501
|
Zhang W, Yang J, Chen Y, Xue R, Mao Z, Lu W, Jiang Y. Lycorine hydrochloride suppresses stress-induced premature cellular senescence by stabilizing the genome of human cells. Aging Cell 2021; 20:e13307. [PMID: 33455051 PMCID: PMC7884038 DOI: 10.1111/acel.13307] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/09/2020] [Accepted: 12/25/2020] [Indexed: 12/11/2022] Open
Abstract
Lycorine, a natural compound isolated from the traditional Chinese medicinal herb Lycoris radiata, exhibits multiple pharmacological effects, such as anti-inflammatory, antiviral, and anticancer effects. Accumulating evidence also indicates that lycorine might hold the potential to treat age-associated Alzheimer's disease. However, whether lycorine is involved in delaying the onset of cellular senescence and its underlying mechanisms has not been determined. Here, we demonstrate that the salt of lycorine, lycorine hydrochloride, significantly suppressed stress-induced premature cellular senescence (SIPS) by ~2-fold, as determined by senescence-associated beta-galactosidase (SA-β-gal) staining and the expression of p16 and p21. In addition, pretreating cells with lycorine hydrochloride significantly inhibited the expression of CXCL1 and IL1α, two factors of the senescence-associated secreted phenotype (SASP) in SIPS cells. Further experiments revealed that lycorine hydrochloride promoted both the homologous recombination (HR) and nonhomologous end joining (NHEJ) pathways of DNA double-strand break (DSB) repair. Mechanistic studies suggested that lycorine hydrochloride treatment promoted the transcription of SIRT1 and SIRT6, critical longevity genes positively regulating both HR and NHEJ repair pathways, thereby stimulating DSB repair and stabilizing genomes. Inhibiting SIRT1 enzymatic activity abrogated the protective effect of lycorine hydrochloride on delaying the onset of SIPS, repairing DSBs, and restoring genome integrity. In summary, our work indicates that lycorine hydrochloride might hold therapeutic potential for treating age-associated diseases or promoting healthy aging by stabilizing genomes.
Collapse
Affiliation(s)
- Weina Zhang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Jiaqing Yang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Yu Chen
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Renhao Xue
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Zhiyong Mao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Wen Lu
- Department of Gynecology of Shanghai First Maternity & Infant Hospital Tongji University School of Medicine Shanghai China
| | - Ying Jiang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| |
Collapse
|
502
|
Senescence under appraisal: hopes and challenges revisited. Cell Mol Life Sci 2021; 78:3333-3354. [PMID: 33439271 PMCID: PMC8038995 DOI: 10.1007/s00018-020-03746-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/20/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023]
Abstract
In recent years, cellular senescence has become the focus of attention in multiple areas of biomedical research. Typically defined as an irreversible cell cycle arrest accompanied by increased cellular growth, metabolic activity and by a characteristic messaging secretome, cellular senescence can impact on multiple physiological and pathological processes such as wound healing, fibrosis, cancer and ageing. These unjustly called 'zombie cells' are indeed a rich source of opportunities for innovative therapeutic development. In this review, we collate the current understanding of the process of cellular senescence and its two-faced nature, i.e. beneficial/detrimental, and reason this duality is linked to contextual aspects. We propose the senescence programme as an endogenous pro-resolving mechanism that may lead to sustained inflammation and damage when dysregulated or when senescent cells are not cleared efficiently. This pro-resolving model reconciles the paradoxical two faces of senescence by emphasising that it is the unsuccessful completion of the programme, and not senescence itself, what leads to pathology. Thus, pro-senescence therapies under the right context, may favour inflammation resolution. We also review the evidence for the multiple therapeutic approaches under development based on senescence, including its induction, prevention, clearance and the use of senolytic and senomorphic drugs. In particular, we highlight the importance of the immune system in the favourable outcome of senescence and the implications of an inefficient immune surveillance in completion of the senescent cycle. Finally, we identify and discuss a number of challenges and existing gaps to encourage and stimulate further research in this exciting and unravelled field, with the hope of promoting and accelerating the clinical success of senescence-based therapies.
Collapse
|
503
|
Santos JC, Ribeiro ML, Gambero A. The Impact of Polyphenols-Based Diet on the Inflammatory Profile in COVID-19 Elderly and Obese Patients. Front Physiol 2021; 11:612268. [PMID: 33584335 PMCID: PMC7874176 DOI: 10.3389/fphys.2020.612268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Abstract
The World Health Organization declared the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-associated disease (coronavirus disease 2019 - COVID-19) as a pandemic in March 2020. COVID-19 is characterized by cytokine storm, acute respiratory distress syndrome (ARDS), and systemic inflammation-related pathology and already kills more than 1.5 million of people worldwide. Since aged and obese COVID-19 patients exhibit an enhanced inflammatory status, they represent a high-risk cluster for rapidly progressive clinical deterioration. These individuals present comorbid disorders and immunosenescence that may promote viral-induced cytokine storm and expression of molecules acting as virus receptor as angiotensin I converting enzyme 2 (ACE2) and CD26 (dipeptidyl-peptidase 4), resulting in respiratory failure and increased morbidity and mortality. A better knowledge of SARS-CoV-2 infection in inflammatory-associated high-risk population is essential in order to develop the therapies needed to combat or prevent severe COVID-19. Here, we review the pathogenesis and clinical implications of inflammatory disorders and disease markers associated to senescence in COVID-19 patients and the emerging evidence to argue that a high intake of polyphenols may have a protective effect on SARS-CoV-2 illness severity.
Collapse
Affiliation(s)
- Juliana Carvalho Santos
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Marcelo Lima Ribeiro
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Bragança Paulista, Brazil
| | - Alessandra Gambero
- Life Science Center, Pontifical Catholic University of Campinas (PUCCAMP), Campinas, Brazil
| |
Collapse
|
504
|
Liao C, Xiao Y, Liu L. The Dynamic Process and Its Dual Effects on Tumors of Therapy-Induced Senescence. Cancer Manag Res 2021; 12:13553-13566. [PMID: 33408525 PMCID: PMC7781229 DOI: 10.2147/cmar.s285083] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence is traditionally considered as stable cell cycle arrest state with other phenotypic alterations including the production of an array of cytokines and growth factors. Cancer cells undergo senescence in response to chemotherapeutic agents, radiotherapy and molecular targeted therapy. This form of senescence is termed therapy-induced senescence (TIS) and represents a desirable target in cancer therapy. Recent studies have shown that cellular senescence is a highly heterogeneous and dynamic process. Apart from being cleared by the immune system, the senescent cancer cells may survive for a long time and escape from senescence state. Notably, these cells even have the potential to regain stem-like state with high aggressiveness that eventually facilitates cancer recurrence. Furthermore, the senescence-associated secretory phenotype (SASP) of senescent cells is not always the same, and could establish immunosuppression and a protumor microenvironment. Given these detrimental effects, senescence-inducing chemotherapy followed by senotherapy (the “one–two punch” approach), has emerged. This combined therapy could mitigate unnecessary side effects of the persistent senescent cells, reduce the toxicity of pro-senescence therapy and prolong the survival of cancer patients, and it has a potential future in the precise treatment of cancer. Herein, we review the complex effects of therapy-induced senescence in cancer and highlight the great promise of two-step strategies in anticancer therapies.
Collapse
Affiliation(s)
- Chenxi Liao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Yin Xiao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| |
Collapse
|
505
|
Robbins PD, Jurk D, Khosla S, Kirkland JL, LeBrasseur NK, Miller JD, Passos JF, Pignolo RJ, Tchkonia T, Niedernhofer LJ. Senolytic Drugs: Reducing Senescent Cell Viability to Extend Health Span. Annu Rev Pharmacol Toxicol 2021; 61:779-803. [PMID: 32997601 PMCID: PMC7790861 DOI: 10.1146/annurev-pharmtox-050120-105018] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Senescence is the consequence of a signaling mechanism activated in stressed cells to prevent proliferation of cells with damage. Senescent cells (Sncs) often develop a senescence-associated secretory phenotype to prompt immune clearance, which drives chronic sterile inflammation and plays a causal role in aging and age-related diseases. Sncs accumulate with age and at anatomical sites of disease. Thus, they are regarded as a logical therapeutic target. Senotherapeutics are a new class of drugs that selectively kill Sncs (senolytics) or suppress their disease-causing phenotypes (senomorphics/senostatics). Since 2015, several senolytics went from identification to clinical trial. Preclinical data indicate that senolytics alleviate disease in numerous organs, improve physical function and resilience, and suppress all causes of mortality, even if administered to the aged. Here, we review the evidence that Sncs drive aging and disease, the approaches to identify and optimize senotherapeutics, and the current status of preclinical and clinical testing of senolytics.
Collapse
Affiliation(s)
- Paul D Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA;
| | - Diana Jurk
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Jordan D Miller
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - João F Passos
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Robert J Pignolo
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA;
| |
Collapse
|
506
|
Coryell PR, Diekman BO, Loeser RF. Mechanisms and therapeutic implications of cellular senescence in osteoarthritis. Nat Rev Rheumatol 2021; 17:47-57. [PMID: 33208917 PMCID: PMC8035495 DOI: 10.1038/s41584-020-00533-7] [Citation(s) in RCA: 420] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 12/13/2022]
Abstract
The development of osteoarthritis (OA) correlates with a rise in the number of senescent cells in joint tissues, and the senescence-associated secretory phenotype (SASP) has been implicated in cartilage degradation and OA. Age-related mitochondrial dysfunction and associated oxidative stress might induce senescence in joint tissue cells. However, senescence is not the only driver of OA, and the mechanisms by which senescent cells contribute to disease progression are not fully understood. Furthermore, it remains uncertain which joint cells and SASP-factors contribute to the OA phenotype. Research in the field has looked at developing therapeutics (namely senolytics and senomorphics) that eliminate or alter senescent cells to stop disease progression and pathogenesis. A better understanding of how senescence contributes to joint dysfunction may enhance the effectiveness of these approaches and provide relief for patients with OA.
