1501
|
Maciel-Barón LÁ, Moreno-Blas D, Morales-Rosales SL, González-Puertos VY, López-Díazguerrero NE, Torres C, Castro-Obregón S, Königsberg M. Cellular Senescence, Neurological Function, and Redox State. Antioxid Redox Signal 2018; 28:1704-1723. [PMID: 28467755 DOI: 10.1089/ars.2017.7112] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Cellular senescence, characterized by permanent cell cycle arrest, has been extensively studied in mitotic cells such as fibroblasts. However, senescent cells have also been observed in the brain. Even though it is recognized that cellular energetic metabolism and redox homeostasis are perturbed in the aged brain and neurodegenerative diseases (NDDs), it is still unknown which alterations in the overall physiology can stimulate cellular senescence induction and their relationship with the former events. Recent Advances: Recent findings have shown that during prolonged inflammatory and pathologic events, the blood-brain barrier could be compromised and immune cells might enter the brain; this fact along with the brain's high oxygen dependence might result in oxidative damage to macromolecules and therefore senescence induction. Thus, cellular senescence in different brain cell types is revised here. CRITICAL ISSUES Most information related to cellular senescence in the brain has been obtained from research in glial cells since it has been assumed that the senescent phenotype is a feature exclusive to mitotic cells. Nevertheless, neurons with senescence hallmarks have been observed in old mouse brains. Therefore, although this is a controversial topic in the field, here we summarize and integrate the observations from several studies and propose that neurons indeed senesce. FUTURE DIRECTIONS It is still unknown which alterations in the overall metabolism can stimulate senescence induction in the aged brain, what are the mechanisms and signaling pathways, and what is their relationship to NDD development. The understanding of these processes will expose new targets to intervene age-associated pathologies.-Antioxid. Redox Signal. 28, 1704-1723.
Collapse
Affiliation(s)
- Luis Ángel Maciel-Barón
- 1 División de Ciencias Biológicas y de la Salud, Department Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa , Iztapalapa, México
| | - Daniel Moreno-Blas
- 2 Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Ciudad de México, México
| | - Sandra Lizbeth Morales-Rosales
- 1 División de Ciencias Biológicas y de la Salud, Department Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa , Iztapalapa, México
| | - Viridiana Yazmín González-Puertos
- 1 División de Ciencias Biológicas y de la Salud, Department Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa , Iztapalapa, México
| | - Norma Edith López-Díazguerrero
- 1 División de Ciencias Biológicas y de la Salud, Department Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa , Iztapalapa, México
| | - Claudio Torres
- 3 Department of Pathology and Laboratory Medicine, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Susana Castro-Obregón
- 2 Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Ciudad de México, México
| | - Mina Königsberg
- 1 División de Ciencias Biológicas y de la Salud, Department Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa , Iztapalapa, México
| |
Collapse
|
1502
|
Kim SJ, Mehta HH, Wan J, Kuehnemann C, Chen J, Hu JF, Hoffman AR, Cohen P. Mitochondrial peptides modulate mitochondrial function during cellular senescence. Aging (Albany NY) 2018; 10:1239-1256. [PMID: 29886458 PMCID: PMC6046248 DOI: 10.18632/aging.101463] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/30/2018] [Indexed: 12/31/2022]
Abstract
Cellular senescence is a complex cell fate response that is thought to underlie several age-related pathologies. Despite a loss of proliferative potential, senescent cells are metabolically active and produce energy-consuming effectors, including senescence-associated secretory phenotypes (SASPs). Mitochondria play crucial roles in energy production and cellular signaling, but the key features of mitochondrial physiology and particularly of mitochondria-derived peptides (MDPs), remain underexplored in senescence responses. Here, we used primary human fibroblasts made senescent by replicative exhaustion, doxorubicin or hydrogen peroxide treatment, and examined the number of mitochondria and the levels of mitochondrial respiration, mitochondrial DNA methylation and the mitochondria-encoded peptides humanin, MOTS-c, SHLP2 and SHLP6. Senescent cells showed increased numbers of mitochondria and higher levels of mitochondrial respiration, variable changes in mitochondrial DNA methylation, and elevated levels of humanin and MOTS-c. Humanin and MOTS-c administration modestly increased mitochondrial respiration and selected components of the SASP in doxorubicin-induced senescent cells partially via JAK pathway. Targeting metabolism in senescence cells is an important strategy to reduce SASP production for eliminating the deleterious effects of senescence. These results provide insight into the role of MDPs in mitochondrial energetics and the production of SASP components by senescent cells.
Collapse
Affiliation(s)
- Su-Jeong Kim
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Hemal H. Mehta
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Junxiang Wan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | | | - Jingcheng Chen
- Stanford University Medical School, Palo Alto Veterans Institute for Research, Palo Alto, CA 94304, USA
| | - Ji-Fan Hu
- Stanford University Medical School, Palo Alto Veterans Institute for Research, Palo Alto, CA 94304, USA
| | - Andrew R. Hoffman
- Stanford University Medical School, Palo Alto Veterans Institute for Research, Palo Alto, CA 94304, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
1503
|
Ness KK, Kirkland JL, Gramatges MM, Wang Z, Kundu M, McCastlain K, Li-Harms X, Zhang J, Tchkonia T, Pluijm SMF, Armstrong GT. Premature Physiologic Aging as a Paradigm for Understanding Increased Risk of Adverse Health Across the Lifespan of Survivors of Childhood Cancer. J Clin Oncol 2018; 36:2206-2215. [PMID: 29874132 DOI: 10.1200/jco.2017.76.7467] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The improvement in survival of childhood cancer observed across the past 50 years has resulted in a growing acknowledgment that simply extending the lifespan of survivors is not enough. It is incumbent on both the cancer research and the clinical care communities to also improve the health span of survivors. It is well established that aging adult survivors of childhood cancer are at increased risk of chronic health conditions, relative to the general population. However, as the first generation of survivors age into their 50s and 60s, it has become increasingly evident that this population is also at risk of early onset of physiologic aging. Geriatric measures have uncovered evidence of reduced strength and speed and increased fatigue, all components of frailty, among survivors with a median age of 33 years, which is similar to adults older than 65 years of age in the general population. Furthermore, frailty in survivors independently increased the risk of morbidity and mortality. Although there has been a paucity of research investigating the underlying biologic mechanisms for advanced physiologic age in survivors, results from geriatric populations suggest five biologically plausible mechanisms that may be potentiated by exposure to cancer therapies: increased cellular senescence, reduced telomere length, epigenetic modifications, somatic mutations, and mitochondrial DNA infidelity. There is now a critical need for research to elucidate the biologic mechanisms of premature aging in survivors of childhood cancer. This research could pave the way for new frontiers in the prevention of these life-changing outcomes.
Collapse
Affiliation(s)
- Kirsten K Ness
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - James L Kirkland
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Maria Monica Gramatges
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Zhaoming Wang
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Mondira Kundu
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Kelly McCastlain
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Xiujie Li-Harms
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jinghui Zhang
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Tamar Tchkonia
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Saskia Martine Francesca Pluijm
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Gregory T Armstrong
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
1504
|
Baar MP, Perdiguero E, Muñoz-Cánoves P, de Keizer PLJ. Musculoskeletal senescence: a moving target ready to be eliminated. Curr Opin Pharmacol 2018; 40:147-155. [DOI: 10.1016/j.coph.2018.05.007] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/16/2018] [Indexed: 11/17/2022]
|
1505
|
Toll-like receptor 4 (TLR4) deficient mice are protected from adipose tissue inflammation in aging. Aging (Albany NY) 2018; 9:1971-1982. [PMID: 28898202 PMCID: PMC5636669 DOI: 10.18632/aging.101288] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022]
Abstract
Adipose tissue (AT) inflammation is a central mechanism for metabolic dysfunction in both diet-induced obesity and age-associated obesity. Studies in diet-induced obesity have characterized the role of Fetuin A (Fet A) in Free Fatty Acids (FFA)-mediated TLR4 activation and adipose tissue inflammation. However, the role of Fet A & TLR4 in aging-related adipose tissue inflammation is unknown. In the current study, analysis of epidymymal fat pads of C57/Bl6 male mice, we found that, in contrast to data from diet-induced obesity models, adipose tissue from aged mice have normal Fet A and TLR4 expression. Interestingly, aged TLR4-deficient mice have diminished adipose tissue inflammation compared to normal controls. We further demonstrated that reduced AT inflammation in old TLR4-deficient mice is linked to impaired ER stress, augmented autophagy activity, and diminished senescence phenomenon. Importantly, old TLR4-deficient mice have improved glucose tolerance compared to age-matched wild type mice, suggesting that the observed reduced AT inflammation in aged TLR4-deficient mice has important physiological consequences. Taken together, our present study establishes novel aspect of aging-associated AT inflammation that is distinct from diet-induced AT inflammation. Our results also provide strong evidence that TLR4 plays a significant role in promoting aging adipose tissue inflammation.
Collapse
|
1506
|
Bourgeois B, Madl T. Regulation of cellular senescence via the FOXO4-p53 axis. FEBS Lett 2018; 592:2083-2097. [PMID: 29683489 PMCID: PMC6033032 DOI: 10.1002/1873-3468.13057] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023]
Abstract
Forkhead box O (FOXO) and p53 proteins are transcription factors that regulate diverse signalling pathways to control cell cycle, apoptosis and metabolism. In the last decade both FOXO and p53 have been identified as key players in aging, and their misregulation is linked to numerous diseases including cancers. However, many of the underlying molecular mechanisms remain mysterious, including regulation of ageing by FOXOs and p53. Several activities appear to be shared between FOXOs and p53, including their central role in the regulation of cellular senescence. In this review, we will focus on the recent advances on the link between FOXOs and p53, with a particular focus on the FOXO4‐p53 axis and the role of FOXO4/p53 in cellular senescence. Moreover, we discuss potential strategies for targeting the FOXO4‐p53 interaction to modulate cellular senescence as a drug target in treatment of aging‐related diseases and morbidity.
