a heterogeneous SASP. Furthermore, age-related bone loss can be alleviated in old mice, with apparent advantages over anti-resorptive therapy, by reducing the senescent cell burden genetically or pharmacologically with the first class of senolytics or a senomorphic. Collectively, these findings point to targeting senescence as a transformational strategy to extend healthspan, therefore providing strong rationale for identifying and optimizing senotherapeutics to alleviate multiple chronic diseases of aging, including osteoporosis, and set the stage for translating senotherapeutics to humans, with clinical trials currently ongoing.
Collapse
Affiliation(s)
- Joshua N Farr
- Division of Endocrinology and Metabolism and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - Sundeep Khosla
- Division of Endocrinology and Metabolism and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
433
|
Sanchis P, Ho CY, Liu Y, Beltran LE, Ahmad S, Jacob AP, Furmanik M, Laycock J, Long DA, Shroff R, Shanahan CM. Arterial "inflammaging" drives vascular calcification in children on dialysis. Kidney Int 2019; 95:958-972. [PMID: 30827513 PMCID: PMC6684370 DOI: 10.1016/j.kint.2018.12.014] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 11/28/2018] [Accepted: 12/06/2018] [Indexed: 01/23/2023]
Abstract
Children on dialysis have a cardiovascular mortality risk equivalent to older adults in the general population, and rapidly develop medial vascular calcification, an age-associated pathology. We hypothesized that premature vascular ageing contributes to calcification in children with advanced chronic kidney disease (CKD). Vessels from children with Stage 5 CKD with and without dialysis had evidence of increased oxidative DNA damage. The senescence markers p16 and p21 were also increased in vessels from children on dialysis. Treatment of vessel rings ex vivo with calcifying media increased oxidative DNA damage in vessels from children with Stage 5 CKD, but not in those from healthy controls. Vascular smooth muscle cells cultured from children on dialysis exhibited persistent DNA damage, impaired DNA damage repair, and accelerated senescence. Under calcifying conditions vascular smooth muscle cells from children on dialysis showed increased osteogenic differentiation and calcification. These changes correlated with activation of the senescence-associated secretory phenotype (SASP), an inflammatory phenotype characterized by the secretion of proinflammatory cytokines and growth factors. Blockade of ataxia-telangiectasia mutated (ATM)-mediated DNA damage signaling reduced both inflammation and calcification. Clinically, children on dialysis had elevated circulating levels of osteogenic SASP factors that correlated with increased vascular stiffness and coronary artery calcification. These data imply that dysregulated mineral metabolism drives vascular "inflammaging" by promoting oxidative DNA damage, premature senescence, and activation of a pro-inflammatory SASP. Drugs that target DNA damage signaling or eliminate senescent cells may have the potential to prevent vascular calcification in patients with advanced CKD.
Collapse
Affiliation(s)
- Pilar Sanchis
- British Heart Foundation Centre of Excellence, Cardiovascular Division, King's College London, London, UK
| | - Chin Yee Ho
- British Heart Foundation Centre of Excellence, Cardiovascular Division, King's College London, London, UK
| | - Yiwen Liu
- British Heart Foundation Centre of Excellence, Cardiovascular Division, King's College London, London, UK
| | - Leilani E Beltran
- British Heart Foundation Centre of Excellence, Cardiovascular Division, King's College London, London, UK
| | - Sadia Ahmad
- British Heart Foundation Centre of Excellence, Cardiovascular Division, King's College London, London, UK
| | - Anne P Jacob
- British Heart Foundation Centre of Excellence, Cardiovascular Division, King's College London, London, UK
| | - Malgorzata Furmanik
- British Heart Foundation Centre of Excellence, Cardiovascular Division, King's College London, London, UK
| | - Joanne Laycock
- British Heart Foundation Centre of Excellence, Cardiovascular Division, King's College London, London, UK
| | - David A Long
- Developmental Biology and Cancer Programme, Great Ormond Street Hospital and University College London Institute of Child Health, London, UK
| | - Rukshana Shroff
- Nephrology Unit, Great Ormond Street Hospital and University College London Institute of Child Health, London, UK
| | - Catherine M Shanahan
- British Heart Foundation Centre of Excellence, Cardiovascular Division, King's College London, London, UK.