Collapse
Affiliation(s)
- Philip R Coryell
- Division of Rheumatology, Allergy, and Immunology, Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Brian O Diekman
- Division of Rheumatology, Allergy, and Immunology, Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Richard F Loeser
- Division of Rheumatology, Allergy, and Immunology, Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
507
|
Targeting senescent cell clearance: An approach to delay aging and age-associated disorders. TRANSLATIONAL MEDICINE OF AGING 2021. [DOI: 10.1016/j.tma.2020.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
508
|
Affiliation(s)
- K Virecoulon Giudici
- Kelly Virecoulon Giudici, Gérontopôle of Toulouse, Institute of Aging, Toulouse University Hospital, Université Toulouse III Paul Sabatier, 37 Allée Jules Guesde, 31000 Toulouse, France, E-mail: Ronda, Hospital Universitario Infanta Sofia, Spain,
| |
Collapse
|
509
|
Mkrtchyan GV, Abdelmohsen K, Andreux P, Bagdonaite I, Barzilai N, Brunak S, Cabreiro F, de Cabo R, Campisi J, Cuervo AM, Demaria M, Ewald CY, Fang EF, Faragher R, Ferrucci L, Freund A, Silva-García CG, Georgievskaya A, Gladyshev VN, Glass DJ, Gorbunova V, de Grey A, He WW, Hoeijmakers J, Hoffmann E, Horvath S, Houtkooper RH, Jensen MK, Jensen MB, Kane A, Kassem M, de Keizer P, Kennedy B, Karsenty G, Lamming DW, Lee KF, MacAulay N, Mamoshina P, Mellon J, Molenaars M, Moskalev A, Mund A, Niedernhofer L, Osborne B, Pak HH, Parkhitko A, Raimundo N, Rando TA, Rasmussen LJ, Reis C, Riedel CG, Franco-Romero A, Schumacher B, Sinclair DA, Suh Y, Taub PR, Toiber D, Treebak JT, Valenzano DR, Verdin E, Vijg J, Young S, Zhang L, Bakula D, Zhavoronkov A, Scheibye-Knudsen M. ARDD 2020: from aging mechanisms to interventions. Aging (Albany NY) 2020; 12:24484-24503. [PMID: 33378272 PMCID: PMC7803558 DOI: 10.18632/aging.202454] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023]
Abstract
Aging is emerging as a druggable target with growing interest from academia, industry and investors. New technologies such as artificial intelligence and advanced screening techniques, as well as a strong influence from the industry sector may lead to novel discoveries to treat age-related diseases. The present review summarizes presentations from the 7th Annual Aging Research and Drug Discovery (ARDD) meeting, held online on the 1st to 4th of September 2020. The meeting covered topics related to new methodologies to study aging, knowledge about basic mechanisms of longevity, latest interventional strategies to target the aging process as well as discussions about the impact of aging research on society and economy. More than 2000 participants and 65 speakers joined the meeting and we already look forward to an even larger meeting next year. Please mark your calendars for the 8th ARDD meeting that is scheduled for the 31st of August to 3rd of September, 2021, at Columbia University, USA.
Collapse
Affiliation(s)
- Garik V. Mkrtchyan
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Pénélope Andreux
- Amazentis SA, EPFL Innovation Park, Bâtiment C, Lausanne, Switzerland
| | - Ieva Bagdonaite
- Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nir Barzilai
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute for Aging Research, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Filipe Cabreiro
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Marco Demaria
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Collin Y. Ewald
- Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute for Technology Zürich, Switzerland
| | - Evandro Fei Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Richard Faragher
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Adam Freund
- Calico Life Sciences, LLC, South San Francisco, CA 94080, USA
| | - Carlos G. Silva-García
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | | | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David J. Glass
- Regeneron Pharmaceuticals, Inc. Tarrytown, NY 10591, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA
| | | | - Wei-Wu He
- Human Longevity Inc., San Diego, CA 92121, USA
| | - Jan Hoeijmakers
- Department of Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eva Hoffmann
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steve Horvath
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Majken K. Jensen
- Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | | | - Alice Kane
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA 94107, USA
| | - Moustapha Kassem
- Molecular Endocrinology Unit, Department of Endocrinology, University Hospital of Odense and University of Southern Denmark, Odense, Denmark
| | - Peter de Keizer
- Department of Molecular Cancer Research, Center for Molecular Medicine, Division of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Brian Kennedy
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University Singapore, Singapore
- Centre for Healthy Ageing, National University Healthy System, Singapore
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison and William S. Middleton Memorial Veterans Hospital, Madison, WI 53792, USA
| | - Kai-Fu Lee
- Sinovation Ventures and Sinovation AI Institute, Beijing, China
| | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Denmark
| | - Polina Mamoshina
- Deep Longevity Inc., Hong Kong Science and Technology Park, Hong Kong
| | - Jim Mellon
- Juvenescence Limited, Douglas, Isle of Man, UK
| | - Marte Molenaars
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Alexey Moskalev
- Institute of Biology of FRC Komi Science Center of Ural Division of RAS, Syktyvkar, Russia
| | - Andreas Mund
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Brenna Osborne
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison and William S. Middleton Memorial Veterans Hospital, Madison, WI 53792, USA
| | | | - Nuno Raimundo
- Institute of Cellular Biochemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Thomas A. Rando
- Department of Neurology and Neurological Sciences and Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Christian G. Riedel
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | | | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, Cologne, Germany
| | - David A. Sinclair
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA 94107, USA
- Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Yousin Suh
- Departments of Obstetrics and Gynecology, Genetics and Development, Columbia University, New York, NY 10027, USA
| | - Pam R. Taub
- Division of Cardiovascular Medicine, University of California, San Diego, CA 92093, USA
| | - Debra Toiber
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jonas T. Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Lei Zhang
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Alex Zhavoronkov
- Insilico Medicine, Hong Kong Science and Technology Park, Hong Kong
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
510
|
Wong A, Kieu T, Robbins PD. The Ercc 1-/Δ mouse model of accelerated senescence and aging for identification and testing of novel senotherapeutic interventions. Aging (Albany NY) 2020; 12:24481-24483. [PMID: 33353886 PMCID: PMC7803498 DOI: 10.18632/aging.202321] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/13/2020] [Indexed: 02/05/2023]
Abstract
Progeroid murine models represent an emerging tool to investigate mechanisms of aging in an expedient and efficient manner. One prominent mechanism of aging is the accumulation of DNA damage and subsequent increase in cellular senescence, leading to age related pathologies. Ercc1-/Δ hypomorphic mice, which have a reduced level of the ERCC1-XPF DNA repair endonuclease complex, accumulate spontaneously occurring endogenous DNA damage similar to naturally aged mice, but at a faster rate. The resulting genomic damage gives rise to a senescent cell burden that is comparable to that of a naturally aged mouse. In fact, the expression of senescence and senescence-associated secretory phenotype (SASP) markers in 4-5-month-old Ercc1-/Δ mice, along with other measurements of senescence, were equivalent and never exceeded the extent of that found in naturally aged mice. Furthermore, many features of both natural murine aging and human aging are present in Ercc1-/Δ mice. An emerging use of these mice is the ability to study age-related signaling pathways, including identifying different types of senescent cells and their key senescent cell anti-apoptotic pathways (SCAPs). Most importantly, this model represents a rapid, cost-effective mouse model for the evaluation in vivo of senolytic drugs and other gerotherapeutics.
Collapse
Affiliation(s)
- Alec Wong
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tra Kieu
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
511
|
Gruendler R, Hippe B, Sendula Jengic V, Peterlin B, Haslberger AG. Nutraceutical Approaches of Autophagy and Neuroinflammation in Alzheimer's Disease: A Systematic Review. Molecules 2020; 25:molecules25246018. [PMID: 33353228 PMCID: PMC7765980 DOI: 10.3390/molecules25246018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Aging and the emergence of age-associated illnesses are one of the major challenges of our present society. Alzheimer’s disease (AD) is closely associated with aging and is defined by increasing memory loss and severe dementia. Currently, there are no therapy options available that halt AD progression. This work investigates three hallmarks of the disease (autophagy, neuroinflammation, and senescence) and systematically analyzes if there is a beneficial effect from three substances derived from food sources, the so called “nutraceuticals” epigallocatechin gallate, fisetin, and spermidine, on these hallmarks. The results imply a positive outlook for the reviewed substances to qualify as a novel treatment option for AD. A combination of nutraceutical substances and other preventive measures could have significant clinical impact in a multi-layered therapy approach to counter AD.