Collapse
Affiliation(s)
- Benjamin Bourgeois
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.,BioTechMed, Graz, Austria
| |
Collapse
|
1507
|
Li M, You L, Xue J, Lu Y. Ionizing Radiation-Induced Cellular Senescence in Normal, Non-transformed Cells and the Involved DNA Damage Response: A Mini Review. Front Pharmacol 2018; 9:522. [PMID: 29872395 PMCID: PMC5972185 DOI: 10.3389/fphar.2018.00522] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 04/30/2018] [Indexed: 02/05/2023] Open
Abstract
Cellular senescence is identified by a living cell in irreversible and persistent cell cycle arrest in response to various cellular stresses. Senescent cells secrete senescence-associated secretory phenotype factors that can amplify cellular senescence and alter the microenvironments. Radiotherapy, via ionizing radiation, serves as an effective treatment for local tumor control with side effects on normal cells, which can induce inflammation and fibrosis in irradiated and nearby regions. Research has revealed that senescent phenotype is observable in irradiated organs. This process starts with DNA damage mediated by radiation, after which a G2 arrest occurs in virtually all eukaryotic cells and a mitotic bypass is possibly necessary to ultimately establish cellular senescence. Within this complex DNA damage response signaling network, ataxia telangiectasia-mutated protein, p53, and p21 stand out as the crucial mediators. Senolytic agents, a class of small molecules that can selectively kill senescent cells, hold great potential to substantially reduce the side effects caused by radiotherapy while reasonably steer clear of carcinogenesis.
Collapse
Affiliation(s)
- Mengqian Li
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Liting You
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - You Lu
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
1508
|
Saleh T, Tyutynuk-Massey L, Cudjoe EK, Idowu MO, Landry JW, Gewirtz DA. Non-Cell Autonomous Effects of the Senescence-Associated Secretory Phenotype in Cancer Therapy. Front Oncol 2018; 8:164. [PMID: 29868482 PMCID: PMC5968105 DOI: 10.3389/fonc.2018.00164] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 04/30/2018] [Indexed: 12/24/2022] Open
Abstract
In addition to promoting various forms of cell death, most conventional anti-tumor therapies also promote senescence. There is now extensive evidence that therapy-induced senescence (TIS) might be transient, raising the concern that TIS could represent an undesirable outcome of therapy by providing a mechanism for tumor dormancy and eventual disease recurrence. The senescence-associated secretory phenotype (SASP) is a hallmark of TIS and may contribute to aberrant effects of cancer therapy. Here, we propose that the SASP may also serve as a major driver of escape from senescence and the re-emergence of proliferating tumor cells, wherein factors secreted from the senescent cells contribute to the restoration of tumor growth in a non-cell autonomous fashion. Accordingly, anti-SASP therapies might serve to mitigate the deleterious outcomes of TIS. In addition to providing an overview of the putative actions of the SASP, we discuss recent efforts to identify and eliminate senescent tumor cells.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Liliya Tyutynuk-Massey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Emmanuel K Cudjoe
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, United States
| | - Michael O Idowu
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, United States
| | - Joseph W Landry
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States.,Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
1509
|
Kouroumalis A, Mavrogonatou E, Savvidou OD, Papagelopoulos PJ, Pratsinis H, Kletsas D. Major traits of the senescent phenotype of nucleus pulposus intervertebral disc cells persist under the specific microenvironmental conditions of the tissue. Mech Ageing Dev 2018; 177:118-127. [PMID: 29778758 DOI: 10.1016/j.mad.2018.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/23/2018] [Accepted: 05/16/2018] [Indexed: 01/25/2023]
Abstract
Intervertebral discs (IVDs) are the joints of the spine, mainly consisting of extracellular matrix (ECM) with a low number of cells embedded therein. Low cellularity stems from nutrient deprivation due to the lack of blood supply, as well as from the hypoxic and hyperosmotic conditions prevailing in the tissue. Intervertebral disc degeneration (IDD) has been firmly connected with low back pain, a major age-related disease, whereas degenerated discs have been characterized by increased proteolytic activity and accumulation of senescent cells. While the catabolic phenotype of senescent IVD cells has been documented, whether this phenotype is preserved under the harsh conditions met in the IVD milieu has never been investigated. Here we showed that a combination of low glucose, hypoxia, high osmolality and absence of serum is anti-proliferative for young disc cells. In addition, we demonstrated for the first time that classical senescence markers, namely p16INK4a, p21WAF1 and ICAM-1, remain up-regulated in senescent cells under these conditions. Finally, up-regulation of the main senescence-associated ECM degrading enzymes, i.e. MMP-1, -2 and -3 was maintained in this strict environment. Conservation of IVD cells' senescent phenotype under the actual conditions these cells are confronted with in vivo further supports their possible implication in IDD.
Collapse
Affiliation(s)
- Anastasios Kouroumalis
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Eleni Mavrogonatou
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Olga D Savvidou
- The First Department of Orthopaedic Surgery, National and Kapodistrian University of Athens, Medical School, ATTIKON University Hospital, Athens, Greece
| | - Panayiotis J Papagelopoulos
- The First Department of Orthopaedic Surgery, National and Kapodistrian University of Athens, Medical School, ATTIKON University Hospital, Athens, Greece
| | - Harris Pratsinis
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece.
| |
Collapse
|
1510
|
Hall BM, Balan V, Gleiberman AS, Strom E, Krasnov P, Virtuoso LP, Rydkina E, Vujcic S, Balan K, Gitlin II, Leonova KI, Consiglio CR, Gollnick SO, Chernova OB, Gudkov AV. p16(Ink4a) and senescence-associated β-galactosidase can be induced in macrophages as part of a reversible response to physiological stimuli. Aging (Albany NY) 2018; 9:1867-1884. [PMID: 28768895 PMCID: PMC5611982 DOI: 10.18632/aging.101268] [Citation(s) in RCA: 273] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/22/2017] [Indexed: 12/22/2022]
Abstract
Constitutive p16Ink4a expression, along with senescence-associated β-galactosidase (SAβG), are commonly accepted biomarkers of senescent cells (SCs). Recent reports attributed improvement of the healthspan of aged mice following p16Ink4a-positive cell killing to the eradication of accumulated SCs. However, detection of p16Ink4a/SAβG-positive macrophages in the adipose tissue of old mice and in the peritoneal cavity of young animals following injection of alginate-encapsulated SCs has raised concerns about the exclusivity of these markers for SCs. Here we report that expression of p16Ink4a and SAβG in macrophages is acquired as part of a physiological response to immune stimuli rather than through senescence, consistent with reports that p16Ink4a plays a role in macrophage polarization and response. Unlike SCs, p16Ink4a/SAβG-positive macrophages can be induced in p53-null mice. Macrophages, but not mesenchymal SCs, lose both markers in response to M1- [LPS, IFN-α, Poly(I:C)] and increase their expression in response to M2-inducing stimuli (IL-4, IL-13). Moreover, interferon-inducing agent Poly(I:C) dramatically reduced p16Ink4a expression in vivo in our alginate bead model and in the adipose tissue of aged mice. These observations suggest that the antiaging effects following eradication of p16Ink4a-positive cells may not be solely attributed to SCs but also to non-senescent p16Ink4a/SAβG-positive macrophages.
Collapse
Affiliation(s)
| | - Vitaly Balan
- Everon Biosciences, Inc., Buffalo, NY 14203, USA
| | | | | | | | | | | | | | - Karina Balan
- Everon Biosciences, Inc., Buffalo, NY 14203, USA
| | - Ilya I Gitlin
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Katerina I Leonova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Camila R Consiglio
- Department of Tumor Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Sandra O Gollnick
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | - Andrei V Gudkov
- Everon Biosciences, Inc., Buffalo, NY 14203, USA.,Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
1511
|
Paredes S, Angulo-Ibanez M, Tasselli L, Carlson SM, Zheng W, Li TM, Chua KF. The epigenetic regulator SIRT7 guards against mammalian cellular senescence induced by ribosomal DNA instability. J Biol Chem 2018; 293:11242-11250. [PMID: 29728458 DOI: 10.1074/jbc.ac118.003325] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 04/21/2018] [Indexed: 12/16/2022] Open
Abstract
In the yeast Saccharomyces cerevisiae, genomic instability in rDNA repeat sequences is an underlying cause of cell aging and is suppressed by the chromatin-silencing factor Sir2. In humans, rDNA instability is observed in cancers and premature aging syndromes, but its underlying mechanisms and functional consequences remain unclear. Here, we uncovered a pivotal role of sirtuin 7 (SIRT7), a mammalian Sir2 homolog, in guarding against rDNA instability and show that this function of SIRT7 protects against senescence in primary human cells. We found that, mechanistically, SIRT7 is required for association of SNF2H (also called SMARCA5, SWI/SNF-related matrix-associated actin-dependent regulator of chromatin, subfamily A, member 5), a component of the nucleolar heterochromatin-silencing complex NoRC, with rDNA sequences. Defective rDNA-heterochromatin silencing in SIRT7-deficient cells unleashed rDNA instability, with excision and loss of rDNA gene copies, which in turn induced acute senescence. Mounting evidence indicates that accumulation of senescent cells significantly contributes to tissue dysfunction in aging-related pathologies. Our findings identify rDNA instability as a driver of mammalian cellular senescence and implicate SIRT7-dependent heterochromatin silencing in protecting against this process.