| |
Collapse
|
434
|
Kritchevsky SB, Forman DE, Callahan KE, Ely EW, High KP, McFarland F, Pérez-Stable EJ, Schmader KE, Studenski SA, Williams J, Zieman S, Guralnik JM. Pathways, Contributors, and Correlates of Functional Limitation Across Specialties: Workshop Summary. J Gerontol A Biol Sci Med Sci 2019; 74:534-543. [PMID: 29697758 PMCID: PMC6417483 DOI: 10.1093/gerona/gly093] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Indexed: 12/25/2022] Open
Abstract
Traditional clinical care models focus on the measurement and normalization of individual organ systems and de-emphasize aspects of health related to the integration of physiologic systems. Measures of physical, cognitive and sensory, and psychosocial or emotional function predict important health outcomes like death and disability independently from the severity of a specific disease, cumulative co-morbidity, or disease severity measures. A growing number of clinical scientists in several subspecialties are exploring the utility of functional assessment to predict complication risk, indicate stress resistance, inform disease screening approaches and risk factor interpretation, and evaluate care. Because a substantial number of older adults in the community have some form of functional limitation, integrating functional assessment into clinical medicine could have a large impact. Although interest in functional implications for health and disease management is growing, the science underlying functional capacity, functional limitation, physical frailty, and functional metrics is often siloed among different clinicians and researchers, with fragmented concepts and methods. On August 25-26, 2016, participants at a trans-disciplinary workshop, supported by the National Institute on Aging and the John A. Hartford Foundation, explored what is known about the pathways, contributors, and correlates of physical, cognitive, and sensory functional measures across conditions and disease states; considered social determinants and health disparities; identified knowledge gaps, and suggested priorities for future research. This article summarizes those discussions.
Collapse
Affiliation(s)
- Stephen B Kritchevsky
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Daniel E Forman
- Department of Medicine, University of Pittsburgh, Pennsylvania
| | - Kathryn E Callahan
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - E Wesley Ely
- VA Tennessee Valley Geriatric Research Education Clinical Center (GRECC) and Department of Medicine, Vanderbilt University, Nashville
| | - Kevin P High
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Frances McFarland
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | | | | | | | - Jack M Guralnik
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
435
|
Short S, Fielder E, Miwa S, von Zglinicki T. Senolytics and senostatics as adjuvant tumour therapy. EBioMedicine 2019; 41:683-692. [PMID: 30737084 PMCID: PMC6441870 DOI: 10.1016/j.ebiom.2019.01.056] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/16/2019] [Accepted: 01/30/2019] [Indexed: 12/25/2022] Open
Abstract
Cell senescence is a driver of ageing, frailty, age-associated disease and functional decline. In oncology, tumour cell senescence may contribute to the effect of adjuvant therapies, as it blocks tumour growth. However, this is frequently incomplete, and tumour cells that recover from senescence may gain a more stem-like state with increased proliferative potential. This might be exaggerated by the induction of senescence in the surrounding niche cells. Finally, senescence will spread through bystander effects, possibly overwhelming the capacity of the immune system to ablate senescent cells. This induces a persistent system-wide senescent cell accumulation, which we hypothesize is the cause for the premature frailty, multi-morbidity and increased mortality in cancer survivors. Senolytics, drugs that selectively kill senescent cells, have been developed recently and have been proposed as second-line adjuvant tumour therapy. Similarly, by blocking accelerated senescence following therapy, senolytics might prevent and potentially even revert premature frailty in cancer survivors. Adjuvant senostatic interventions, which suppress senescence-associated bystander signalling, might also have therapeutic potential. This becomes pertinent because treatments that are senostatic in vitro (e.g. dietary restriction mimetics) persistently reduce numbers of senescent cells in vivo, i.e. act as net senolytics in immunocompetent hosts.