Collapse
Affiliation(s)
- Reinhard Gruendler
- Department of Pharmacology and Toxicology, University of Vienna, A-1090 Vienna, Austria;
| | - Berit Hippe
- Department of Nutritional Sciences, University of Vienna, A-1090 Vienna, Austria;
| | | | | | - Alexander G. Haslberger
- Department of Nutritional Sciences, University of Vienna, A-1090 Vienna, Austria;
- Correspondence:
| |
Collapse
|
512
|
Henze L, Walter U, Murua Escobar H, Junghanss C, Jaster R, Köhling R, Lange F, Salehzadeh-Yazdi A, Wolkenhauer O, Hamed M, Barrantes I, Palmer D, Möller S, Kowald A, Heussen N, Fuellen G. Towards biomarkers for outcomes after pancreatic ductal adenocarcinoma and ischaemic stroke, with focus on (co)-morbidity and ageing/cellular senescence (SASKit): protocol for a prospective cohort study. BMJ Open 2020; 10:e039560. [PMID: 33334830 PMCID: PMC7747584 DOI: 10.1136/bmjopen-2020-039560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Ageing-related processes such as cellular senescence are believed to underlie the accumulation of diseases in time, causing (co)morbidity, including cancer, thromboembolism and stroke. Interfering with these processes may delay, stop or reverse morbidity. The aim of this study is to investigate the link between (co)morbidity and ageing by exploring biomarkers and molecular mechanisms of disease-triggered deterioration in patients with pancreatic ductal adenocarcinoma (PDAC) and (thromboembolic) ischaemic stroke (IS). METHODS AND ANALYSIS We will recruit 50 patients with PDAC, 50 patients with (thromboembolic) IS and 50 controls at Rostock University Medical Center, Germany. We will gather routine blood data, clinical performance measurements and patient-reported outcomes at up to seven points in time, alongside in-depth transcriptomics and proteomics at two of the early time points. Aiming for clinically relevant biomarkers, the primary outcome is a composite of probable sarcopenia, clinical performance (described by ECOG Performance Status for patients with PDAC and the Modified Rankin Scale for patients with stroke) and quality of life. Further outcomes cover other aspects of morbidity such as cognitive decline and of comorbidity such as vascular or cancerous events. The data analysis is comprehensive in that it includes biostatistics and machine learning, both following standard role models and additional explorative approaches. Prognostic and predictive biomarkers for interventions addressing senescence may become available if the biomarkers that we find are specifically related to ageing/cellular senescence. Similarly, diagnostic biomarkers will be explored. Our findings will require validation in independent studies, and our dataset shall be useful to validate the findings of other studies. In some of the explorative analyses, we shall include insights from systems biology modelling as well as insights from preclinical animal models. We anticipate that our detailed study protocol and data analysis plan may also guide other biomarker exploration trials. ETHICS AND DISSEMINATION The study was approved by the local ethics committee (Ethikkommission an der Medizinischen Fakultät der Universität Rostock, A2019-0174), registered at the German Clinical Trials Register (DRKS00021184), and results will be published following standard guidelines.
Collapse
Affiliation(s)
- Larissa Henze
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center and Research Focus Oncology, Rostock, Germany
| | - Uwe Walter
- Department of Neurology, Rostock University Medical Center and Centre for Transdisciplinary Neurosciences Rostock, Rostock, Germany
| | - Hugo Murua Escobar
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center and Research Focus Oncology, Rostock, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center and Research Focus Oncology, Rostock, Germany
| | - Robert Jaster
- Department of Gastroenterology, Rostock University Medical Center and Research Focus Oncology, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, Rostock University Medical Center and Centre for Transdisciplinary Neurosciences Rostock and Ageing of Individuals and Society, Interdisciplinary Faculty, Rostock University, Rostock, Germany
| | - Falko Lange
- Oscar Langendorff Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | - Ali Salehzadeh-Yazdi
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock and Centre for Transdisciplinary Neurosciences Rostock, Rostock University Medical Center, Rostock, Germany
| | - Mohamed Hamed
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center and Research Focus Oncology, Rostock, Germany
| | - Israel Barrantes
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center and Research Focus Oncology, Rostock, Germany
| | - Daniel Palmer
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Steffen Möller
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Axel Kowald
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Nicole Heussen
- Department of Medical Statistics, RWTH Aachen, Aachen, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center and Centre for Transdisciplinary Neurosciences Rostock and Research Focus Oncology, Rostock and Ageing of Individuals and Society, Interdisciplinary Faculty, Rostock University, Rostock, Germany
| |
Collapse
|
513
|
Zhang L, Tong X, Huang J, Wu M, Zhang S, Wang D, Liu S, Fan H. Fisetin Alleviated Bleomycin-Induced Pulmonary Fibrosis Partly by Rescuing Alveolar Epithelial Cells From Senescence. Front Pharmacol 2020; 11:553690. [PMID: 33381023 PMCID: PMC7768023 DOI: 10.3389/fphar.2020.553690] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis is an aging-associated disease, satisfactory therapies are not yet available. Accelerated senescence of alveolar epithelial cells plays an important part in Idiopathic pulmonary fibrosis pathogenesis. Fisetin (FIS) is a natural non-toxic flavonoid, which has many pharmacological functions. However, the role of FIS in pulmonary fibrosis has not been established. In this study, we found that FIS treatment apparently alleviated BLM-induced weight loss, inflammatory cells infiltration, inflammatory factors expression, collagen deposition and alveolar epithelial cell senescence, along with AMPK activation and the down regulation of NF-κB and TGF-β/Smad3 in vivo. In vitro, FIS administration significantly inhibited the senescence of alveolar epithelial cells and senescence-associated secretory phenotype, followed by reduced transdifferentiation of fibroblasts to myofibroblasts as well as collagen deposition in fibroblasts, which was blocked by an AMPK inhibitor, Compound C. Together, these results suggest that FIS can alleviate the development of BLM-induced pulmonary fibrosis, which is related to the inhibition of TGF-β/Smad3 signaling and the reduction of alveolar epithelium cell senescence by regulating AMPK/NF-κB signaling pathway. FIS may be a promising candidate for patients with pulmonary fibrosis.
Collapse
Affiliation(s)
- Li Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Chengdu, China
| | - Xiang Tong
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Chengdu, China
| | - Jizhen Huang
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Chengdu, China
| | - Man Wu
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Chengdu, China
| | - Shijie Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Chengdu, China
| | - Dongguang Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Chengdu, China
| | - SiTong Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Chengdu, China
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Chengdu, China
| |
Collapse
|
514
|
Adamczyk-Grochala J, Lewinska A. Nano-Based Theranostic Tools for the Detection and Elimination of Senescent Cells. Cells 2020; 9:E2659. [PMID: 33322013 PMCID: PMC7764355 DOI: 10.3390/cells9122659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
The progressive accumulation of apoptosis-resistant and secretory active senescent cells (SCs) in animal and human aged tissues may limit lifespan and healthspan and lead to age-related diseases such as cancer, neurodegenerative disorders, and metabolic syndrome. Thus, SCs are suggested targets in anti-aging therapy. In the last two decades, a number of nanomaterials have gained much attention as innovative tools in theranostic applications due to their unique properties improving target visualization, drug and gene delivery, controlled drug release, effective diagnosis, and successful therapy. Although the healthcare industry has focused on a plethora of applications of nanomaterials, it remains elusive how nanomaterials may modulate cellular senescence, a hallmark of aging. In this review paper, we consider novel nanotechnology-based strategies for healthspan promotion and the prevention of age-related dysfunctions that are based on the delivery of therapeutic compounds capable to preferentially killing SCs (nano-senolytics) and/or modulating a proinflammatory secretome (nano-senomorphics/nano-senostatics). Recent examples of SC-targeted nanomaterials and the mechanisms underlying different aspects of the nanomaterial-mediated senolysis are presented and discussed.
Collapse
Affiliation(s)
- Jagoda Adamczyk-Grochala
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Anna Lewinska
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| |
Collapse
|
515
|
Abstract
Bien que la sénescence cellulaire joue un rôle essentiel dans le développent embryonnaire, la cicatrisation ou l’hémostase, il est maintenant également démontré qu’elle est à l’origine de nombreux processus dégénératifs qui caractérisent le vieillissement. Cette sénescence est induite en réponse à divers stress ou stimulus inappropriés, conduisant à un arrêt de la prolifération et des adaptations géniques, épigénétiques, métaboliques, structurelles et fonctionnelles. Ces cellules sénescentes, lorsqu’elles ne sont pas éliminées, favorisent la propagation de leur phénotype de proche en proche dans le tissu environnant, par l’établissement d’un profil sécrétoire spécifique. Éliminer ou bloquer l’action de ces cellules par des agents dits sénothérapeutiques pourrait prévenir la dégénérescence tissulaire et améliorer la longévité en bonne santé. Nous nous proposons dans cette revue de présenter les dernières avancées et applications développées en sénothérapie et discuterons les résultats très prometteurs des premiers essais cliniques chez l’homme.
Collapse
|
516
|
Yeh CH, Chou YJ, Kao CH, Tsai TF. Mitochondria and Calcium Homeostasis: Cisd2 as a Big Player in Cardiac Ageing. Int J Mol Sci 2020; 21:ijms21239238. [PMID: 33287440 PMCID: PMC7731030 DOI: 10.3390/ijms21239238] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/27/2022] Open
Abstract
The ageing of human populations has become a problem throughout the world. In this context, increasing the healthy lifespan of individuals has become an important target for medical research and governments. Cardiac disease remains the leading cause of morbidity and mortality in ageing populations and results in significant increases in healthcare costs. Although clinical and basic research have revealed many novel insights into the pathways that drive heart failure, the molecular mechanisms underlying cardiac ageing and age-related cardiac dysfunction are still not fully understood. In this review we summarize the most updated publications and discuss the central components that drive cardiac ageing. The following characters of mitochondria-related dysfunction have been identified during cardiac ageing: (a) disruption of the integrity of mitochondria-associated membrane (MAM) contact sites; (b) dysregulation of energy metabolism and dynamic flexibility; (c) dyshomeostasis of Ca2+ control; (d) disturbance to mitochondria–lysosomal crosstalk. Furthermore, Cisd2, a pro-longevity gene, is known to be mainly located in the endoplasmic reticulum (ER), mitochondria, and MAM. The expression level of Cisd2 decreases during cardiac ageing. Remarkably, a high level of Cisd2 delays cardiac ageing and ameliorates age-related cardiac dysfunction; this occurs by maintaining correct regulation of energy metabolism and allowing dynamic control of metabolic flexibility. Together, our previous studies and new evidence provided here highlight Cisd2 as a novel target for developing therapies to promote healthy ageing
Collapse
Affiliation(s)
- Chi-Hsiao Yeh
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Linko 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan
| | - Yi-Ju Chou
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 350, Taiwan;
| | - Cheng-Heng Kao
- Center of General Education, Chang Gung University, Taoyuan 333, Taiwan
- Correspondence: (C.-H.K.); (T.-F.T.); Tel.: +886-3-211-8800 (ext. 5149) (C.-H.K.); +886-2-2826-7293 (T.-F.T.); Fax: +886-3-211-8700 (C.-H.K.); +886-2-2828-0872 (T.-F.T.)