Collapse
Affiliation(s)
- Silvana Paredes
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Maria Angulo-Ibanez
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Luisa Tasselli
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Scott M Carlson
- Department of Biology, Stanford University, Stanford, California 94305
| | - Wei Zheng
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Tie-Mei Li
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Katrin F Chua
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, .,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| |
Collapse
|
1512
|
Briot A, Decaunes P, Volat F, Belles C, Coupaye M, Ledoux S, Bouloumié A. Senescence Alters PPARγ (Peroxisome Proliferator–Activated Receptor Gamma)-Dependent Fatty Acid Handling in Human Adipose Tissue Microvascular Endothelial Cells and Favors Inflammation. Arterioscler Thromb Vasc Biol 2018; 38:1134-1146. [DOI: 10.1161/atvbaha.118.310797] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 03/01/2018] [Indexed: 02/06/2023]
Abstract
Objective—
Adipose tissue (AT) dysfunction associated with obesity or aging is a major cause for lipid redistribution and the progression of cardiometabolic disorders. Our goal is to decipher the contribution of human AT microvascular endothelial cells (ECs) in the maintenance of fatty acid (FA) fluxes and the impact of senescence on their function.
Approach and Results—
We used freshly isolated primary microvascular ECs from human AT. Our data identified the endothelial FA handling machinery including FATPs (FA transport proteins) FATP1, FATP3, FATP4, and CD36 as well as FABP4 (FA binding protein 4). We showed that PPARγ (peroxisome proliferator–activated receptor gamma) regulates the expression of FATP1, CD36, and FABP4 and is a major regulator of FA uptake in human AT EC (hATEC). We provided evidence that endothelial PPARγ activity is modulated by senescence. Indeed, the positive regulation of FA transport by PPARγ agonist was abolished, whereas the emergence of an inflammatory response was favored in senescent hATEC. This was associated with the retention of nuclear FOXO1 (forkhead box protein O1), whereas nuclear PPARγ translocation was impaired.
Conclusions—
These data support the notion that PPARγ is a key regulator of primary hATEC function including FA handling and inflammatory response. However, the outcome of PPARγ activation is modulated by senescence, a phenomenon that may impact the ability of hATEC to properly respond to and handle lipid fluxes. Finally, our work highlights the role of hATEC in the regulation of FA fluxes and reveals that dysfunction of these cells with accelerated aging is likely to participate to AT dysfunction and the redistribution of lipids.
Collapse
Affiliation(s)
- Anaïs Briot
- From the Inserm, UMR1048, Team 1, I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, Toulouse, Cedex 4, France (A. Briot, P.D., F.V., C.B., A. Bouloumié)
| | - Pauline Decaunes
- From the Inserm, UMR1048, Team 1, I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, Toulouse, Cedex 4, France (A. Briot, P.D., F.V., C.B., A. Bouloumié)
| | - Fanny Volat
- From the Inserm, UMR1048, Team 1, I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, Toulouse, Cedex 4, France (A. Briot, P.D., F.V., C.B., A. Bouloumié)
| | - Chloé Belles
- From the Inserm, UMR1048, Team 1, I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, Toulouse, Cedex 4, France (A. Briot, P.D., F.V., C.B., A. Bouloumié)
| | - Muriel Coupaye
- Center Support of Obesity, Hôpital Louis Mourier (APHP), Colombes, and Faculté Paris Diderot, France (M.C., S.L.)
| | - Séverine Ledoux
- Center Support of Obesity, Hôpital Louis Mourier (APHP), Colombes, and Faculté Paris Diderot, France (M.C., S.L.)
| | - Anne Bouloumié
- From the Inserm, UMR1048, Team 1, I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, Toulouse, Cedex 4, France (A. Briot, P.D., F.V., C.B., A. Bouloumié)
| |
Collapse
|
1513
|
Logan S, Owen D, Chen S, Chen WJ, Ungvari Z, Farley J, Csiszar A, Sharpe A, Loos M, Koopmans B, Richardson A, Sonntag WE. Simultaneous assessment of cognitive function, circadian rhythm, and spontaneous activity in aging mice. GeroScience 2018; 40:123-137. [PMID: 29687240 DOI: 10.1007/s11357-018-0019-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 04/11/2018] [Indexed: 12/27/2022] Open
Abstract
Cognitive function declines substantially with age in both humans and animal models. In humans, this decline is associated with decreases in independence and quality of life. Although the methodology for analysis of cognitive function in human models is relatively well established, similar analyses in animal models have many technical issues (e.g., unintended experimenter bias, motivational issues, stress, and testing during the light phase of the light dark cycle) that limit interpretation of the results. These caveats, and others, potentially bias the interpretation of studies in rodents and prevent the application of current tests of learning and memory as part of an overall healthspan assessment in rodent models of aging. The goal of this study was to establish the methodology to assess cognitive function in aging animals that addresses many of these concerns. Here, we use a food reward-based discrimination procedure with minimal stress in C57Bl/6J male mice at 6, 21, and 27 months of age, followed by a reversal task to assess behavioral flexibility. Importantly, the procedures minimize issues related to between-experimenter confounds and are conducted during both the dark and light phases of the light dark cycle in a home-cage setting. During cognitive testing, we were able to assess multiple measures of spontaneous movement and diurnal activity in young and aged mice including, distance moved, velocity, and acceleration over a 90-h period. Both initial discrimination and reversal learning significantly decreased with age and, similar to rats and humans, not all old mice demonstrated impairments in learning with age. These results permitted classification of animals based on their cognitive status. Analysis of movement parameters indicated decreases in distance moved as well as velocity and acceleration with increasing age. Based on these data, we developed preliminary models indicating, as in humans, a close relationship exists between age-related movement parameters and cognitive ability. Our results provide a reliable method for assessing cognitive performance with minimal stress and simultaneously provide key information on movement and diurnal activity. These methods represent a novel approach to developing non-invasive healthspan measures in rodent models that allow standardization across laboratories.
Collapse
Affiliation(s)
- Sreemathi Logan
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10TH Street, SLY-BRC 1303, Oklahoma City, OK, 73104, USA.
| | - Daniel Owen
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10TH Street, SLY-BRC 1303, Oklahoma City, OK, 73104, USA
| | - Sixia Chen
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Wei-Jen Chen
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10TH Street, SLY-BRC 1303, Oklahoma City, OK, 73104, USA
| | - Julie Farley
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10TH Street, SLY-BRC 1303, Oklahoma City, OK, 73104, USA
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10TH Street, SLY-BRC 1303, Oklahoma City, OK, 73104, USA
| | - Amanda Sharpe
- College of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Maarten Loos
- Sylics (Synaptologics BV), Amsterdam, The Netherlands
| | | | - Arlan Richardson
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10TH Street, SLY-BRC 1303, Oklahoma City, OK, 73104, USA
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10TH Street, SLY-BRC 1303, Oklahoma City, OK, 73104, USA
| |
Collapse
|
1514
|
Gonzalez-Meljem JM, Apps JR, Fraser HC, Martinez-Barbera JP. Paracrine roles of cellular senescence in promoting tumourigenesis. Br J Cancer 2018; 118:1283-1288. [PMID: 29670296 PMCID: PMC5959857 DOI: 10.1038/s41416-018-0066-1] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 03/02/2018] [Accepted: 03/02/2018] [Indexed: 12/26/2022] Open
Abstract
Senescent cells activate genetic programmes that irreversibly inhibit cellular proliferation, but also endow these cells with distinctive metabolic and signalling phenotypes. Although senescence has historically been considered a protective mechanism against tumourigenesis, the activities of senescent cells are increasingly being associated with age-related diseases, including cancer. An important feature of senescent cells is the secretion of a vast array of pro-inflammatory cytokines, chemokines, and growth factors collectively known as the senescence-associated secretory phenotype (SASP). Recent research has shown that SASP paracrine signalling can mediate several pro-tumourigenic effects, such as enhancing malignant phenotypes and promoting tumour initiation. In this review, we summarise the paracrine activities of senescent cells and their role in tumourigenesis through direct effects on growth and proliferation of tumour cells, tumour angiogenesis, invasion and metastasis, cellular reprogramming and emergence of tumour-initiating cells, and tumour interactions with the local immune environment. The evidence described here suggests cellular senescence acts as a double-edged sword in cancer pathogenesis, which demands further attention in order to support the use of senolytic or SASP-modulating compounds for cancer treatment.