Collapse
Affiliation(s)
- Susan Short
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Beckett St, Leeds LS9 7TF, UK
| | - Edward Fielder
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biology, 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 Biology, 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 Biology, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| |
Collapse
|
436
|
Wakasugi T, Shimizu I, Yoshida Y, Hayashi Y, Ikegami R, Suda M, Katsuumi G, Nakao M, Hoyano M, Kashimura T, Nakamura K, Ito H, Nojiri T, Soga T, Minamino T. Role of smooth muscle cell p53 in pulmonary arterial hypertension. PLoS One 2019; 14:e0212889. [PMID: 30807606 PMCID: PMC6391010 DOI: 10.1371/journal.pone.0212889] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/11/2019] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by remodeling and narrowing of the pulmonary arteries, which lead to elevation of right ventricular pressure, heart failure, and death. Proliferation of pulmonary artery smooth muscle cells (PASMCs) is thought to be central to the pathogenesis of PAH, although the underlying mechanisms are still being explored. The protein p53 is involved in cell cycle coordination, DNA repair, apoptosis, and cellular senescence, but its role in pulmonary hypertension (PH) is not fully known. We developed a mouse model of hypoxia-induced pulmonary hypertension (PH) and found significant reduction of p53 expression in the lungs. Our in vitro experiments with metabolomic analyses and the Seahorse XF extracellular flux analyzer indicated that suppression of p53 expression in PASMCs led to upregulation of glycolysis and downregulation of mitochondrial respiration, suggesting a proliferative phenotype resembling that of cancer cells. It was previously shown that systemic genetic depletion of p53 in a murine PH model led to more severe lung manifestations. Lack of information about the role of cell-specific p53 signaling promoted us to investigate it in our mouse PH model with the inducible Cre-loxP system. We generated a mouse model with SMC-specific gain or loss of p53 function by crossing Myh11-Cre/ERT2 mice with floxed Mdm4 mice or floxed Trp53 mice. After these animals were exposed to hypoxia for 4 weeks, we conducted hemodynamic and echocardiographic studies. Surprisingly, the severity of PH was similar in both groups of mice and there were no differences between the genotypes. Our findings in these mice indicate that activation or suppression of p53 signaling in SMCs has a minor role in the pathogenesis of PH and suggest that p53 signaling in other cells (endothelial cells, immune cells, or fibroblasts) may be involved in the progression of this condition.
Collapse
Affiliation(s)
- Takayuki Wakasugi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuka Hayashi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryutaro Ikegami
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masaaki Nakao
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Makoto Hoyano
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takeshi Kashimura
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takashi Nojiri
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita-City, Osaka, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- * E-mail: ,
| |
Collapse
|
437
|
Melo Pereira S, Ribeiro R, Logarinho E. Approaches towards Longevity: Reprogramming, Senolysis, and Improved Mitotic Competence as Anti-Aging Therapies. Int J Mol Sci 2019; 20:E938. [PMID: 30795536 PMCID: PMC6413205 DOI: 10.3390/ijms20040938] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/09/2019] [Accepted: 02/18/2019] [Indexed: 02/06/2023] Open
Abstract
Mainstream approaches that are currently used as anti-aging therapies primarily explore the senescence and epigenetic drift aging hallmarks and they are at two ends of the spectrum. While senolytic therapies include either the selective elimination of senescent cells or the disruption of their secretome with the use of drugs or natural compounds, cellular reprogramming uses genetic manipulation to revert cells all the way back to pluripotency. Here, we describe the progress that has been made on these therapies, while highlighting the major challenges involved. Moreover, based on recent findings elucidating the impact of mitotic shutdown and aneuploidy in cellular senescence, we discuss the modulation of mitotic competence as an alternative strategy to delay the hallmarks of aging. We propose that a regulated rise in mitotic competence of cells could circumvent certain limitations that are present in the senolytic and reprogramming approaches, by acting to decelerate senescence and possibly restore the epigenetic landscape.
Collapse
Affiliation(s)
- Sofia Melo Pereira
- Ageing and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Rui Ribeiro
- Ageing and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Elsa Logarinho
- Ageing and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Cell Division Unit, Faculty of Medicine, Department of Experimental Biology, Universidade do Porto, 4200-319 Porto, Portugal.
| |
Collapse
|
438
|
Yokoyama M, Shimizu I, Nagasawa A, Yoshida Y, Katsuumi G, Wakasugi T, Hayashi Y, Ikegami R, Suda M, Ota Y, Okada S, Fruttiger M, Kobayashi Y, Tsuchida M, Kubota Y, Minamino T. p53 plays a crucial role in endothelial dysfunction associated with hyperglycemia and ischemia. J Mol Cell Cardiol 2019; 129:105-117. [PMID: 30790589 DOI: 10.1016/j.yjmcc.2019.02.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 02/12/2019] [Accepted: 02/16/2019] [Indexed: 12/23/2022]
Abstract
p53 is a guardian of the genome that protects against carcinogenesis. There is accumulating evidence that p53 is activated with aging. Such activation has been reported to contribute to various age-associated pathologies, but its role in vascular dysfunction is largely unknown. The aim of this study was to investigate whether activation of endothelial p53 has a pathological effect in relation to endothelial function. We established endothelial p53 loss-of-function and gain-of-function models by breeding endothelial-cell specific Cre mice with floxed Trp53 or floxed Mdm2/Mdm4 mice, respectively. Then we induced diabetes by injection of streptozotocin. In the diabetic state, endothelial p53 expression was markedly up-regulated and endothelium-dependent vasodilatation was significantly impaired. Impairment of vasodilatation was significantly ameliorated in endothelial p53 knockout (EC-p53 KO) mice, and deletion of endothelial p53 also significantly enhanced the induction of angiogenesis by ischemia. Conversely, activation of endothelial p53 by deleting Mdm2/Mdm4 reduced both endothelium-dependent vasodilatation and ischemia-induced angiogenesis. Introduction of p53 into human endothelial cells up-regulated the expression of phosphatase and tensin homolog (PTEN), thereby reducing phospho-eNOS levels. Consistent with these results, the beneficial impact of endothelial p53 deletion on endothelial function was attenuated in EC-p53 KO mice with an eNOS-deficient background. These results show that endothelial p53 negatively regulates endothelium-dependent vasodilatation and ischemia-induced angiogenesis, suggesting that inhibition of endothelial p53 could be a novel therapeutic target in patients with metabolic disorders.