| | - Ting-Fen Tsai
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 350, Taiwan;
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei 112, Taiwan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 350, Taiwan
- Aging and Health Research Center, National Yang-Ming University, Taipei 112, Taiwan
- Correspondence: (C.-H.K.); (T.-F.T.); Tel.: +886-3-211-8800 (ext. 5149) (C.-H.K.); +886-2-2826-7293 (T.-F.T.); Fax: +886-3-211-8700 (C.-H.K.); +886-2-2828-0872 (T.-F.T.)
| |
Collapse
|
517
|
Kaur J, Farr JN. Cellular senescence in age-related disorders. Transl Res 2020; 226:96-104. [PMID: 32569840 PMCID: PMC7572662 DOI: 10.1016/j.trsl.2020.06.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023]
Abstract
Much of the population is now faced with an enormous burden of age-associated chronic diseases. Recent discoveries in geroscience indicate that healthspan in model organisms such as mice can be manipulated by targeting cellular senescence, a hallmark mechanism of aging, defined as an irreversible proliferative arrest that occurs when cells experience oncogenic or other diverse forms of damage. Senescent cells and their proinflammatory secretome have emerged as contributors to age-related tissue dysfunction and morbidity. Cellular senescence has causal roles in mediating osteoporosis, frailty, cardiovascular diseases, osteoarthritis, pulmonary fibrosis, renal diseases, neurodegenerative diseases, hepatic steatosis, and metabolic dysfunction. Therapeutically targeting senescent cells in mice can prevent, delay, or alleviate each of these conditions. Therefore, senotherapeutic approaches, including senolytics and senomorphics, that either selectively eliminate senescent cells or interfere with their ability to promote tissue dysfunction, are gaining momentum as potential realistic strategies to abrogate human senescence to thereby compress morbidity and extend healthspan.
Collapse
Affiliation(s)
- Japneet Kaur
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester Minnesota; Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester Minnesota
| | - Joshua N Farr
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester Minnesota; Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester Minnesota; Division of Physiology and Biomedical Engineering; Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
518
|
Song S, Tchkonia T, Jiang J, Kirkland JL, Sun Y. Targeting Senescent Cells for a Healthier Aging: Challenges and Opportunities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002611. [PMID: 33304768 PMCID: PMC7709980 DOI: 10.1002/advs.202002611] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/11/2020] [Indexed: 05/02/2023]
Abstract
Aging is a physiological decline in both structural homeostasis and functional integrity, progressively affecting organismal health. A major hallmark of aging is the accumulation of senescent cells, which have entered a state of irreversible cell cycle arrest after experiencing inherent or environmental stresses. Although cellular senescence is essential in several physiological events, it plays a detrimental role in a large array of age-related pathologies. Recent biomedical advances in specifically targeting senescent cells to improve healthy aging, or alternatively, postpone natural aging and age-related diseases, a strategy termed senotherapy, have attracted substantial interest in both scientific and medical communities. Challenges for aging research are highlighted and potential avenues that can be leveraged for therapeutic interventions to control aging and age-related disorders in the current era of precision medicine.
Collapse
Affiliation(s)
- Shuling Song
- Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthShanghai Institutes for Biological SciencesUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
- School of GerontologyBinzhou Medical UniversityYantaiShandong264003China
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMN55905USA
| | - Jing Jiang
- School of PharmacyBinzhou Medical UniversityYantaiShandong264003China
| | - James L. Kirkland
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMN55905USA
| | - Yu Sun
- Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthShanghai Institutes for Biological SciencesUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
- School of PharmacyBinzhou Medical UniversityYantaiShandong264003China
- Department of Medicine and VAPSHCSUniversity of WashingtonSeattleWA98195USA
| |
Collapse
|
519
|
Yan L, Jia Q, Cao H, Chen C, Xing S, Huang Y, Shen D. Fisetin ameliorates atherosclerosis by regulating PCSK9 and LOX-1 in apoE -/- mice. Exp Ther Med 2020; 21:25. [PMID: 33262811 PMCID: PMC7690243 DOI: 10.3892/etm.2020.9457] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
The purpose of the current study was to investigate the mechanism by which fisetin improves atherosclerosis (AS) by regulating lipid metabolism and senescence in apolipoprotein E-deficient (apoE-/-) mice. An AS model was established by feeding apoE-/- mice a high-fat diet. Mice were randomly divided into the model group (n=18), the fisetin group (n=18) and the atorvastatin group (n=18). The control group (n=18) was composed of wild-type C57BL/6 mice of the same age and genetic background. The fisetin and atorvastatin groups were respectively treated with aqueous solutions of fisetin (12.5 mg/kg) and atorvastatin (2 mg/kg) via oral gavage daily for 12 weeks. The pathological morphology, lipid accumulation, collagen deposition of the aortic sinus were observed, serum lipids, superoxide dismutase (SOD) and malondialdehyde (MDA) levels and alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities were measured in the peripheral blood serum. Additionally, the expressions of proprotein convertase subtilisin/kexin type 9 (PCSK9), lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), tumor suppressor protein p53 (p53), cyclin-dependent kinase inhibitor 1A (p21) and multiple tumor suppressor-1 (p16) were analyzed in the aorta. The results of the current study indicated that compared with the control group, a large area of AS plaque in the aortic sinus that contained a large amount of red-stained lipids and decreased collagen fiber content were found in the model group, which exhibited higher total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), oxidized low-density lipoprotein (ox-LDL) and MDA levels; higher ALT and AST activities, lower high-density lipoprotein cholesterol (HDL-C) and SOD levels and increased expression levels of PCSK9, LOX-1, p53, p21 and p16. Fisetin is a phytochemical and bioflavonoid that serves a potential role in chronic diseases including AS, obesity, diabetes and cancer due to its wide biological activities, such as regulating lipid metabolism and anti-aging, anti-oxidation and anti-inflammatory. Atorvastatin is recognized as a first-line treatment drug for AS; therefore it was used as a positive control in the current study. Following fisetin and atorvastatin treatment, both the AS plaque and the lipid accumulation in the aortic sinus were significantly reduced, and the expressions of PCSK9, LOX-1 and aging markers, including p53, p21 and p16 were downregulated.
Collapse
Affiliation(s)
- Li Yan
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Qingling Jia
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Hui Cao
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Sanli Xing
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Yan Huang
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Dingzhu Shen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| |
Collapse
|
520
|
The use of geroprotectors to prevent multimorbidity: Opportunities and challenges. Mech Ageing Dev 2020; 193:111391. [PMID: 33144142 DOI: 10.1016/j.mad.2020.111391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Over 60 % of people over the age of 65 will suffer from multiple diseases concomitantly but the common approach is to treat each disease separately. As age-associated diseases have common underlying mechanisms there is potential to tackle many diseases with the same pharmacological intervention. These are known as geroprotectors and could overcome the problems related to polypharmacy seen with the use of the single disease model. With some geroprotectors now reaching the end stage of preclinical studies and early clinical trials, there is a need to review the evidence and assess how they can be translated practically and effectively into routine practice. Despite promising evidence, there are many gaps and challenges in our understanding that must be addressed to make geroprotective medicine effective in the treatment of age-associated multimorbidity. Here we highlight the key barriers to clinical translation and discuss whether geroprotectors such as metformin, rapamycin and senolytics can tackle all age-associated diseases at the same dose, or whether a more nuanced approach is required. The evidence suggests that geroprotectors' mode of action may differ in different tissues or in response to different inducers of accelerating ageing, suggesting that a blunt 'one drug for many diseases' approach may not work. We make the case for the use of artificial intelligence to better understand multimorbidity, allowing identification of clusters and networks of diseases that are significantly associated beyond chance and the underpinning molecular pathway of ageing causal to each cluster. This will allow us to better understand the development of multimorbidity, select a more homogenous group of patients for intervention, match them with the appropriate geroprotector and identify biomarkers specific to the cluster.
Collapse
|
521
|
Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med 2020; 288:518-536. [PMID: 32686219 PMCID: PMC7405395 DOI: 10.1111/joim.13141] [Citation(s) in RCA: 633] [Impact Index Per Article: 126.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/31/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
Senolytics are a class of drugs that selectively clear senescent cells (SC). The first senolytic drugs Dasatinib, Quercetin, Fisetin and Navitoclax were discovered using a hypothesis-driven approach. SC accumulate with ageing and at causal sites of multiple chronic disorders, including diseases accounting for the bulk of morbidity, mortality and health expenditures. The most deleterious SC are resistant to apoptosis and have up-regulation of anti-apoptotic pathways which defend SC against their own inflammatory senescence-associated secretory phenotype (SASP), allowing them to survive, despite killing neighbouring cells. Senolytics transiently disable these SCAPs, causing apoptosis of those SC with a tissue-destructive SASP. Because SC take weeks to reaccumulate, senolytics can be administered intermittently - a 'hit-and-run' approach. In preclinical models, senolytics delay, prevent or alleviate frailty, cancers and cardiovascular, neuropsychiatric, liver, kidney, musculoskeletal, lung, eye, haematological, metabolic and skin disorders as well as complications of organ transplantation, radiation and cancer treatment. As anticipated for agents targeting the fundamental ageing mechanisms that are 'root cause' contributors to multiple disorders, potential uses of senolytics are protean, potentially alleviating over 40 conditions in preclinical studies, opening a new route for treating age-related dysfunction and diseases. Early pilot trials of senolytics suggest they decrease senescent cells, reduce inflammation and alleviate frailty in humans. Clinical trials for diabetes, idiopathic pulmonary fibrosis, Alzheimer's disease, COVID-19, osteoarthritis, osteoporosis, eye diseases and bone marrow transplant and childhood cancer survivors are underway or beginning. Until such studies are done, it is too early for senolytics to be used outside of clinical trials.