Collapse
Affiliation(s)
- Jose Mario Gonzalez-Meljem
- Developmental Biology and Cancer Research Programme, UCL Great Ormond Street Institute of Child Health, Guilford Street, London, WC1N 1EH, UK.,Basic Research Department, Instituto Nacional de Geriatría, Anillo Periférico 2767, Magdalena Contreras, 10200, Mexico City, Mexico
| | - John Richard Apps
- Developmental Biology and Cancer Research Programme, UCL Great Ormond Street Institute of Child Health, Guilford Street, London, WC1N 1EH, UK
| | - Helen Christina Fraser
- Developmental Biology and Cancer Research Programme, UCL Great Ormond Street Institute of Child Health, Guilford Street, London, WC1N 1EH, UK
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Research Programme, UCL Great Ormond Street Institute of Child Health, Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
1515
|
Grezella C, Fernandez-Rebollo E, Franzen J, Ventura Ferreira MS, Beier F, Wagner W. Effects of senolytic drugs on human mesenchymal stromal cells. Stem Cell Res Ther 2018; 9:108. [PMID: 29669575 PMCID: PMC5907463 DOI: 10.1186/s13287-018-0857-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/26/2018] [Accepted: 03/28/2018] [Indexed: 02/08/2023] Open
Abstract
Background Senolytic drugs are thought to target senescent cells and might thereby rejuvenate tissues. In fact, such compounds were suggested to increase health and lifespan in various murine aging models. So far, effects of senolytic drugs have not been analysed during replicative senescence of human mesenchymal stromal cells (MSCs). Methods In this study, we tested four potentially senolytic drugs: ABT-263 (navitoclax), quercetin, nicotinamide riboside, and danazol. The effects of these compounds were analysed during long-term expansion of MSCs, until replicative senescence. Furthermore, we determined the effect on molecular markers for replicative senescence, such as senescence-associated beta-galactosidase staining (SA-β-gal), telomere attrition, and senescence-associated DNA methylation changes. Results Co-culture experiments of fluorescently labelled early and late passages revealed that particularly ABT-263 had a significant but moderate senolytic effect. This was in line with reduced SA-β-gal staining in senescent MSCs upon treatment with ABT-263. However, none of the drugs had significant effects on the maximum number of population doublings, telomere length, or epigenetic senescence predictions. Conclusions Of the four tested drugs, only ABT-263 revealed a senolytic effect in human MSCs—and even treatment with this compound did not rejuvenate MSCs with regard to telomere length or epigenetic senescence signature. It will be important to identify more potent senolytic drugs to meet the high hopes for regenerative medicine. Electronic supplementary material The online version of this article (10.1186/s13287-018-0857-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Clara Grezella
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstraße 20, 52074, Aachen, Germany
| | - Eduardo Fernandez-Rebollo
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstraße 20, 52074, Aachen, Germany
| | - Julia Franzen
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstraße 20, 52074, Aachen, Germany
| | - Mónica Sofia Ventura Ferreira
- Department for Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Fabian Beier
- Department for Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstraße 20, 52074, Aachen, Germany.
| |
Collapse
|
1516
|
|
1517
|
Lämmermann I, Terlecki-Zaniewicz L, Weinmüllner R, Schosserer M, Dellago H, de Matos Branco AD, Autheried D, Sevcnikar B, Kleissl L, Berlin I, Morizot F, Lejeune F, Fuzzati N, Forestier S, Toribio A, Tromeur A, Weinberg L, Higareda Almaraz JC, Scheideler M, Rietveld M, El Ghalbzouri A, Tschachler E, Gruber F, Grillari J. Blocking negative effects of senescence in human skin fibroblasts with a plant extract. NPJ Aging Mech Dis 2018; 4:4. [PMID: 29675264 PMCID: PMC5895844 DOI: 10.1038/s41514-018-0023-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 03/13/2018] [Accepted: 03/19/2018] [Indexed: 12/26/2022] Open
Abstract
There is increasing evidence that senescent cells are a driving force behind many age-related pathologies and that their selective elimination increases the life- and healthspan of mice. Senescent cells negatively affect their surrounding tissue by losing their cell specific functionality and by secreting a pro-tumorigenic and pro-inflammatory mixture of growth hormones, chemokines, cytokines and proteases, termed the senescence-associated secretory phenotype (SASP). Here we identified an extract from the plant Solidago virgaurea subsp. alpestris, which exhibited weak senolytic activity, delayed the acquisition of a senescent phenotype and induced a papillary phenotype with improved functionality in human dermal fibroblasts. When administered to stress-induced premature senescent fibroblasts, this extract changed their global mRNA expression profile and particularly reduced the expression of various SASP components, thereby ameliorating the negative influence on nearby cells. Thus, the investigated plant extract represents a promising possibility to block age-related loss of tissue functionality.
Collapse
Affiliation(s)
- Ingo Lämmermann
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Lucia Terlecki-Zaniewicz
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Regina Weinmüllner
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Markus Schosserer
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Hanna Dellago
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - André Dargen de Matos Branco
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Dominik Autheried
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Benjamin Sevcnikar
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Lisa Kleissl
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Irina Berlin
- Department of Biology and Women Beauty, Chanel R&T, Pantin, France
| | | | - Francois Lejeune
- Department of Biology and Women Beauty, Chanel R&T, Pantin, France
| | | | | | | | | | | | - Juan Carlos Higareda Almaraz
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research, Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcel Scheideler
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research, Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marion Rietveld
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Abdoel El Ghalbzouri
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erwin Tschachler
- Division for Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Florian Gruber
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Division for Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
1518
|
Childs BG, Li H, van Deursen JM. Senescent cells: a therapeutic target for cardiovascular disease. J Clin Invest 2018; 128:1217-1228. [PMID: 29608141 DOI: 10.1172/jci95146] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cellular senescence, a major tumor-suppressive cell fate, has emerged from humble beginnings as an in vitro phenomenon into recognition as a fundamental mechanism of aging. In the process, senescent cells have attracted attention as a therapeutic target for age-related diseases, including cardiovascular disease (CVD), the leading cause of morbidity and mortality in the elderly. Given the aging global population and the inadequacy of current medical management, attenuating the health care burden of CVD would be transformative to clinical practice. Here, we review the evidence that cellular senescence drives CVD in a bimodal fashion by both priming the aged cardiovascular system for disease and driving established disease forward. Hence, the growing field of senotherapy (neutralizing senescent cells for therapeutic benefit) is poised to contribute to both prevention and treatment of CVD.
Collapse
Affiliation(s)
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, and
| | - Jan M van Deursen
- Department of Biochemistry and Molecular Biology.,Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
1519
|
Baker DJ, Petersen RC. Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives. J Clin Invest 2018; 128:1208-1216. [PMID: 29457783 PMCID: PMC5873891 DOI: 10.1172/jci95145] [Citation(s) in RCA: 351] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Along with a general decline in overall health, most chronic degenerative human diseases are inherently associated with increasing age. Age-associated cognitive impairments and neurodegenerative diseases, such as Parkinson's and Alzheimer's diseases, are potentially debilitating conditions that lack viable options for treatment, resulting in a tremendous economic and societal cost. Most high-profile clinical trials for neurodegenerative diseases have led to inefficacious results, suggesting that novel approaches to treating these pathologies are needed. Numerous recent studies have demonstrated that senescent cells, which are characterized by sustained cell cycle arrest and production of a distinct senescence-associated secretory phenotype, accumulate with age and at sites of age-related diseases throughout the body, where they actively promote tissue deterioration. Cells with features of senescence have been detected in the context of brain aging and neurodegenerative disease, suggesting that they may also promote dysfunction. Here, we discuss the evidence implicating senescent cells in neurodegenerative diseases, the mechanistic contribution of these cells that may actively drive neurodegeneration, and how these cells or their effects may be targeted therapeutically.
Collapse
Affiliation(s)
- Darren J. Baker
- Department of Biochemistry and Molecular Biology
- Department of Pediatric and Adolescent Medicine, and
| | | |
Collapse
|
1520
|
Abstract
Cellular senescence is a physiological phenomenon that has both beneficial and detrimental consequences. Senescence limits tumorigenesis and tissue damage throughout the lifetime. However, at the late stages of life, senescent cells increasingly accumulate in tissues and might also contribute to the development of various age-related pathologies. Recent studies have revealed the molecular pathways that preserve the viability of senescent cells and the ones regulating their immune surveillance. These studies provide essential initial insights for the development of novel therapeutic strategies for targeting senescent cells. At the same time they stress the need to understand the limitations of the existing strategies, their efficacy and safety, and the possible deleterious consequences of senescent cell elimination. Here we discuss the existing strategies for targeting senescent cells and upcoming challenges in translating these strategies into safe and efficient therapies. Successful translation of these strategies could have implications for treating a variety of diseases at old age and could potentially reshape our view of health management during aging.
Collapse
|
1521
|
Kang DH, Park YS, Lee DY. Senotherapy for attenuation of cellular senescence in aging and organ implantation. J IND ENG CHEM 2018. [DOI: 10.1016/j.jiec.2017.08.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
1522
|
Khosla S, Farr JN, Kirkland JL. Inhibiting Cellular Senescence: A New Therapeutic Paradigm for Age-Related Osteoporosis. J Clin Endocrinol Metab 2018; 103:1282-1290. [PMID: 29425296 PMCID: PMC6276719 DOI: 10.1210/jc.2017-02694] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/02/2018] [Indexed: 12/31/2022]
Abstract
Context With the aging of the population and projected increase in osteoporotic fractures coupled with the declining use of osteoporosis medications, there is a compelling need for new approaches to treat osteoporosis. Given that age-related osteoporosis generally coexists with multiple other comorbidities (e.g., atherosclerosis, diabetes, frailty) that share aging as the leading risk factor, there is growing interest in the "Geroscience Hypothesis," which posits that manipulation of fundamental aging mechanisms will delay the appearance or severity of multiple chronic diseases because these diseases share aging as the underlying risk factor. In this context, one fundamental aging mechanism that has received considerable attention recently as contributing to multiple age-related morbidities is cellular senescence. This mini-review provides an overview on cellular senescence with a focus on its role in mediating age-related bone loss. Methods This summary is based on the authors' knowledge of the field supplemented by a PubMed search using the terms "senescence," "aging," and "bone." Results There is compelling evidence from preclinical models and supportive human data demonstrating an increase in senescent cells in the bone microenvironment with aging. These cells produce a proinflammatory secretome that leads to increased bone resorption and decreased bone formation, and approaches that either eliminate senescent cells or impair the production of their proinflammatory secretome have been shown to prevent age-related bone loss in mice. Conclusions Targeting cellular senescence represents a novel therapeutic strategy to prevent not only bone loss but potentially multiple age-related diseases simultaneously.