Collapse
Affiliation(s)
- Masataka Yokoyama
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Ayako Nagasawa
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Department of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Takayuki Wakasugi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yuka Hayashi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Ryutaro Ikegami
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yusuke Ota
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Sho Okada
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Masanori Tsuchida
- Department of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
439
|
Anderson R, Lagnado A, Maggiorani D, Walaszczyk A, Dookun E, Chapman J, Birch J, Salmonowicz H, Ogrodnik M, Jurk D, Proctor C, Correia-Melo C, Victorelli S, Fielder E, Berlinguer-Palmini R, Owens A, Greaves LC, Kolsky KL, Parini A, Douin-Echinard V, LeBrasseur NK, Arthur HM, Tual-Chalot S, Schafer MJ, Roos CM, Miller JD, Robertson N, Mann J, Adams PD, Tchkonia T, Kirkland JL, Mialet-Perez J, Richardson GD, Passos JF. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J 2019; 38:embj.2018100492. [PMID: 30737259 PMCID: PMC6396144 DOI: 10.15252/embj.2018100492] [Citation(s) in RCA: 337] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/18/2018] [Accepted: 01/02/2019] [Indexed: 01/08/2023] Open
Abstract
Ageing is the biggest risk factor for cardiovascular disease. Cellular senescence, a process driven in part by telomere shortening, has been implicated in age‐related tissue dysfunction. Here, we address the question of how senescence is induced in rarely dividing/post‐mitotic cardiomyocytes and investigate whether clearance of senescent cells attenuates age‐related cardiac dysfunction. During ageing, human and murine cardiomyocytes acquire a senescent‐like phenotype characterised by persistent DNA damage at telomere regions that can be driven by mitochondrial dysfunction and crucially can occur independently of cell division and telomere length. Length‐independent telomere damage in cardiomyocytes activates the classical senescence‐inducing pathways, p21CIP and p16INK4a, and results in a non‐canonical senescence‐associated secretory phenotype, which is pro‐fibrotic and pro‐hypertrophic. Pharmacological or genetic clearance of senescent cells in mice alleviates detrimental features of cardiac ageing, including myocardial hypertrophy and fibrosis. Our data describe a mechanism by which senescence can occur and contribute to age‐related myocardial dysfunction and in the wider setting to ageing in post‐mitotic tissues.
Collapse
Affiliation(s)
- Rhys Anderson
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Anthony Lagnado
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Damien Maggiorani
- INSERM Institute of metabolic and cardiovascular diseases, University of Toulouse, Toulouse, France
| | - Anna Walaszczyk
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Emily Dookun
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.,Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - James Chapman
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jodie Birch
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Hanna Salmonowicz
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mikolaj Ogrodnik
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Diana Jurk
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Carole Proctor
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Clara Correia-Melo
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Stella Victorelli
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Edward Fielder
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | | | - Andrew Owens
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Laura C Greaves
- Wellcome Trust Centre for Mitochondrial Research, Centre for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Kathy L Kolsky
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Angelo Parini
- INSERM Institute of metabolic and cardiovascular diseases, University of Toulouse, Toulouse, France
| | - Victorine Douin-Echinard
- INSERM Institute of metabolic and cardiovascular diseases, University of Toulouse, Toulouse, France
| | | | - Helen M Arthur
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Simon Tual-Chalot
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Marissa J Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Carolyn M Roos
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Jordan D Miller
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Neil Robertson
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Jelena Mann
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Peter D Adams
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK.,Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Jeanne Mialet-Perez
- INSERM Institute of metabolic and cardiovascular diseases, University of Toulouse, Toulouse, France
| | - Gavin D Richardson
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - João F Passos
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK .,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
440
|
Hoffman CM, Han J, Calvi LM. Impact of aging on bone, marrow and their interactions. Bone 2019; 119:1-7. [PMID: 30010082 DOI: 10.1016/j.bone.2018.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/12/2018] [Accepted: 07/12/2018] [Indexed: 12/24/2022]
Abstract
Hematopoiesis in land dwelling vertebrates and marine mammals occurs within the bone marrow, continually providing mature progeny over the course of an organism's lifetime. This conserved dependency highlights the critical relationship between these two organs, yet the skeletal and hematopoietic systems are often thought of as separate. In fact, data are beginning to show that skeletal disease pathogenesis influences hematopoiesis and viceversa, offering novel opportunities to approach disease affecting bone and blood. With a growing global population of aged individuals, interest has focused on cell autonomous changes in hematopoietic and skeletal systems that result in dysfunction. The purpose of this review is to summarize the literature on aging effects in both fields, and provide critical examples of organ cross-talk in the aging process.