Collapse
Affiliation(s)
- J L Kirkland
- From the, Mayo Clinic Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - T Tchkonia
- From the, Mayo Clinic Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| |
Collapse
|
522
|
Abstract
The prevalence of osteoarthritis (OA) and the burden associated with the disease are steadily increasing worldwide, representing a major public health challenge for the coming decades. The lack of specific treatments for OA has led to it being recognized as a serious disease that has an unmet medical need. Advances in the understanding of OA pathophysiology have enabled the identification of a variety of potential therapeutic targets involved in the structural progression of OA, some of which are promising and under clinical investigation in randomized controlled trials. Emerging therapies include those targeting matrix-degrading proteases or senescent chondrocytes, promoting cartilage repair or limiting bone remodelling, local low-grade inflammation or Wnt signalling. In addition to these potentially disease-modifying OA drugs (DMOADs), several targets are being explored for the treatment of OA-related pain, such as nerve growth factor inhibitors. The results of these studies are expected to considerably reshape the landscape of OA management over the next few years. This Review describes the pathophysiological processes targeted by emerging therapies for OA, along with relevant clinical data and discussion of the main challenges for the further development of these therapies, to provide context for the latest advances in the field of pharmaceutical therapies for OA.
Collapse
|
523
|
Al-Naggar IMA, Kuchel GA, Xu M. Senolytics: targeting senescent cells for age-associated diseases. ACTA ACUST UNITED AC 2020; 6:161-172. [PMID: 33777657 DOI: 10.1007/s40610-020-00140-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
| | - George A Kuchel
- UConn Center on Aging, UConn Health, Farmington, Connecticut, USA
| | - Ming Xu
- UConn Center on Aging, UConn Health, Farmington, Connecticut, USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
524
|
Zhu M, Meng P, Ling X, Zhou L. Advancements in therapeutic drugs targeting of senescence. Ther Adv Chronic Dis 2020; 11:2040622320964125. [PMID: 33133476 PMCID: PMC7576933 DOI: 10.1177/2040622320964125] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
Aging leads to a high burden on society, both medically and economically. Cellular senescence plays an essential role in the initiation of aging and age-related diseases. Recent studies have highlighted the therapeutic value of senescent cell deletion in natural aging and many age-related disorders. However, the therapeutic strategies for manipulating cellular senescence are still at an early stage of development. Among these strategies, therapeutic drugs that target cellular senescence are arguably the most highly anticipated. Many recent studies have demonstrated that a variety of drugs exhibit healthy aging effects. In this review, we summarize different types of drugs promoting healthy aging – such as senolytics, senescence-associated secretory phenotype (SASP) inhibitors, and nutrient signaling regulators – and provide an update on their potential therapeutic merits. Taken together, our review synthesizes recent advancements in the therapeutic potentialities of drugs promoting healthy aging with regard to their clinical implications.
Collapse
Affiliation(s)
- Mingsheng Zhu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Meng
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Xian Ling
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Ave, Guangzhou 510515, China
| |
Collapse
|
525
|
Abstract
PURPOSE OF REVIEW Senescent cells are now known to accumulate in multiple tissues with aging and through their inflammation (the senescence-associated secretory phenotype, SASP) contribute to aging and chronic diseases. Here, we review the roles of senescent osteocytes in the context of bone loss. RECENT FINDINGS Numerous studies have established that senescent osteocytes accumulate in the bone microenvironment with aging in mice and in humans. Moreover, at least in mice, elimination of senescent cells results in attenuation of age-related bone loss. Osteocyte senescence also occurs in response to other cellular stressors, including radiotherapy, chemotherapy, and metabolic dysfunction, where it appears to mediate skeletal deterioration. Osteocyte senescence is linked to bone loss associated with aging and other conditions. Senescent osteocytes are potential therapeutic targets to alleviate skeletal dysfunction. Additional studies better defining the underlying mechanisms as well as translating these exciting findings from mouse models to humans are needed.
Collapse
Affiliation(s)
- Joshua N Farr
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Division of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Japneet Kaur
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Madison L Doolittle
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sundeep Khosla
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
526
|
Hall BM, Gleiberman AS, Strom E, Krasnov PA, Frescas D, Vujcic S, Leontieva OV, Antoch MP, Kogan V, Koman IE, Zhu Y, Tchkonia T, Kirkland JL, Chernova OB, Gudkov AV. Immune checkpoint protein VSIG4 as a biomarker of aging in murine adipose tissue. Aging Cell 2020; 19:e13219. [PMID: 32856419 PMCID: PMC7576241 DOI: 10.1111/acel.13219] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 05/26/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue is recognized as a major source of systemic inflammation with age, driving age-related tissue dysfunction and pathogenesis. Macrophages (Mφ) are central to these changes yet adipose tissue Mφ (ATMs) from aged mice remain poorly characterized. To identify biomarkers underlying changes in aged adipose tissue, we performed an unbiased RNA-seq analysis of ATMs from young (8-week-old) and healthy aged (80-week-old) mice. One of the genes identified, V-set immunoglobulin-domain-containing 4 (VSIG4/CRIg), encodes a Mφ-associated complement receptor and B7 family-related immune checkpoint protein. Here, we demonstrate that Vsig4 expression is highly upregulated with age in perigonadal white adipose tissue (gWAT) in two mouse strains (inbred C57BL/6J and outbred NIH Swiss) independent of gender. The accumulation of VSIG4 was mainly attributed to a fourfold increase in the proportion of VSIG4+ ATMs (13%-52%). In a longitudinal study, VSIG4 expression in gWAT showed a strong correlation with age within a cohort of male and female mice and correlated strongly with physiological frailty index (PFI, a multi-parameter assessment of health) in male mice. Our results indicate that VSIG4 is a novel biomarker of aged murine ATMs. VSIG4 expression was also found to be elevated in other aging tissues (e.g., thymus) and was strongly induced in tumor-adjacent stroma in cases of spontaneous and xenograft lung cancer models. VSIG4 expression was recently associated with cancer and several inflammatory diseases with diagnostic and prognostic potential in both mice and humans. Further investigation is required to determine whether VSIG4-positive Mφ contribute to immunosenescence and/or systemic age-related deficits.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Olga V. Leontieva
- Department of Pharmacology and TherapeuticsRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Marina P. Antoch
- Department of Pharmacology and TherapeuticsRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Valeria Kogan
- Institute for Translational ResearchAriel UniversityArielIsrael
| | - Igor E. Koman
- Institute for Translational ResearchAriel UniversityArielIsrael
| | - Yi Zhu
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | | | | | - Andrei V. Gudkov
- Everon Biosciences IncBuffaloNYUSA
- Department of Cell Stress BiologyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
- Genome Protection IncBuffaloNYUSA
| |
Collapse
|
527
|
Food as medicine: targeting the uraemic phenotype in chronic kidney disease. Nat Rev Nephrol 2020; 17:153-171. [PMID: 32963366 DOI: 10.1038/s41581-020-00345-8] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2020] [Indexed: 02/07/2023]
Abstract
The observation that unhealthy diets (those that are low in whole grains, fruits and vegetables, and high in sugar, salt, saturated fat and ultra-processed foods) are a major risk factor for poor health outcomes has boosted interest in the concept of 'food as medicine'. This concept is especially relevant to metabolic diseases, such as chronic kidney disease (CKD), in which dietary approaches are already used to ameliorate metabolic and nutritional complications. Increased awareness that toxic uraemic metabolites originate not only from intermediary metabolism but also from gut microbial metabolism, which is directly influenced by diet, has fuelled interest in the potential of 'food as medicine' approaches in CKD beyond the current strategies of protein, sodium and phosphate restriction. Bioactive nutrients can alter the composition and metabolism of the microbiota, act as modulators of transcription factors involved in inflammation and oxidative stress, mitigate mitochondrial dysfunction, act as senolytics and impact the epigenome by altering one-carbon metabolism. As gut dysbiosis, inflammation, oxidative stress, mitochondrial dysfunction, premature ageing and epigenetic changes are common features of CKD, these findings suggest that tailored, healthy diets that include bioactive nutrients as part of the foodome could potentially be used to prevent and treat CKD and its complications.
Collapse
|
528
|
Carpenter VJ, Patel BB, Autorino R, Smith SC, Gewirtz DA, Saleh T. Senescence and castration resistance in prostate cancer: A review of experimental evidence and clinical implications. Biochim Biophys Acta Rev Cancer 2020; 1874:188424. [PMID: 32956765 DOI: 10.1016/j.bbcan.2020.188424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 01/10/2023]
Abstract
The development of Castration-Resistant Prostate Cancer (CRPC) remains a major challenge in the treatment of this disease. While Androgen Deprivation Therapy (ADT) can result in tumor shrinkage, a primary response of Prostate Cancer (PCa) cells to ADT is a senescent growth arrest. As a response to cancer therapies, senescence has often been considered as a beneficial outcome due to its association with stable growth abrogation, as well as the potential for immune system activation via the Senescence-Associated Secretory Phenotype (SASP). However, there is increasing evidence that not only can senescent cells regain proliferative capacity, but that senescence contributes to deleterious effects of cancer chemotherapy, including disease recurrence. Notably, the preponderance of work investigating the consequences of therapy-induced senescence on tumor progression has been performed in non-PCa models. Here, we summarize the evidence that ADT promotes a senescent response in PCa and postulate mechanisms by which senescence may contribute to the development of castration-resistance. Primarily, we suggest that ADT-induced senescence may support CRPC development via escape from senescence, by cell autonomous-reprogramming, and by the formation of a pro-tumorigenic SASP. However, due to the scarcity of direct evidence from PCa models, the consequences of ADT-induced senescence outlined here remain speculative until the relationship between senescence and CRPC can be experimentally defined.