Collapse
Affiliation(s)
- Sundeep Khosla
- Division of Endocrinology and Metabolism and Kogod Center on Aging, Mayo
Clinic, Rochester, Minnesota 55905
| | - Joshua N Farr
- Division of Endocrinology and Metabolism and Kogod Center on Aging, Mayo
Clinic, Rochester, Minnesota 55905
| | - James L Kirkland
- Division of Endocrinology and Metabolism and Kogod Center on Aging, Mayo
Clinic, Rochester, Minnesota 55905
| |
Collapse
|
1523
|
Yousefzadeh MJ, Schafer MJ, Noren Hooten N, Atkinson EJ, Evans MK, Baker DJ, Quarles EK, Robbins PD, Ladiges WC, LeBrasseur NK, Niedernhofer LJ. Circulating levels of monocyte chemoattractant protein-1 as a potential measure of biological age in mice and frailty in humans. Aging Cell 2018; 17:e12706. [PMID: 29290100 PMCID: PMC5847863 DOI: 10.1111/acel.12706] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2017] [Indexed: 11/29/2022] Open
Abstract
A serum biomarker of biological versus chronological age would have significant impact on clinical care. It could be used to identify individuals at risk of early-onset frailty or the multimorbidities associated with old age. It may also serve as a surrogate endpoint in clinical trials targeting mechanisms of aging. Here, we identified MCP-1/CCL2, a chemokine responsible for recruiting monocytes, as a potential biomarker of biological age. Circulating monocyte chemoattractant protein-1 (MCP-1) levels increased in an age-dependent manner in wild-type (WT) mice. That age-dependent increase was accelerated in Ercc1-/Δ and Bubr1H/H mouse models of progeria. Genetic and pharmacologic interventions that slow aging of Ercc1-/Δ and WT mice lowered serum MCP-1 levels significantly. Finally, in elderly humans with aortic stenosis, MCP-1 levels were significantly higher in frail individuals compared to nonfrail. These data support the conclusion that MCP-1 can be used as a measure of mammalian biological age that is responsive to interventions that extend healthy aging.
Collapse
Affiliation(s)
- Matthew J. Yousefzadeh
- Department of Molecular MedicineCenter on AgingThe Scripps Research InstituteJupiterFLUSA
| | - Marissa J. Schafer
- Robert and Arlene Kogod Center on AgingMayo Clinic College of MedicineRochesterMNUSA
- Department of Physical Medicine and RehabilitationMayo Clinic College of MedicineRochesterMNUSA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population ScienceNational Institute on AgingNational Institutes of HealthBaltimoreMDUSA
| | - Elizabeth J. Atkinson
- Division of Biomedical Statistics and InformaticsDepartment of Health Sciences ResearchMayo Clinic College of MedicineRochesterMNUSA
| | - Michele K. Evans
- Laboratory of Epidemiology and Population ScienceNational Institute on AgingNational Institutes of HealthBaltimoreMDUSA
| | - Darren J. Baker
- Department of Pediatric and Adolescent MedicineMayo Clinic College of MedicineRochesterMNUSA
| | | | - Paul D. Robbins
- Department of Molecular MedicineCenter on AgingThe Scripps Research InstituteJupiterFLUSA
| | - Warren C. Ladiges
- Department of Comparative MedicineUniversity of WashingtonSeattleWAUSA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on AgingMayo Clinic College of MedicineRochesterMNUSA
- Department of Physical Medicine and RehabilitationMayo Clinic College of MedicineRochesterMNUSA
| | - Laura J. Niedernhofer
- Department of Molecular MedicineCenter on AgingThe Scripps Research InstituteJupiterFLUSA
| |
Collapse
|
1524
|
Current Progress in the Rejuvenation of Aging Stem/Progenitor Cells for Improving the Therapeutic Effectiveness of Myocardial Repair. Stem Cells Int 2018; 2018:9308301. [PMID: 29760740 PMCID: PMC5926481 DOI: 10.1155/2018/9308301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 03/13/2018] [Indexed: 12/31/2022] Open
Abstract
Ischemic heart disease affects a majority of people, especially elderly patients. Recent studies have utilized autologous adult stem/progenitor cells as a treatment option to heal cardiac tissue after myocardial infarction. However, donor cells from aging patients are more likely to be in a senescent stage. Rejuvenation is required to reverse the damage levied by aging and promote a youthful phenotype. This review aims to discuss current strategies that are effective in rejuvenating aging cardiac stem cells and represent novel therapeutic methods to treat the aging heart. Recent literature mainly focuses on three approaches that aim to reverse cardiac aging: genetic modification, pharmaceutical administration, and optimization of extracellular factors. In vitro genetic modification can be used to overexpress or knock down certain genes and allow for reversal of the aging phenotype. Pharmaceutical administration is another approach that allows for manipulation of signaling pathways related to cell proliferation and cell senescence. Since the stem cell niche can contribute to the age-related decline in stem cell function, rejuvenation strategies also include optimization of extracellular factors. Overall, improving the intrinsic properties of aging stem cells as well as the surrounding environment allows these cells to adopt a phenotype similar to their younger counterparts.
Collapse
|
1525
|
Bjørklund G, Dadar M, Martins N, Chirumbolo S, Goh BH, Smetanina K, Lysiuk R. Brief Challenges on Medicinal Plants: An Eye-Opening Look at Ageing-Related Disorders. Basic Clin Pharmacol Toxicol 2018; 122:539-558. [DOI: 10.1111/bcpt.12972] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/15/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine; Mo i Rana Norway
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute; Agricultural Research, Education and Extension Organization (AREEO); Karaj Iran
| | - Natália Martins
- Mountain Research Centre (CIMO), ESA; Polytechnic Institute of Bragança, Campus de Santa Apolónia; Bragança Portugal
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences; University of Verona; Verona Italy
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group (BMEX); School of Pharmacy; Monash University Malaysia; Bandar Sunway Malaysia
- Novel Bacteria and Drug Discovery Research Group (NBDD); School of Pharmacy; Monash University Malaysia; Bandar Sunway Malaysia
- Center of Health Outcomes Research and Therapeutic Safety; School of Pharmaceutical Sciences; University of Phayao; Phayao Thailand
- Asian Centre for Evidence Synthesis in Population; Implementation and Clinical Outcomes; Health and Well-Being Cluster; Global Asia in the 21st Century Platform; Monash University Malaysia; Bandar Sunway Malaysia
| | - Kateryna Smetanina
- Department of Management and Economy of Pharmacy; Postgraduate Faculty; Drug Technology and Pharmacoeconomics; Danylo Halytsky Lviv National Medical University; Lviv Ukraine
| | - Roman Lysiuk
- Department of Pharmacognosy and Botany; Danylo Halytsky Lviv National Medical University; Lviv Ukraine
| |
Collapse
|
1526
|
Copper accumulation in senescent cells: Interplay between copper transporters and impaired autophagy. Redox Biol 2018; 16:322-331. [PMID: 29579719 PMCID: PMC5953000 DOI: 10.1016/j.redox.2018.03.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/24/2022] Open
Abstract
Cellular senescence is characterized by irreversible growth arrest incurred through either replicative exhaustion or by pro-oncogenic cellular stressors (radioactivity, oxidative stress, oncogenic activation). The enrichment of senescent cells in tissues with age has been associated with tissue dyshomeostasis and age-related pathologies including cancers, neurodegenerative disorders (e.g. Alzheimer's, Parkinson's, etc.) and metabolic disorders (e.g. diabetes). We identified copper accumulation as being a universal feature of senescent cells [mouse embryonic fibroblasts (MEF), human prostate epithelial cells and human diploid fibroblasts] in vitro. Elevated copper in senescent MEFs was accompanied by elevated levels of high-affinity copper uptake protein 1 (Ctr1), diminished levels of copper-transporting ATPase 1 (Atp7a) (copper export) and enhanced antioxidant defence reflected by elevated levels of glutathione (GSH), superoxide dismutase 1 (SOD1) and glutaredoxin 1 (Grx1). The levels of intracellular copper were further increased in senescent MEFs cultured in copper supplemented medium and in senescent Mottled Brindled (Mobr) MEFs lacking functional Atp7a. Finally, we demonstrated that the restoration/preservation of autophagic-lysosomal degradation in senescent MEFs following rapamycin treatment correlated with attenuation of copper accumulation in these cells despite a further decrease in Atp7a levels. This study for the first time establishes a link between Atp7a and the autophagic-lysosomal pathway, and a requirement for both to effect efficient copper export. Such a connection between cellular autophagy and copper homeostasis is significant, as both have emerged as important facets of age-associated degenerative disease. Copper accumulation is a feature of cellular senescence. Elevated copper in senescent cells is associated with impaired autophagic-lysosomal function. Restoration/preservation of lysosomal function attenuates copper accumulation. Both Atp7a and autophagic-lysosomal function are required for copper export.
Collapse
|
1527
|
Abstract
When ageing, cells profoundly reprogram to enter a state called senescence. Although the link between senescence and cancer is well established, the nature of this link remains unclear and debated. We will describe in this article the properties of senescent cells and make clear on how they could promote or oppose to cancer initiation and progression. We will also consider senescence as a response to classical anti-cancer therapies and discuss how to take advantage of senescence to improve the efficacy of these therapies while decreasing their toxicity.