Collapse
Affiliation(s)
- Corey M Hoffman
- University of Rochester Medical Center, Rochester, NY, United States of America
| | - Jimin Han
- University of Rochester Medical Center, Rochester, NY, United States of America
| | - Laura M Calvi
- University of Rochester Medical Center, Rochester, NY, United States of America.
| |
Collapse
|
441
|
Aavik E, Babu M, Ylä-Herttuala S. DNA methylation processes in atherosclerotic plaque. Atherosclerosis 2019; 281:168-179. [DOI: 10.1016/j.atherosclerosis.2018.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/09/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022]
|
442
|
Ogrodnik M, Salmonowicz H, Gladyshev VN. Integrating cellular senescence with the concept of damage accumulation in aging: Relevance for clearance of senescent cells. Aging Cell 2019; 18:e12841. [PMID: 30346102 PMCID: PMC6351832 DOI: 10.1111/acel.12841] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/31/2018] [Accepted: 08/20/2018] [Indexed: 12/12/2022] Open
Abstract
Understanding the aging process and ways to manipulate it is of major importance for biology and medicine. Among the many aging theories advanced over the years, the concept most consistent with experimental evidence posits the buildup of numerous forms of molecular damage as a foundation of the aging process. Here, we discuss that this concept integrates well with recent findings on cellular senescence, offering a novel view on the role of senescence in aging and age‐related disease. Cellular senescence has a well‐established role in cellular aging, but its impact on the rate of organismal aging is less defined. One of the most prominent features of cellular senescence is its association with macromolecular damage. The relationship between cell senescence and damage concerns both damage as a molecular signal of senescence induction and accelerated accumulation of damage in senescent cells. We describe the origin, regulatory mechanisms, and relevance of various damage forms in senescent cells. This view on senescent cells as carriers and inducers of damage puts new light on senescence, considering it as a significant contributor to the rise in organismal damage. Applying these ideas, we critically examine current evidence for a role of cellular senescence in aging and age‐related diseases. We also discuss the differential impact of longevity interventions on senescence burden and other types of age‐related damage. Finally, we propose a model on the role of aging‐related damage accumulation and the rate of aging observed upon senescent cell clearance.
Collapse
Affiliation(s)
- Mikolaj Ogrodnik
- Institute for Cell and Molecular Biosciences; Newcastle University Institute for Ageing; Newcastle upon Tyne UK
| | - Hanna Salmonowicz
- Institute for Cell and Molecular Biosciences; Newcastle University Institute for Ageing; Newcastle upon Tyne UK
| | - Vadim N. Gladyshev
- Division of Genetics; Department of Medicine; Brigham and Women's Hospital and Harvard Medical School; Boston Massachusetts
| |
Collapse
|
443
|
Hobson S, Arefin S, Kublickiene K, Shiels PG, Stenvinkel P. Senescent Cells in Early Vascular Ageing and Bone Disease of Chronic Kidney Disease-A Novel Target for Treatment. Toxins (Basel) 2019; 11:toxins11020082. [PMID: 30717151 PMCID: PMC6409791 DOI: 10.3390/toxins11020082] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/13/2022] Open
Abstract
Together with bone-mineral disorders, premature vascular ageing is a common feature of the uremic phenotype. A detailed understanding of mechanisms involved remains unclear and warrants further research. Available treatment options for end stage renal disease are principally dialysis and organ transplantation, as other treatment alternatives have proven insufficient. Chronic kidney disease (CKD) has been proposed as a model of early vascular and bone ageing, with accumulating evidence supporting the contribution of cellular senescence and the senescence-associated secretory phenotype (SASP) to cardiovascular pathology in CKD. Correspondingly, novel therapies based around the use of senolytic compounds and nuclear factor-erythroid-2-related factor 2 (Nrf2) agonists, have been suggested as attractive novel treatment options. In this review, we detail the contribution of the uremic environment to these processes underpinning ageing and how these relate to vascular health.