Collapse
Affiliation(s)
- Valerie J Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Bhaumik B Patel
- Department of Internal Medicine, Division of Hematology, Oncology & Palliative Care, VCU Health, Richmond, VA, USA
| | - Riccardo Autorino
- Department of Surgery, Division of Urology, VCU Health, Richmond, VA, USA
| | | | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tareq Saleh
- The Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan.
| |
Collapse
|
529
|
Garg S, Khan SI, Malhotra RK, Sharma MK, Kumar M, Kaur P, Nag TC, RumaRay, Bhatia J, Arya DS. The molecular mechanism involved in cardioprotection by the dietary flavonoid fisetin as an agonist of PPAR-γ in a murine model of myocardial infarction. Arch Biochem Biophys 2020; 694:108572. [PMID: 32926843 DOI: 10.1016/j.abb.2020.108572] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023]
Abstract
The methodology exploring the cardioprotective potential of the flavonoid Fisetin through its ability to modulate PPAR-γ was unraveled in the present study. Computational modelling through molecular docking based binding study of interactions between Fiestin and PPAR-γ revealed the potential role of Fisetin as an agonist of PPAR-γ. A murine model of cardiac ischemia-reperfusion injury was used to explore this further. Male Wistar Rats were randomly assigned to five groups. Fisetin (20 mg/kg; p. o) was administered for 28 days. Ischemia was induced for 45 min on the 29th day followed by 60 min of reperfusion. Fisetin pretreatment upregulated the expression of PPAR-γ in heart tissue significantly Cardioprotection was assessed by measurement of hemodynamic parameters, infarct size, ELISA for oxidative stress, immunohistochemistry and TUNEL assay for apoptosis, and western blot analysis for MAPK proteins and inflammation. PPAR-γ activation by fisetin led to significantly reduced infarct size, suppression of oxidative stress, reduction of cardiac injury markers, alleviation of inflammation, and inhibition of apoptosis The MAPK-based molecular mechanism showed a rise in a key prosurvival kinase, ERK1/ERK2 and suppression of JNK and p38 proteins. The aforementioned beneficial findings of fisetin were reversed on the administration of a specific antagonist of PPAR-γ. In conclusion, through our experiments, we have proved that fisetin protects the heart against ischemia-reperfusion injury and the evident cardioprotection is PPAR-γ dependant. In conclusion, our study has revealed a prime mechanism involved in the cardioprotective effects of fisetin. Hence, Fisetin may be evaluated in further clinical studies as a cardioprotective agent in patients undergoing reperfusion interventions.
Collapse
Affiliation(s)
- Shanky Garg
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Sana Irfan Khan
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Rajiv Kumar Malhotra
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Manish Kumar Sharma
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Manoj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - RumaRay
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Jagriti Bhatia
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
530
|
Oladele JO, Ajayi EI, Oyeleke OM, Oladele OT, Olowookere BD, Adeniyi BM, Oyewole OI, Oladiji AT. A systematic review on COVID-19 pandemic with special emphasis on curative potentials of Nigeria based medicinal plants. Heliyon 2020; 6:e04897. [PMID: 32929412 PMCID: PMC7480258 DOI: 10.1016/j.heliyon.2020.e04897] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/16/2020] [Accepted: 09/07/2020] [Indexed: 01/08/2023] Open
Abstract
Despite the frightening mortality rate associated with COVID-19, there is no known approved drug to effectively combat the pandemic. COVID-19 clinical manifestations include fever, fatigue, cough, shortness of breath, and other complications. At present, there is no known effective treatment or vaccine that can mitigate/inhibit SARS-CoV-2. Available clinical intervention for COVID-19 is only palliative and limited to support. Thus, there is an exigent need for effective and non-invasive treatment. This article evaluates the possible mechanism of actions of SARS-CoV-2 and present Nigeria based medicinal plants which have pharmacological and biological activities that can mitigate the hallmarks of the pathogenesis of COVID-19. SARS-CoV-2 mode of actions includes hyper-inflammation characterized by a severe and fatal hyper-cytokinaemia with multi-organ failure; immunosuppression; reduction of angiotensin-converting enzyme 2 (ACE2) to enhance pulmonary vascular permeability causing damage to the alveoli; and further activated by open reading frame (ORF)3a, ORF3b, and ORF7a via c-Jun N- terminal kinase (JNK) pathway which induces lung damage. These mechanisms of action of SARS-CoV-2 can be mitigated by a combination therapy of medicinal herbs based on their pharmacological activities. Since the clinical manifestations of COVID-19 are multifactorial with co-morbidities, we strongly recommend the use of combined therapy such that two or more herbs with specific therapeutic actions are administered to combat the mediators of the disease.
Collapse
Affiliation(s)
- Johnson O. Oladele
- Biochemistry Unit, Department of Chemical Sciences, Kings University, Ode-Omu, Osun State, Nigeria
| | - Ebenezer I. Ajayi
- Membrane Biophysics and Nanotechnology Laboratories, Mercedes and Martin Ferreyra Institute of Medicine, IMMF-INIMEC-CONICET-UNC, Cordoba, Argentina
- Diabesity Complications & Other Neglected Infectious Diseases Group, Department of Biochemistry, Osun State University, Osogbo, Nigeria
| | - Oyedotun M. Oyeleke
- Biochemistry Unit, Department of Chemical Sciences, Kings University, Ode-Omu, Osun State, Nigeria
| | - Oluwaseun T. Oladele
- Phytomedicine and Molecular Toxicology Research Laboratories, Department of Biochemistry, Osun State University, Osogbo, Nigeria
| | - Boyede D. Olowookere
- Biochemistry Unit, Department of Chemical Sciences, Kings University, Ode-Omu, Osun State, Nigeria
| | - Boluwaji M. Adeniyi
- Centre of Excellence for Food Technology and Research -Benue State University, Nigerian Stored Products Research Institute, Ibadan, Nigeria
| | - Olu I. Oyewole
- Phytomedicine and Molecular Toxicology Research Laboratories, Department of Biochemistry, Osun State University, Osogbo, Nigeria
| | | |
Collapse
|
531
|
Cherif H, Bisson DG, Mannarino M, Rabau O, Ouellet JA, Haglund L. Senotherapeutic drugs for human intervertebral disc degeneration and low back pain. eLife 2020; 9:54693. [PMID: 32821059 PMCID: PMC7442487 DOI: 10.7554/elife.54693] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence is a contributor to intervertebral disc (IVD) degeneration and low back pain. Here, we found that RG-7112, a potent mouse double-minute two protein inhibitor, selectively kills senescent IVD cells through apoptosis. Gene expression pathway analysis was used to compare the functional networks of genes affected by RG-7112, a pure synthetic senolytic with o-Vanillin a natural and anti-inflammatory senolytic. Both affected a functional gene network related to cell death and survival. O-Vanillin also affected networks related to cell cycle progression as well as connective tissue development and function. Both senolytics effectively decreased the senescence-associated secretory phenotype (SASP) of IVD cells. Furthermore, bioavailability and efficacy were verified ex vivo in the physiological environment of degenerating intact human discs where a single dose improved disc matrix homeostasis. Matrix improvement correlated with a reduction in senescent cells and SASP, supporting a translational potential of targeting senescent cells as a therapeutic intervention. Pain in the lower back affects about four in five people during their lifetime. Over time, the discs that provide cushioning between the vertebrae of the spine can degenerate, which can be one of the major causes of lower back pain. It has been shown that when the cells of these discs are exposed to different stress factors, they stop growing and become irreversibly dormant. Such ‘senescent’ cells release a range of proteins and small molecules that lead to painful inflammation and further degeneration of the discs. Moreover, it is thought that a high number of senescent cells may be linked to other degenerative diseases such as arthritis. Current treatments can only reduce the severity of the symptoms, but they cannot prevent the degeneration from progressing. Now, Cherif et al. set out to test the effects of two different compounds on human disc cells grown in the laboratory. One of the molecules studied, RG-7112, is a synthetic drug that has been approved for safety by the US Food and Drug Administration and has been shown to remove senescent cells. The other, o-Vanillin, is a natural compound that has anti-inflammatory and anti-senescence properties. The results showed that both compounds were able to trigger changes to that helped new, healthy cells to grow and at the same time kill senescent cells. They also reduced the production of molecules linked to inflammation and pain. Further analyses revealed that the compounds were able to strengthen the fibrous matrix that surrounds and supports the discs. Cherif et al. hope that this could form the basis for a new family of drugs for back pain to slow the degeneration of the discs and reduce pain. This may also have benefits for other similar degenerative diseases caused by cell senescence, such as arthritis.
Collapse
Affiliation(s)
- Hosni Cherif
- Orthopaedic Research Lab, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Daniel G Bisson
- Orthopaedic Research Lab, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Matthew Mannarino
- Orthopaedic Research Lab, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Oded Rabau
- McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,Shriner's Hospital for Children, 1003 Decarie Blvd, Montreal, Canada
| | - Jean A Ouellet
- McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,Shriner's Hospital for Children, 1003 Decarie Blvd, Montreal, Canada
| | - Lisbet Haglund
- Orthopaedic Research Lab, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,Shriner's Hospital for Children, 1003 Decarie Blvd, Montreal, Canada
| |
Collapse
|
532
|
Borghesan M, Hoogaars WMH, Varela-Eirin M, Talma N, Demaria M. A Senescence-Centric View of Aging: Implications for Longevity and Disease. Trends Cell Biol 2020; 30:777-791. [PMID: 32800659 DOI: 10.1016/j.tcb.2020.07.002] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/03/2020] [Accepted: 07/10/2020] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a state of stable cell cycle arrest associated with macromolecular alterations and secretion of proinflammatory cytokines and molecules. From their initial discovery in the 1960s, senescent cells have been hypothesized as potential contributors to the age-associated loss of regenerative potential. Here, we discuss recent evidence that implicates cellular senescence as a central regulatory mechanism of the aging process. We provide a comprehensive overview of age-associated pathologies in which cellular senescence has been implicated. We describe mechanisms by which senescent cells drive aging and diseases, and we discuss updates on exploiting these mechanisms as therapeutic targets. Finally, we critically analyze the use of senotherapeutics and their translation to the clinic, highlighting limitations and suggesting ideas for future applications and developments.