Collapse
Affiliation(s)
- Erwan Goy
- Université de Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T -mécanismes de tumorigenèse et thérapies ciblées, F-59000 Lille, France
| | - Corinne Abbadie
- Université de Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T -mécanismes de tumorigenèse et thérapies ciblées, F-59000 Lille, France
| |
Collapse
|
1528
|
Su Y, Wang P, Shen H, Sun Z, Xu C, Li G, Tong T, Chen J. The protein kinase D1-mediated classical protein secretory pathway regulates the Ras oncogene-induced senescence response. J Cell Sci 2018; 131:jcs.207217. [PMID: 29420297 DOI: 10.1242/jcs.207217] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/15/2018] [Indexed: 12/13/2022] Open
Abstract
Senescent cells develop a senescence-associated secretory phenotype (SASP). The factors secreted by cells with a SASP have multiple biological functions that are mediated in an autocrine or paracrine manner. However, the status of the protein kinase D1 (PKD1; also known as PRKD1)-mediated classical protein secretory pathway, from the trans-Golgi network (TGN) to the cell surface, during cellular senescence and its role in the cellular senescence response remain unknown. Here, we show that the activities or quantities of critical components of this pathway, including PKD1, ADP-ribosylation factor 1 (ARF1) and phosphatidylinositol 4-kinase IIIβ (PI4KIIIβ), at the TGN are increased in senescent cells. Blocking of this pathway decreases IL-6 and IL-8 (hereafter IL-6/IL-8) secretion and results in IL-6/IL-8 accumulation in SASP-competent senescent cells. Inhibition of this pathway reduces IL-6/IL-8 secretion during Ras oncogene-induced senescence (OIS), retards Ras OIS and alleviates its associated ER stress and autophagy. Finally, targeting of this pathway triggers cell death in SASP factor-producing senescent cells due to the intracellular accumulation of massive amounts of IL-6/IL-8. Taken together, our results unveil the hyperactive state of the protein secretory pathway in SASP-competent senescent cells and its critical functions in mediating SASP factor secretion and the Ras OIS process, as well as in determining the fate of senescent cells.
Collapse
Affiliation(s)
- Yuanyuan Su
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China
| | - Pengfeng Wang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China
| | - Hong Shen
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China
| | - Zhaomeng Sun
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China
| | - Chenzhong Xu
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China
| | - Guodong Li
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China
| | - Tanjun Tong
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China
| | - Jun Chen
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
1529
|
Katsuumi G, Shimizu I, Yoshida Y, Minamino T. Vascular Senescence in Cardiovascular and Metabolic Diseases. Front Cardiovasc Med 2018; 5:18. [PMID: 29556500 PMCID: PMC5845435 DOI: 10.3389/fcvm.2018.00018] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/21/2018] [Indexed: 01/08/2023] Open
Abstract
In mammals, aging is associated with accumulation of senescent cells. Stresses such as telomere shortening and reactive oxygen species induce “cellular senescence”, which is characterized by growth arrest and alteration of the gene expression profile. Chronological aging is associated with development of age-related diseases, including heart failure, diabetes, and atherosclerotic disease, and studies have shown that accumulation of senescent cells has a causative role in the pathology of these age-related disorders. Endothelial cell senescence has been reported to develop in heart failure and promotes pathologic changes in the failing heart. Senescent endothelial cells and vascular smooth muscle cells are found in atherosclerotic plaque, and studies indicate that these cells are involved in progression of plaque. Diabetes is also linked to accumulation of senescent vascular endothelial cells, while endothelial cell senescence per se induces systemic glucose intolerance by inhibiting skeletal muscle metabolism. A close connection between derangement of systemic metabolism and cellular senescence is also well recognized. Aging is a complex phenomenon, and there is no simple approach to understanding the whole process. However, there is accumulating evidence that cellular senescence has a central role in the development and progression of various undesirable aspects of aging. Suppression of cellular senescence or elimination of senescent cells reverses phenotypic changes of aging in several models, and proof-of-concept has been established that inhibiting accumulation of senescent cells could become a next generation therapy for age-related disorders. It is clear that cellular senescence drives various pathological changes associated with aging. Accordingly, further investigation into the role of this biological process in age-related disorders and discovery of senolytic compounds are important fields for future exploration.
Collapse
Affiliation(s)
- Goro Katsuumi
- 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
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
1530
|
Hoare M, Narita M. The Power Behind the Throne: Senescence and the Hallmarks of Cancer. ANNUAL REVIEW OF CANCER BIOLOGY 2018; 2:175-194. [DOI: 10.1146/annurev-cancerbio-030617-050352] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Cellular senescence is a state of stable proliferative arrest triggered by various stimuli, including oncogenic and other cellular stress. Senescent cells are highly metabolically active and have diverse and profound nonautonomous effects through the senescence-associated secretory phenotype (SASP). It has become increasingly evident that senescent cells can have tumour suppressive or pro-oncogenic effects on adjacent cancer cells and other players in the tumor microenvironment such as the stroma, vasculature, and immune system. Thus, the last decade or so has witnessed a huge leap forward in our understanding of the biology of senescence, promoting it from an autonomous tumor suppressor to a complex, dynamic, and interactive phenotype. It is perhaps not a coincidence that the concept of the “hallmarks of cancer” has also evolved during this period, with the latest iteration ( Hanahan & Weinberg 2011 ) focusing more on the microenvironment. Here, we suggest that cellular senescence could underpin the biology of many of the hallmarks of cancer, making it the true power behind the throne.
Collapse
Affiliation(s)
- Matthew Hoare
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom;,
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom;,
| |
Collapse
|
1531
|
Stress, cell senescence and organismal ageing. Mech Ageing Dev 2018; 170:2-9. [DOI: 10.1016/j.mad.2017.07.001] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/17/2017] [Accepted: 07/04/2017] [Indexed: 12/25/2022]
|
1532
|
Shilovsky GA, Putyatina TS, Ashapkin VV, Luchkina OS, Markov AV. Coefficient of Variation of Lifespan Across the Tree of Life: Is It a Signature of Programmed Aging? BIOCHEMISTRY (MOSCOW) 2018; 82:1480-1492. [PMID: 29486698 DOI: 10.1134/s0006297917120070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Measurements of variation are of great importance for studying the stability of pathological phenomena and processes. For the biology of aging, it is very important not only to determine average mortality, but also to study its stability in time and the size of fluctuations that are indicated by the variation coefficient of lifespan (CVLS). It is believed that a relatively small (~20%) value of CVLS in humans, comparable to the coefficients of variation of other events programmed in ontogenesis (for example, menarche and menopause), indicates a relatively rigid determinism (N. S. Gavrilova et al. (2012) Biochemistry (Moscow), 77, 754-760). To assess the prevalence of this phenomenon, we studied the magnitude of CVLS, as well as the coefficients of skewness and kurtosis in diverse representatives of the animal kingdom using data provided by the Institute for Demographic Research (O. R. Jones et al. (2014) Nature, 505, 169-173). We found that, unlike humans and laboratory animals, in most examined species the values of CVLS are rather high, indicating heterogeneity of the lifespan in the cohorts studied. This is probably due to the large influence of background mortality, as well as the non-monotonicity of total mortality in the wild, especially at the earliest ages. One way to account for this influence is to "truncate" the data (removing the earliest and latest ages from consideration). To reveal the effect of this procedure, we proposed a new indicator, the stability coefficient of mortality dynamics, which indicates how quickly CVLS is reduced to values that characterize a relatively homogeneous population (33%) when the data are "truncated". Such indicators facilitate the use of the parameters of survival curves for analysis of the effects of geroprotectors, lifestyle, and other factors on lifespan, and for the quantification of relative contributions of genetic and environmental factors to the dynamics of aging in human and animal populations, including those living in the wild.
Collapse
Affiliation(s)
- G A Shilovsky
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | | | | | | | | |
Collapse
|
1533
|
Nelson G, Kucheryavenko O, Wordsworth J, von Zglinicki T. The senescent bystander effect is caused by ROS-activated NF-κB signalling. Mech Ageing Dev 2018; 170:30-36. [PMID: 28837845 PMCID: PMC5861994 DOI: 10.1016/j.mad.2017.08.005] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/13/2017] [Accepted: 08/14/2017] [Indexed: 12/27/2022]
Abstract
Cell senescence is an important driver of the ageing process. The accumulation of senescent cells in tissues is accelerated by stress signals from senescent cells that induce DNA damage and ultimately senescence in bystander cells. We examine here the interplay of senescence-associated mitochondrial dysfunction (SAMD)-driven production of reactive oxygen species (ROS) and senescence-associated secretory phenotype (SASP) in causing the bystander effect. We show that in various modes of fibroblast senescence ROS are necessary and sufficient to activate the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which facilitates a large part of the SASP. This ROS-NF-κB axis causes the DNA damage response in bystander cells. Cytokines IL-6 and IL-8 are major components of the pro-inflammatory SASP in senescent fibroblasts. However, their activation in senescence is only partially controlled by NF-κB, and they are thus not strong candidates as intercellular mediators of the bystander effect as mediated by the ROS-NF-κB axis.
Collapse
Affiliation(s)
- Glyn Nelson
- The Ageing Biology Centre, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Olena Kucheryavenko
- The Ageing Biology Centre, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - James Wordsworth
- The Ageing Biology Centre, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Thomas von Zglinicki
- The Ageing Biology Centre, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| |
Collapse
|
1534
|
|
1535
|
van Willigenburg H, de Keizer PLJ, de Bruin RWF. Cellular senescence as a therapeutic target to improve renal transplantation outcome. Pharmacol Res 2018; 130:322-330. [PMID: 29471104 DOI: 10.1016/j.phrs.2018.02.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/02/2018] [Accepted: 02/12/2018] [Indexed: 01/18/2023]
Abstract
Kidney transplants from aged donors are more vulnerable to ischemic injury, suffer more from delayed graft function and have a lower graft survival compared to kidneys from younger donors. On a cellular level, aging results in an increase in cells that are in a permanent cell cycle arrest, termed senescence, which secrete a range of pro-inflammatory cytokines and growth factors. Consequently, these senescent cells negatively influence the local milieu by causing inflammaging, and by reducing the regenerative capacity of the kidney. Moreover, the oxidative damage that is inflicted by ischemia-reperfusion injury during transplantation can induce senescence and accelerate aging. In this review, we describe recent developments in the understanding of the biology of aging that have led to the development of a new class of therapeutic agents aimed at eliminating senescent cells. These compounds have already shown to be able to restore tissue homeostasis in old mice, improve kidney function and general health- and lifespan. Use of these anti-senescence compounds holds great promise to improve the quality of marginal donor kidneys as well as to remove senescent cells induced by ischemia-reperfusion injury. Altogether, senescent cell removal may increase the donor pool, relieving the growing organ shortage and improve long-term transplantation outcome.