Collapse
Affiliation(s)
- Sam Hobson
- Division of Renal Medicine, Department of Clinical Science, Technology and Intervention, Karolinska University Hospital, 14186 Stockholm, Sweden.
| | - Samsul Arefin
- Division of Renal Medicine, Department of Clinical Science, Technology and Intervention, Karolinska University Hospital, 14186 Stockholm, Sweden.
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Technology and Intervention, Karolinska University Hospital, 14186 Stockholm, Sweden.
| | - Paul G Shiels
- Institute of Cancer Sciences, MVLS, University of Glasgow, Glasgow G61 1QH, UK.
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Technology and Intervention, Karolinska University Hospital, 14186 Stockholm, Sweden.
| |
Collapse
|
444
|
Noly PE, Labbé P, Thorin-Trescases N, Fortier A, Nguyen A, Thorin E, Carrier M. Reduction of plasma angiopoietin-like 2 after cardiac surgery is related to tissue inflammation and senescence status of patients. J Thorac Cardiovasc Surg 2019; 158:792-802.e5. [PMID: 30745045 DOI: 10.1016/j.jtcvs.2018.12.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/04/2018] [Accepted: 12/15/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVES A strong relationship between high circulating angiopoietin-like 2 (ANGPTL2) levels, a proinflammatory adipokine, and cardiovascular diseases has been reported. Our objective was to determine whether plasma ANGPTL2 and high-sensitivity C-reactive protein (hs-CRP) levels change postoperatively in patients who underwent heart valve surgery and/or coronary artery bypass grafting. We hypothesized that a corrective cardiac surgery would decrease ANGPTL2 levels. METHODS In 47 prospectively recruited patients who underwent coronary artery bypass grafting (n = 16), valve replacement (n = 16), or both (n = 15), we measured plasma ANGPTL2 and hs-CRP levels preoperatively, at 24 hours, at 3 to 5 days (hospital discharge), and at 30 to 90 days (follow-up) after surgery. Mediastinal adipose tissue and distal fragments of the left internal mammary artery (IMA) were harvested during surgery and mRNA expression of inflammatory and senescence markers was assessed using real-time quantitative polymerase chain reaction. RESULTS ANGPTL2 and hs-CRP levels were elevated 24 hours after surgery and then returned to baseline levels. We noted, however, a dichotomy among patients: compared with baseline, plasma ANGPTL2 levels either significantly decreased (n = 21/47) or increased (n = 26/47) after surgery. In contrast, hs-CRP levels were identical between groups (P = .997). Patients in the increased group were older (P = .002) with a higher systolic blood pressure (P = .038) at baseline. Moreover, changes in ANGPTL2 levels (ΔANGPTL2 = final minus initial levels) positively correlated with mRNA expression of tumor necrosis factor α and interleukin 8 in mediastinal adipose tissue and IMA (P < .05) and with the senescence-associated marker cyclin-dependent kinase inhibitor 1 in IMA (P = .009). CONCLUSIONS In younger patients with lower levels of tissue inflammation and arterial senescence load, ANGPTL2, but not hs-CRP levels decreased after cardiac surgery, suggesting that circulating ANGPTL2 reflects tissue inflammation and senescence.
Collapse
Affiliation(s)
- Pierre-Emmanuel Noly
- Faculty of Medicine, Department of Surgery, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Pauline Labbé
- Montreal Heart Institute Research Center, Université de Montréal, Montreal, Quebec, Canada; Department of Pharmacology, Université de Montréal, Montreal, Quebec, Canada
| | | | - Annik Fortier
- Montreal Health Innovations Coordinating Center, Montreal, Quebec, Canada
| | - Albert Nguyen
- Montreal Heart Institute Research Center, Université de Montréal, Montreal, Quebec, Canada
| | - Eric Thorin
- Faculty of Medicine, Department of Surgery, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada.