Collapse
Affiliation(s)
- M Borghesan
- European Research Institute for the Biology of Ageing (ERIBA);, University Medical Center Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9715RA, Groningen, The Netherlands
| | - W M H Hoogaars
- European Research Institute for the Biology of Ageing (ERIBA);, University Medical Center Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9715RA, Groningen, The Netherlands
| | - M Varela-Eirin
- European Research Institute for the Biology of Ageing (ERIBA);, University Medical Center Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9715RA, Groningen, The Netherlands
| | - N Talma
- European Research Institute for the Biology of Ageing (ERIBA);, University Medical Center Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9715RA, Groningen, The Netherlands
| | - M Demaria
- European Research Institute for the Biology of Ageing (ERIBA);, University Medical Center Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9715RA, Groningen, The Netherlands.
| |
Collapse
|
533
|
Tobin SW, Alibhai FJ, Weisel RD, Li RK. Considering Cause and Effect of Immune Cell Aging on Cardiac Repair after Myocardial Infarction. Cells 2020; 9:E1894. [PMID: 32823583 PMCID: PMC7465938 DOI: 10.3390/cells9081894] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
The importance of the immune system for cardiac repair following myocardial infarction is undeniable; however, the complex nature of immune cell behavior has limited the ability to develop effective therapeutics. This limitation highlights the need for a better understanding of the function of each immune cell population during the inflammatory and resolution phases of cardiac repair. The development of reliable therapies is further complicated by aging, which is associated with a decline in cell and organ function and the onset of cardiovascular and immunological diseases. Aging of the immune system has important consequences on heart function as both chronic cardiac inflammation and an impaired immune response to cardiac injury are observed in older individuals. Several studies have suggested that rejuvenating the aged immune system may be a valid therapeutic candidate to prevent or treat heart disease. Here, we review the basic patterns of immune cell behavior after myocardial infarction and discuss the autonomous and nonautonomous manners of hematopoietic stem cell and immune cell aging. Lastly, we identify prospective therapies that may rejuvenate the aged immune system to improve heart function such as anti-inflammatory and senolytic therapies, bone marrow transplant, niche remodeling and regulation of immune cell differentiation.
Collapse
Affiliation(s)
- Stephanie W. Tobin
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
| | - Faisal J. Alibhai
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
| | - Richard D. Weisel
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
- Division of Cardiac Surgery, Peter Munk Cardiac Centre, Toronto General Hospital and University of Toronto, Toronto, ON M5G 2N2, Canada
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
- Division of Cardiac Surgery, Peter Munk Cardiac Centre, Toronto General Hospital and University of Toronto, Toronto, ON M5G 2N2, Canada
| |
Collapse
|
534
|
Wilkinson HN, Hardman MJ. Senescence in Wound Repair: Emerging Strategies to Target Chronic Healing Wounds. Front Cell Dev Biol 2020; 8:773. [PMID: 32850866 PMCID: PMC7431694 DOI: 10.3389/fcell.2020.00773] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/22/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a fundamental stress response that restrains tumour formation. Yet, senescence cells are also present in non-cancerous states, accumulating exponentially with chronological age and contributing to age- and diabetes-related cellular dysfunction. The identification of hypersecretory and phagocytic behaviours in cells that were once believed to be non-functional has led to a recent explosion of senescence research. Here we discuss the profound, and often opposing, roles identified for short-lived vs. chronic tissue senescence. Transiently induced senescence is required for development, regeneration and acute wound repair, while chronic senescence is widely implicated in tissue pathology. We recently demonstrated that sustained senescence contributes to impaired diabetic healing via the CXCR2 receptor, which when blocked promotes repair. Further studies have highlighted the beneficial effects of targeting a range of senescence-linked processes to fight disease. Collectively, these findings hold promise for developing clinically viable strategies to tackle senescence in chronic wounds and other cutaneous pathologies.
Collapse
Affiliation(s)
- Holly N Wilkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Matthew J Hardman
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, United Kingdom
| |
Collapse
|
535
|
Wissler Gerdes EO, Zhu Y, Weigand BM, Tripathi U, Burns TC, Tchkonia T, Kirkland JL. Cellular senescence in aging and age-related diseases: Implications for neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:203-234. [PMID: 32854855 PMCID: PMC7656525 DOI: 10.1016/bs.irn.2020.03.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging is the major predictor for developing multiple neurodegenerative diseases, including Alzheimer's disease (AD) other dementias, and Parkinson's disease (PD). Senescent cells, which can drive aging phenotypes, accumulate at etiological sites of many age-related chronic diseases. These cells are resistant to apoptosis and can cause local and systemic dysfunction. Decreasing senescent cell abundance using senolytic drugs, agents that selectively target these cells, alleviates neurodegenerative diseases in preclinical models. In this review, we consider roles of senescent cells in neurodegenerative diseases and potential implications of senolytic agents as an innovative treatment.
Collapse
Affiliation(s)
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - B Melanie Weigand
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Utkarsh Tripathi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Terence C Burns
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
536
|
Ermogenous C, Green C, Jackson T, Ferguson M, Lord JM. Treating age-related multimorbidity: the drug discovery challenge. Drug Discov Today 2020; 25:1403-1415. [DOI: 10.1016/j.drudis.2020.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/19/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022]
|
537
|
Wyld L, Bellantuono I, Tchkonia T, Morgan J, Turner O, Foss F, George J, Danson S, Kirkland JL. Senescence and Cancer: A Review of Clinical Implications of Senescence and Senotherapies. Cancers (Basel) 2020; 12:cancers12082134. [PMID: 32752135 PMCID: PMC7464619 DOI: 10.3390/cancers12082134] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
Cellular senescence is a key component of human aging that can be induced by a range of stimuli, including DNA damage, cellular stress, telomere shortening, and the activation of oncogenes. Senescence is generally regarded as a tumour suppressive process, both by preventing cancer cell proliferation and suppressing malignant progression from pre-malignant to malignant disease. It may also be a key effector mechanism of many types of anticancer therapies, such as chemotherapy, radiotherapy, and endocrine therapies, both directly and via bioactive molecules released by senescent cells that may stimulate an immune response. However, senescence may contribute to reduced patient resilience to cancer therapies and may provide a pathway for disease recurrence after cancer therapy. A new group of drugs, senotherapies, (drugs which interact with senescent cells to interfere with their pro-aging impacts by either selectively destroying senescent cells (senolytic drugs) or inhibiting their function (senostatic drugs)) are under active investigation to determine whether they can enhance the efficacy of cancer therapies and improve resilience to cancer treatments. Senolytic drugs include quercetin, navitoclax, and fisetin and preclinical and early phase clinical data are emerging of their potential role in cancer treatments, although none are yet in routine use clinically. This article provides a review of these issues.
Collapse
Affiliation(s)
- Lynda Wyld
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
- Correspondence:
| | - Ilaria Bellantuono
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA;
| | - Jenna Morgan
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - Olivia Turner
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - Fiona Foss
- Department of Pathology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield S10 2JF, UK;
| | - Jayan George
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - Sarah Danson
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - James L. Kirkland
- Departments of Internal Medicine, Geriatric Medicine and Gerontology, The Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
538
|
Currais A, Huang L, Petrascheck M, Maher P, Schubert D. A chemical biology approach to identifying molecular pathways associated with aging. GeroScience 2020; 43:353-365. [PMID: 32705410 DOI: 10.1007/s11357-020-00238-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/19/2020] [Indexed: 12/26/2022] Open
Abstract
The understanding of how aging contributes to dementia remains obscure. To address this problem, a chemical biology approach was used employing CAD031, an Alzheimer's disease (AD) drug candidate identified using a discovery platform based upon phenotypic screens that mimic toxicities associated with the aging brain. Since CAD031 has therapeutic efficacy when fed to old symptomatic transgenic AD mice, the chemical biology hypothesis is that it can be used to determine the molecular pathways associated with age-related disease by identifying those that are modified by the compound. Here we show that when CAD031 was fed to rapidly aging SAMP8 mice starting in the last quadrant of their lifespan, it reduced many of the changes in gene, protein, and small molecule expression associated with mitochondrial aging, maintaining mitochondria at the younger molecular phenotype. Network analysis integrating the metabolomics and transcription data followed by mechanistic validation showed that CAD031 targets acetyl-CoA and fatty acid metabolism via the AMPK/ACC1 pathway. Importantly, CAD031 extended the median lifespan of SAMP8 mice by about 30%. These data show that specific alterations in mitochondrial composition and metabolism highly correlate with aging, supporting the use AD drug candidates that limit physiological aging in the brain.
Collapse
Affiliation(s)
- Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA.
| | - Ling Huang
- The Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Michael Petrascheck
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - David Schubert
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA.
| |
Collapse
|
539
|
Karaman Mayack B, Sippl W, Ntie-Kang F. Natural Products as Modulators of Sirtuins. Molecules 2020; 25:molecules25143287. [PMID: 32698385 PMCID: PMC7397027 DOI: 10.3390/molecules25143287] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 07/12/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
Natural products have been used for the treatment of human diseases since ancient history. Over time, due to the lack of precise tools and techniques for the separation, purification, and structural elucidation of active constituents in natural resources there has been a decline in financial support and efforts in characterization of natural products. Advances in the design of chemical compounds and the understanding of their functions is of pharmacological importance for the biomedical field. However, natural products regained attention as sources of novel drug candidates upon recent developments and progress in technology. Natural compounds were shown to bear an inherent ability to bind to biomacromolecules and cover an unparalleled chemical space in comparison to most libraries used for high-throughput screening. Thus, natural products hold a great potential for the drug discovery of new scaffolds for therapeutic targets such as sirtuins. Sirtuins are Class III histone deacetylases that have been linked to many diseases such as Parkinson`s disease, Alzheimer’s disease, type II diabetes, and cancer linked to aging. In this review, we examine the revitalization of interest in natural products for drug discovery and discuss natural product modulators of sirtuins that could serve as a starting point for the development of isoform selective and highly potent drug-like compounds, as well as the potential application of naturally occurring sirtuin inhibitors in human health and those in clinical trials.
Collapse
Affiliation(s)
- Berin Karaman Mayack
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Istanbul University, Istanbul 34116, Turkey
- Correspondence:
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany; (W.S.); (F.N.-K.)
| | - Fidele Ntie-Kang
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany; (W.S.); (F.N.-K.)