Collapse
Affiliation(s)
- Hester van Willigenburg
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Peter L J de Keizer
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Ron W F de Bruin
- Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
1536
|
Senescence-associated secretory factors induced by cisplatin in melanoma cells promote non-senescent melanoma cell growth through activation of the ERK1/2-RSK1 pathway. Cell Death Dis 2018; 9:260. [PMID: 29449532 PMCID: PMC5833767 DOI: 10.1038/s41419-018-0303-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/25/2017] [Accepted: 01/10/2018] [Indexed: 12/12/2022]
Abstract
Although targeted therapy and immunotherapy greatly improve the outcome of melanoma, drug resistance and low response rates still maintain the unsubstitutability of traditional chemotherapy. Cisplatin (CDDP) is widely used in different types of tumours with high response rates, but it generally has low efficiency in melanoma. The mechanisms underpinning the phenomena are not sufficiently understood. Here we demonstrated that various melanoma cell lines adopted senescence phenotype after CDDP treatment in contrast to the other types of tumour cells. CDDP treatment induced melanoma A375 cells into senescence through the sequential activation of the DNA damage response and the P53/P21 pathway. All the senescent melanoma cells induced by CDDP alone or the combination of CDDP and dacarbazine developed robust senescence-associated secretory phenotype (SASP), that is, the secretion of multiple cytokines. IL-1α was an early component and an upstream regulator of SASP. Similarly, CDDP either alone or combined with dacarbazine could induce melanoma cell senescence and SASP in either A375 or B16F10 melanoma xenograft mice. The supernatant of senescent A375 cells promoted the growth of normal non-senescent A375 cells and enhanced their expression and secretion of IL-8 through the activation of the ERK1/2-RSK1 pathway. The transplantation of non-senescent and senescent A375 cells together into nude mice showed accelerated tumour growth compared with transplanting non-senescent cells alone; no tumours developed when transplanting senescent cells alone. Following CDDP administration in A375-bearing mice, the intratumour injection of neutralisation antibodies targeting the SASP factors IL-1α or IL-8 evidently delayed tumour growth. The results suggest that the CDDP-induced senescent melanoma cells promote non-senescent cells proliferation through the activation of ERK1/2-RSK1 pathway by the SASP factors. Cell senescence and concomitant SASP may be the particular mechanisms for melanoma to resist chemotherapeutics.
Collapse
|
1537
|
|
1538
|
Inducers of Senescence, Toxic Compounds, and Senolytics: The Multiple Faces of Nrf2-Activating Phytochemicals in Cancer Adjuvant Therapy. Mediators Inflamm 2018; 2018:4159013. [PMID: 29618945 PMCID: PMC5829354 DOI: 10.1155/2018/4159013] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/19/2017] [Indexed: 12/18/2022] Open
Abstract
The reactivation of senescence in cancer and the subsequent clearance of senescent cells are suggested as therapeutic intervention in the eradication of cancer. Several natural compounds that activate Nrf2 (nuclear factor erythroid-derived 2-related factor 2) pathway, which is involved in complex cytoprotective responses, have been paradoxically shown to induce cell death or senescence in cancer. Promoting the cytoprotective Nrf2 pathway may be desirable for chemoprevention, but it might be detrimental in later stages and advanced cancers. However, senolytic activity shown by some Nrf2-activating compounds could be used to target senescent cancer cells (particularly in aged immune-depressed organisms) that escape immunosurveillance. We herein describe in vitro and in vivo effects of fifteen Nrf2-interacting natural compounds (tocotrienols, curcumin, epigallocatechin gallate, quercetin, genistein, resveratrol, silybin, phenethyl isothiocyanate, sulforaphane, triptolide, allicin, berberine, piperlongumine, fisetin, and phloretin) on cellular senescence and discuss their use in adjuvant cancer therapy. In light of available literature, it can be concluded that the meaning and the potential of adjuvant therapy with natural compounds in humans remain unclear, also taking into account the existence of few clinical trials mostly characterized by uncertain results. Further studies are needed to investigate the therapeutic potential of those compounds that display senolytic activity.
Collapse
|
1539
|
Liu A, Guo E, Yang J, Yang Y, Liu S, Jiang X, Hu Q, Dirsch O, Dahmen U, Zhang C, Gewirtz DA, Fang H. Young plasma reverses age-dependent alterations in hepatic function through the restoration of autophagy. Aging Cell 2018; 17. [PMID: 29210183 PMCID: PMC5770779 DOI: 10.1111/acel.12708] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2017] [Indexed: 12/13/2022] Open
Abstract
Recent studies showing the therapeutic effect of young blood on aging‐associated deterioration of organs point to young blood as the solution for clinical problems related to old age. Given that defective autophagy has been implicated in aging and aging‐associated organ injuries, this study was designed to determine the effect of young blood on aging‐induced alterations in hepatic function and underlying mechanisms, with a focus on autophagy. Aged rats (22 months) were treated with pooled plasma (1 ml, intravenously) collected from young (3 months) or aged rats three times per week for 4 weeks, and 3‐methyladenine or wortmannin was used to inhibit young blood‐induced autophagy. Aging was associated with elevated levels of alanine transaminase and aspartate aminotransferase, lipofuscin accumulation, steatosis, fibrosis, and defective liver regeneration after partial hepatectomy, which were significantly attenuated by young plasma injections. Young plasma could also restore aging‐impaired autophagy activity. Inhibition of the young plasma‐restored autophagic activity abrogated the beneficial effect of young plasma against hepatic injury with aging. In vitro, young serum could protect old hepatocytes from senescence, and the antisenescence effect of young serum was abrogated by 3‐methyladenine, wortmannin, or small interfering RNA to autophagy‐related protein 7. Collectively, our data indicate that young plasma could ameliorate age‐dependent alterations in hepatic function partially via the restoration of autophagy.
Collapse
Affiliation(s)
- Anding Liu
- Experimental Medicine Center; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Enshuang Guo
- Department of Infectious Diseases; Wuhan General Hospital; Wuhan China
| | - Jiankun Yang
- Experimental Medicine Center; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Yan Yang
- Experimental Medicine Center; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Shenpei Liu
- Experimental Medicine Center; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Xiaojing Jiang
- Department of Infectious Diseases; Wuhan General Hospital; Wuhan China
| | - Qi Hu
- Department of Geriatrics; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Olaf Dirsch
- Experimental Transplantation Surgery; Department of General, Visceral and Vascular Surgery; Friedrich-Schiller-University Jena; Jena Germany
| | - Uta Dahmen
- Experimental Transplantation Surgery; Department of General, Visceral and Vascular Surgery; Friedrich-Schiller-University Jena; Jena Germany
| | - Cuntai Zhang
- Department of Geriatrics; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - David A Gewirtz
- Department of Pharmacology and Toxicology; Massey Cancer Center; Virginia Commonwealth University; Richmond VA USA
| | - Haoshu Fang
- Department of Pathophysiology; Anhui Medical University; Hefei China
| |
Collapse
|
1540
|
Santoro A, Spinelli CC, Martucciello S, Nori SL, Capunzo M, Puca AA, Ciaglia E. Innate immunity and cellular senescence: The good and the bad in the developmental and aged brain. J Leukoc Biol 2018; 103:509-524. [PMID: 29389023 DOI: 10.1002/jlb.3mr0118-003r] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 12/21/2022] Open
Abstract
Ongoing studies evidence cellular senescence in undifferentiated and specialized cells from tissues of all ages. Although it is believed that senescence plays a wider role in several stress responses in the mature age, its participation in certain physiological and pathological processes throughout life is coming to light. The "senescence machinery" has been observed in all brain cell populations, including components of innate immunity (e.g., microglia and astrocytes). As the beneficial versus detrimental implications of senescence is an open question, we aimed to analyze the contribution of immune responses in regulatory mechanisms governing its distinct functions in healthy (development, organogenesis, danger patrolling events) and diseased brain (glioma, neuroinflammation, neurodeneration), and the putative connection between cellular and molecular events governing the 2 states. Particularly this review offers new insights into the complex roles of senescence both as a chronological event as age advances, and as a molecular mechanism of brain homeostasis through the important contribution of innate immune responses and their crosstalk with neighboring cells in brain parenchyma. We also highlight the impact of the recently described glymphatic system and brain lymphatic vasculature in the interplay between peripheral and central immune surveillance and its potential implication during aging. This will open new ways to understand brain development, its deterioration during aging, and the occurrence of several oncological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Antonietta Santoro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| | | | | | - Stefania Lucia Nori
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| | - Mario Capunzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| | - Annibale Alessandro Puca
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy.,Cardiovascular Research Unit, IRCCS MultiMedica, Milan, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| |
Collapse
|
1541
|
Abstract
Senescence is a durable cell cycle arrest that can be induced in response to various stress factors, such as telomere erosion, DNA damage or the aberrant activation of oncogenes. In addition to its well-established role as a stress response programme, research has revealed important physiological roles of senescence in nondisease settings, such as embryonic development, wound healing, tissue repair and ageing. Senescent cells secrete various cytokines, chemokines, matrix remodelling proteases and growth factors, a phenotype collectively referred to as the senescence-associated secretory phenotype. These factors evoke immune responses that, depending on the pathophysiological context, can either prevent or even fuel disease and tumorigenesis. Remarkably, even the gut microbiota can influence senescence in various organs. In this Review, we provide an introduction to cellular senescence, addressed particularly to gastroenterologists and hepatologists, and discuss the implications of senescence for the pathogenesis of malignant and nonmalignant gastrointestinal and hepatobiliary diseases. We conclude with an outlook on how modulation of cellular senescence might be used for therapeutic purposes.