| | - Michel Carrier
- Faculty of Medicine, Department of Surgery, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
445
|
Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, Prata L, Masternak MM, Kritchevsky SB, Musi N, Kirkland JL. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019; 40:554-563. [PMID: 30616998 PMCID: PMC6412088 DOI: 10.1016/j.ebiom.2018.12.052] [Citation(s) in RCA: 796] [Impact Index Per Article: 132.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/14/2018] [Accepted: 12/21/2018] [Indexed: 01/19/2023] Open
Abstract
Background Cellular senescence is a key mechanism that drives age-related diseases, but has yet to be targeted therapeutically in humans. Idiopathic pulmonary fibrosis (IPF) is a progressive, fatal cellular senescence-associated disease. Selectively ablating senescent cells using dasatinib plus quercetin (DQ) alleviates IPF-related dysfunction in bleomycin-administered mice. Methods A two-center, open-label study of intermittent DQ (D:100 mg/day, Q:1250 mg/day, three-days/week over three-weeks) was conducted in participants with IPF (n = 14) to evaluate feasibility of implementing a senolytic intervention. The primary endpoints were retention rates and completion rates for planned clinical assessments. Secondary endpoints were safety and change in functional and reported health measures. Associations with the senescence-associated secretory phenotype (SASP) were explored. Findings Fourteen patients with stable IPF were recruited. The retention rate was 100% with no DQ discontinuation; planned clinical assessments were complete in 13/14 participants. One serious adverse event was reported. Non-serious events were primarily mild-moderate, with respiratory symptoms (n = 16 total events), skin irritation/bruising (n = 14), and gastrointestinal discomfort (n = 12) being most frequent. Physical function evaluated as 6-min walk distance, 4-m gait speed, and chair-stands time was significantly and clinically-meaningfully improved (p < .05). Pulmonary function, clinical chemistries, frailty index (FI-LAB), and reported health were unchanged. DQ effects on circulat.ing SASP factors were inconclusive, but correlations were observed between change in function and change in SASP-related matrix-remodeling proteins, microRNAs, and pro-inflammatory cytokines (23/48 markers r ≥ 0.50). Interpretation Our first-in-humans open-label pilot supports study feasibility and provides initial evidence that senolytics may alleviate physical dysfunction in IPF, warranting evaluation of DQ in larger randomized controlled trials for senescence-related diseases. ClinicalTrials.gov identifier: NCT02874989 (posted 2016–2018).
Collapse
Affiliation(s)
- Jamie N Justice
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine (WFSM), 1 Medical Center Blvd, Winston-Salem, NC 27157, United States.
| | - Anoop M Nambiar
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Internal Medicine, University of Texas Health Sciences Center at San Antonio (UTHSCSA) and South Texas Veterans Health Care System, San Antonio, TX 78229, United States.
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| | - Rodolfo Pascual
- Internal Medicine - Pulmonary, Critical Care, Allergy, Immunologic Medicine, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC 27157, United States.
| | - Shahrukh K Hashmi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| | - Larissa Prata
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, United States.
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine (WFSM), 1 Medical Center Blvd, Winston-Salem, NC 27157, United States.
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, Center for Healthy Aging, University of Texas Health Sciences Center at San Antonio and South Texas Veterans Health Care System, San Antonio, TX 78229, United States; San Antonio Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX 78229, United States.
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| |
Collapse
|
446
|
Lau A, Kennedy BK, Kirkland JL, Tullius SG. Mixing old and young: enhancing rejuvenation and accelerating aging. J Clin Invest 2019; 129:4-11. [PMID: 30601138 DOI: 10.1172/jci123946] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Donor age and recipient age are factors that influence transplantation outcomes. Aside from age-associated differences in intrinsic graft function and alloimmune responses, the ability of young and old cells to exert either rejuvenating or aging effects extrinsically may also apply to the transplantation of hematopoietic stem cells or solid organ transplants. While the potential for rejuvenation mediated by the transfer of youthful cells is currently being explored for therapeutic applications, aspects that relate to accelerating aging are no less clinically significant. Those effects may be particularly relevant in transplantation with an age discrepancy between donor and recipient. Here, we review recent advances in understanding the mechanisms by which young and old cells modify their environments to promote rejuvenation- or aging-associated phenotypes. We discuss their relevance to clinical transplantation and highlight potential opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Ashley Lau
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian K Kennedy
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Institute for Clinical Sciences, Singapore.,Agency for Science, Technology and Research (A*STAR), Singapore.,Buck Institute for Research on Aging, Novato, California, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
447
|
Mahmoudi S, Xu L, Brunet A. Turning back time with emerging rejuvenation strategies. Nat Cell Biol 2019; 21:32-43. [PMID: 30602763 DOI: 10.1038/s41556-018-0206-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/24/2018] [Indexed: 01/10/2023]
Abstract
Ageing is associated with the functional decline of all tissues and a striking increase in many diseases. Although ageing has long been considered a one-way street, strategies to delay and potentially even reverse the ageing process have recently been developed. Here, we review four emerging rejuvenation strategies-systemic factors, metabolic manipulations, senescent cell ablation and cellular reprogramming-and discuss their mechanisms of action, cellular targets, potential trade-offs and application to human ageing.