Collapse
|
1542
|
Abstract
PURPOSE OF THE REVIEW Senescent cells have the capacity to both effect and limit fibrosis. Senotherapeutics target senescent cells to improve aging conditions. Here, we review the contexts in which senescent cells mediate wound healing and fibrotic pathology and the potential utility of senotherapeutic drugs for treatment of fibrotic disease. RECENT FINDINGS Multi-action and temporal considerations influence deleterious versus beneficial actions of senescent cells. Acutely generated senescent cells can limit proliferation, and the senescence-associated secretory phenotype (SASP) contains factors that can facilitate tissue repair. Long-lived senescent cells that evade clearance or are generated outside of programmed remodeling can deplete the progenitor pool to exhaust regenerative capacity and through the SASP, stimulate continual activation, leading to disorganized tissue architecture, fibrotic damage, sterile inflammation, and induction of bystander senescence. Senescent cells contribute to fibrotic pathogenesis in multiple tissues, including the liver, kidney, and lung. Senotherapeutics may be a viable strategy for treatment of a range of fibrotic conditions.
Collapse
|
1543
|
Novel treatment strategies for chronic kidney disease: insights from the animal kingdom. Nat Rev Nephrol 2018; 14:265-284. [PMID: 29332935 DOI: 10.1038/nrneph.2017.169] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many of the >2 million animal species that inhabit Earth have developed survival mechanisms that aid in the prevention of obesity, kidney disease, starvation, dehydration and vascular ageing; however, some animals remain susceptible to these complications. Domestic and captive wild felids, for example, show susceptibility to chronic kidney disease (CKD), potentially linked to the high protein intake of these animals. By contrast, naked mole rats are a model of longevity and are protected from extreme environmental conditions through mechanisms that provide resistance to oxidative stress. Biomimetic studies suggest that the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) offers protection in extreme environmental conditions and promotes longevity in the animal kingdom. Similarly, during months of fasting, immobilization and anuria, hibernating bears are protected from muscle wasting, azotaemia, thrombotic complications, organ damage and osteoporosis - features that are often associated with CKD. Improved understanding of the susceptibility and protective mechanisms of these animals and others could provide insights into novel strategies to prevent and treat several human diseases, such as CKD and ageing-associated complications. An integrated collaboration between nephrologists and experts from other fields, such as veterinarians, zoologists, biologists, anthropologists and ecologists, could introduce a novel approach for improving human health and help nephrologists to find novel treatment strategies for CKD.
Collapse
|
1544
|
Hervault M, Clavel MA. Sex-related Differences in Calcific Aortic Valve Stenosis: Pathophysiology, Epidemiology, Etiology, Diagnosis, Presentation, and Outcomes. STRUCTURAL HEART-THE JOURNAL OF THE HEART TEAM 2018. [DOI: 10.1080/24748706.2017.1420273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Maxime Hervault
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| | - Marie-Annick Clavel
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| |
Collapse
|
1545
|
Hwang HV, Tran DT, Rebuffatti MN, Li CS, Knowlton AA. Investigation of quercetin and hyperoside as senolytics in adult human endothelial cells. PLoS One 2018; 13:e0190374. [PMID: 29315311 PMCID: PMC5760026 DOI: 10.1371/journal.pone.0190374] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/13/2017] [Indexed: 01/09/2023] Open
Abstract
Quercetin has been reported to act as a senolytic by selectively removing senescent endothelial cells, and thus it would seem quercetin could revolutionize the field of gerontology. However, given quercetin's narrow therapeutic index reported in work done with human umbilical vein endothelial cells (HUVECs), we hypothesized that quercetin is not innocuous for non-senescent adult human vascular endothelial cells at concentrations that have been reported to be safe for proliferating HUVECs. Furthermore, we investigated quercetin 3-D-galactoside (Q3G; hyperoside), an inactive quercetin derivative that needs to be cleaved by beta-galactosidase overexpressed in senescent cells to release quercetin, as a potential safer senolytic. We compared the effectiveness of quercetin and Q3G in primary human coronary artery endothelial cells (HCAEC), which are adult microvascular cells. We found that quercetin caused cell death in non-senescent endothelial cells at a concentration that has been reported to selectively remove senescent cells, and that Q3G was not cytotoxic to either young or senescent cells. Thus, in primary adult human endothelial cells, quercetin and Q3G are not senolytics. Earlier work reporting positive results was done with HUVECs, and given their origin and the disparate findings from the current study, these may not be the best cells for evaluating potential senolytics in clinically relevant endothelial cells.
Collapse
Affiliation(s)
- HyunTae V. Hwang
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
| | - Darlene Thuy Tran
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
| | - Michelle Nicole Rebuffatti
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
| | - Chin-Shang Li
- Division of Biostatistics, Department of Public Health Sciences, University of California-Davis, Davis, CA, United States of America
| | - Anne A. Knowlton
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
- VA Medical Center, Sacramento, CA, United States of America
- Pharmacology Department, University of California-Davis, Davis, CA, United States of America
- * E-mail:
| |
Collapse
|
1546
|
Abstract
Brown adipose tissue aging and the concomitant loss of thermogenic capacity have been linked to an inability to maintain normal energy homeostasis in late life. Similarly, the ability of white fat to convert into brite/beige adipose tissue declines. This may ultimately exacerbate the progression of age-related metabolic pathologies, such as insulin resistance and obesity. The depletion of all types of brown adipocytes during aging is well-established and has been described in rodent models as well as humans. We here review the available literature on the potential mechanisms leading to cell-autonomous and microenvironment-related aspects of brown adipocyte dysfunction. Among these, cellular senescence, mitochondrial impairment, and deteriorating changes to the local and endocrine microenvironments have been proposed. An important goal of aging research is to develop approaches that may not only extend life expectancy but also prolong health-span. These efforts may also be aimed at maintaining metabolic health throughout life by targeting brown adipocyte function.
Collapse
Affiliation(s)
- Antonia Graja
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Sabrina Gohlke
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany. .,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany. .,German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany.
| |
Collapse
|
1547
|
Cesselli D, Aleksova A, Mazzega E, Caragnano A, Beltrami AP. Cardiac stem cell aging and heart failure. Pharmacol Res 2018; 127:26-32. [DOI: 10.1016/j.phrs.2017.01.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 12/11/2022]
|
1548
|
Leveraging Epigenetics to Enhance the Cellular Response to Chemotherapies and Improve Tumor Immunogenicity. Adv Cancer Res 2018; 138:1-39. [PMID: 29551125 DOI: 10.1016/bs.acr.2018.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer chemotherapeutic drugs have greatly advanced our ability to successfully treat a variety of human malignancies. The different forms of stress produced by these agents in cancer cells result in both cell autonomous and cell nonautonomous effects. Desirable cell autonomous effects include reduced proliferative potential, cellular senescence, and cell death. More recently recognized cell nonautonomous effects, usually in the form of stimulating an antitumor immune response, have significant roles in therapeutic efficiency for a select number of chemotherapies. Unfortunately, the success of these therapeutics is not universal as not all tumors respond to treatment, and those that do respond will frequently relapse into therapy-resistant disease. Numerous strategies have been developed to sensitize tumors toward chemotherapies as a means to either improve initial responses, or serve as a secondary treatment strategy for therapy-resistant disease. Recently, targeting epigenetic regulators has emerged as a viable method of sensitizing tumors to the effects of chemotherapies, many of which are cytotoxic. In this review, we summarize these strategies and propose a path for future progress.
Collapse
|
1549
|
Brooks RW, Robbins PD. Treating Age-Related Diseases with Somatic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1056:29-45. [DOI: 10.1007/978-3-319-74470-4_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
1550
|
Yanai H, Fraifeld VE. The role of cellular senescence in aging through the prism of Koch-like criteria. Ageing Res Rev 2018; 41:18-33. [PMID: 29106993 DOI: 10.1016/j.arr.2017.10.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/11/2017] [Accepted: 10/23/2017] [Indexed: 12/13/2022]
Abstract
Since Hayflick's discovery of cellular senescence (CS), a great volume of knowledge in the field has been accumulated and intensively discussed. Here, we attempted to organize the evidence "for" and "against" the hypothesized causal role of CS in aging. For that purpose, we utilized robust Koch-like logical criteria, based on the assumption that some quantitative relationships between the accumulation of senescent cells and aging rate should exist. If so, it could be expected that (i) the "CS load" would be greater in the premature aging phenotype and lesser in longevity phenotype; (ii) CS would promote age-related diseases, and (iii) the interventions that modulate the levels of senescent cells should also modulate health/lifespan. The analysis shows that CS can be considered a causal factor of aging and an important player in various age-related diseases, though its contribution may greatly vary across species. While the relative impact of senescent cells to aging could overall be rather limited and their elimination is hardly expected to be the "fountain of youth", the potential benefits of the senolytic strategy seems a promising option in combating age-related diseases and extending healthspan.
Collapse
|