Collapse
Affiliation(s)
- Salah Mahmoudi
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lucy Xu
- Department of Genetics, Stanford University, Stanford, CA, USA.,Department of Biology, Stanford University, Stanford, CA, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA. .,Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA, USA.
| |
Collapse
|
448
|
Larsson L, Degens H, Li M, Salviati L, Lee YI, Thompson W, Kirkland JL, Sandri M. Sarcopenia: Aging-Related Loss of Muscle Mass and Function. Physiol Rev 2019; 99:427-511. [PMID: 30427277 PMCID: PMC6442923 DOI: 10.1152/physrev.00061.2017] [Citation(s) in RCA: 965] [Impact Index Per Article: 160.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 05/14/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia is a loss of muscle mass and function in the elderly that reduces mobility, diminishes quality of life, and can lead to fall-related injuries, which require costly hospitalization and extended rehabilitation. This review focuses on the aging-related structural changes and mechanisms at cellular and subcellular levels underlying changes in the individual motor unit: specifically, the perikaryon of the α-motoneuron, its neuromuscular junction(s), and the muscle fibers that it innervates. Loss of muscle mass with aging, which is largely due to the progressive loss of motoneurons, is associated with reduced muscle fiber number and size. Muscle function progressively declines because motoneuron loss is not adequately compensated by reinnervation of muscle fibers by the remaining motoneurons. At the intracellular level, key factors are qualitative changes in posttranslational modifications of muscle proteins and the loss of coordinated control between contractile, mitochondrial, and sarcoplasmic reticulum protein expression. Quantitative and qualitative changes in skeletal muscle during the process of aging also have been implicated in the pathogenesis of acquired and hereditary neuromuscular disorders. In experimental models, specific intervention strategies have shown encouraging results on limiting deterioration of motor unit structure and function under conditions of impaired innervation. Translated to the clinic, if these or similar interventions, by saving muscle and improving mobility, could help alleviate sarcopenia in the elderly, there would be both great humanitarian benefits and large cost savings for health care systems.
Collapse
Affiliation(s)
- Lars Larsson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Hans Degens
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Meishan Li
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Leonardo Salviati
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Young Il Lee
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Wesley Thompson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - James L Kirkland
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Marco Sandri
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| |
Collapse
|
449
|
de Almeida Alvarenga L, Borges NA, Moreira LDSG, Resende Teixeira KT, Carraro-Eduardo JC, Dai L, Stenvinkel P, Lindholm B, Mafra D. Cranberries – potential benefits in patients with chronic kidney disease. Food Funct 2019; 10:3103-3112. [DOI: 10.1039/c9fo00375d] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Patients with chronic kidney disease (CKD) present many complications that potentially could be linked to increased cardiovascular mortality such as inflammation, oxidative stress, cellular senescence and gut dysbiosis.
Collapse
Affiliation(s)
| | - Natália Alvarenga Borges
- Graduate Program in Nutrition Sciences
- Fluminense Federal University (UFF)
- Niterói
- Brazil
- Graduate Program in Cardiovascular Sciences
| | | | | | | | - Lu Dai
- Division of Renal Medicine and Baxter Novum
- Department of Clinical Science
- Technology and Intervention
- Karolinska Institutet
- Stockholm
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum
- Department of Clinical Science
- Technology and Intervention
- Karolinska Institutet
- Stockholm
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum
- Department of Clinical Science
- Technology and Intervention
- Karolinska Institutet
- Stockholm
| | - Denise Mafra
- Graduate Program in Medical Sciences
- Fluminense Federal University (UFF)
- Niterói
- Brazil
- Graduate Program in Nutrition Sciences
| |
Collapse
|
450
|
Swenson BL, Meyer CF, Bussian TJ, Baker DJ. Senescence in aging and disorders of the central nervous system. TRANSLATIONAL MEDICINE OF AGING 2019. [DOI: 10.1016/j.tma.2019.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|