- Department of Chemistry, University of Buea, P.O. Box 63, Buea CM-00237, Cameroon
- Institute of Botany, Technical University of Dresden, 01217 Dresden, Germany
| |
Collapse
|
540
|
The role of adipose tissue senescence in obesity- and ageing-related metabolic disorders. Clin Sci (Lond) 2020; 134:315-330. [PMID: 31998947 DOI: 10.1042/cs20190966] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 12/19/2022]
Abstract
Adipose tissue as the largest energy reservoir and endocrine organ is essential for maintenance of systemic glucose, lipid and energy homeostasis, but these metabolic functions decline with ageing and obesity. Adipose tissue senescence is one of the common features in obesity and ageing. Although cellular senescence is a defensive mechanism preventing tumorigenesis, its occurrence in adipose tissue causatively induces defective adipogenesis, inflammation, aberrant adipocytokines production and insulin resistance, leading to adipose tissue dysfunction. In addition to these paracrine effects, adipose tissue senescence also triggers systemic inflammation and senescence as well as insulin resistance in the distal metabolic organs, resulting in Type 2 diabetes and other premature physiological declines. Multiple cell types including mature adipocytes, immune cells, endothelial cells and progenitor cells gradually senesce at different levels in different fat depots with ageing and obesity, highlighting the heterogeneity and complexity of adipose tissue senescence. In this review, we discuss the causes and consequences of adipose tissue senescence, and the major cell types responsible for adipose tissue senescence in ageing and obesity. In addition, we summarize the pharmacological approaches and lifestyle intervention targeting adipose tissue senescence for the treatment of obesity- and ageing-related metabolic diseases.
Collapse
|
541
|
Abstract
For centuries, people believed that bats possessed sinister powers. Bats are thought to be ancestral hosts to many deadly viruses affecting humans including Ebola, rabies, and most recently SARS-CoV-2 coronavirus. However, bats themselves tolerate these viruses without ill effects. The second power that bats have is their longevity. Bats live much longer than similar-sized land mammals. Here we review how bats' ability to control inflammation may be contributing to their longevity. The underlying mechanisms may hold clues to developing new treatments for age-related diseases. Now may be the time to use science to exploit the secret powers of bats for human benefit.
Collapse
Affiliation(s)
- Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA.
| | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA
| | - Brian K Kennedy
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore; Centre for Healthy Longevity, National University Health System, Singapore 117609, Singapore; Singapore Institute of Clinical Sciences, A(∗)STAR, Singapore 117609, Singapore.
| |
Collapse
|
542
|
Grosse L, Wagner N, Emelyanov A, Molina C, Lacas-Gervais S, Wagner KD, Bulavin DV. Defined p16 High Senescent Cell Types Are Indispensable for Mouse Healthspan. Cell Metab 2020; 32:87-99.e6. [PMID: 32485135 DOI: 10.1016/j.cmet.2020.05.002] [Citation(s) in RCA: 295] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/13/2020] [Accepted: 05/04/2020] [Indexed: 12/29/2022]
Abstract
The accumulation of senescent cells can drive many age-associated phenotypes and pathologies. Consequently, it has been proposed that removing senescent cells might extend lifespan. Here, we generated two knockin mouse models targeting the best-characterized marker of senescence, p16Ink4a. Using a genetic lineage tracing approach, we found that age-induced p16High senescence is a slow process that manifests around 10-12 months of age. The majority of p16High cells were vascular endothelial cells mostly in liver sinusoids (LSECs), and to lesser extent macrophages and adipocytes. In turn, continuous or acute elimination of p16High senescent cells disrupted blood-tissue barriers with subsequent liver and perivascular tissue fibrosis and health deterioration. Our data show that senescent LSECs are not replaced after removal and have important structural and functional roles in the aging organism. In turn, delaying senescence or replacement of senescent LSECs could represent a powerful tool in slowing down aging.
Collapse
Affiliation(s)
- Laurent Grosse
- Institute for Research on Cancer and Aging of Nice (IRCAN), INSERM, Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS), Nice, France
| | - Nicole Wagner
- Université Côte d'Azur, INSERM, Centre National de la Recherche Scientifique (CNRS), Institute of Biology Valrose, Nice, France
| | - Alexander Emelyanov
- Institute for Research on Cancer and Aging of Nice (IRCAN), INSERM, Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS), Nice, France
| | - Clement Molina
- Institute for Research on Cancer and Aging of Nice (IRCAN), INSERM, Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS), Nice, France
| | - Sandra Lacas-Gervais
- Electron Microscopy Facility, Centre Commun de Microscopie Appliquée, CCMA, Université Côte d'Azur, UCA, Nice, France
| | - Kay-Dietrich Wagner
- Université Côte d'Azur, INSERM, Centre National de la Recherche Scientifique (CNRS), Institute of Biology Valrose, Nice, France
| | - Dmitry V Bulavin
- Institute for Research on Cancer and Aging of Nice (IRCAN), INSERM, Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS), Nice, France.
| |
Collapse
|
543
|
Kunihiro AG, Luis PB, Frye JB, Chew W, Chow HHS, Schneider C, Funk JL. Bone-Specific Metabolism of Dietary Polyphenols in Resorptive Bone Diseases. Mol Nutr Food Res 2020; 64:e2000072. [PMID: 32506808 PMCID: PMC7712627 DOI: 10.1002/mnfr.202000072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/27/2020] [Indexed: 12/14/2022]
Abstract
SCOPE Curcumin prevents bone loss in resorptive bone diseases and inhibits osteoclast formation, a key process driving bone loss. Curcumin circulates as an inactive glucuronide that can be deconjugated in situ by bone's high β-glucuronidase (GUSB) content, forming the active aglycone. Because curcumin is a common remedy for musculoskeletal disease, effects of microenvironmental changes consequent to skeletal development or disease on bone curcumin metabolism are explored. METHODS AND RESULTS Across sexual/skeletal development or between sexes in C57BL/6 mice ingesting curcumin (500 mg kg-1 ), bone curcumin metabolism and GUSB enzyme activity are unchanged, except for >twofold higher (p < 0.05) bone curcumin-glucuronide substrate levels in immature (4-6-week-old) mice. In ovariectomized (OVX) or bone metastasis-bearing female mice, bone substrate levels are also >twofold higher. Aglycone curcumin levels tend to increase proportional to substrate such that the majority of glucuronide distributing to bone is deconjugated, including OVX mice where GUSB decreases by 24% (p < 0.01). GUSB also catalyzes deconjugation of resveratrol and quercetin glucuronides by bone, and a requirement for the aglycones for anti-osteoclastogenic bioactivity, analogous to curcumin, is confirmed. CONCLUSION Dietary polyphenols circulating as glucuronides may require in situ deconjugation for bone-protective effects, a process influenced by bone microenvironmental changes.
Collapse
Affiliation(s)
- Andrew G Kunihiro
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ
| | - Paula B Luis
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| | | | - Wade Chew
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - H-H. Sherry Chow
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Claus Schneider
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| | - Janet L Funk
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ
- Department of Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
544
|
Song S, Lam EWF, Tchkonia T, Kirkland JL, Sun Y. Senescent Cells: Emerging Targets for Human Aging and Age-Related Diseases. Trends Biochem Sci 2020; 45:578-592. [PMID: 32531228 PMCID: PMC7649645 DOI: 10.1016/j.tibs.2020.03.008] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/03/2020] [Accepted: 03/13/2020] [Indexed: 01/10/2023]
Abstract
Aging is a major risk factor for numerous human pathologies, including cardiovascular, metabolic, musculoskeletal, and neurodegenerative conditions and various malignancies. While our understanding of aging is far from complete, recent advances suggest that targeting fundamental aging processes can delay, prevent, or alleviate age-related disorders. Cellular senescence is physiologically beneficial in several contexts, but it has causal roles in multiple chronic diseases. New studies have illustrated the promising feasibility and safety to selectively ablate senescent cells from tissues, a therapeutic modality that holds potential for treating multiple chronic pathologies and extending human healthspan. Here, we review molecular links between cellular senescence and age-associated complications and highlight novel therapeutic avenues that may be exploited to target senescent cells in future geriatric medicine.
Collapse
Affiliation(s)
- Shuling Song
- School of Gerontology, Binzhou Medical University, Yantai, Shandong 264003, China; Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, W12 0NN, UK
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Yu Sun
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; School of Pharmacy, Binzhou Medical University, Yantai, 264003, China; Department of Medicine and VAPSHCS, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
545
|
Maher PA. Using Plants as a Source of Potential Therapeutics for the Treatment of Alzheimer's Disease. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:365-373. [PMID: 32607095 PMCID: PMC7309672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with the numbers expected to increase dramatically as our society ages. There are no treatments to cure, prevent, or slow down the progression of the disease. Age is the single greatest risk factor for AD. However, to date, AD drug discovery efforts have generally not taken this fact into consideration. Multiple changes associated with brain aging, including neuroinflammation and oxidative stress, are important contributors to disease development and progression. Thus, due to the multifactorial nature of AD, the one target strategy to fight the disease needs to be replaced by a more general approach using pleiotropic compounds to deal with the complexity of the disease. In this perspectives piece, our alternative approach to AD drug development based on the biology of aging is described. Starting with plants or plant-derived natural products, we have used a battery of cell-based screening assays that reflect multiple, age-associated toxicity pathways to identify compounds that can target the aspects of aging that contribute to AD pathology. We have found that this combination of assays provides a replicable, cost- and time-effective screening approach that has to date yielded one compound in clinical trials for AD (NCT03838185) and several others that show significant promise.
Collapse
Affiliation(s)
- Pamela A. Maher
- To whom all correspondence should be addressed: Pamela A. Maher, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037; Tel: 858-453-4100 x1932;
| |
Collapse
|
546
|
Das J, Singh R, Ladol S, Nayak SK, Sharma D. Fisetin prevents the aging-associated decline in relative spectral power of α, β and linked MUA in the cortex and behavioral alterations. Exp Gerontol 2020; 138:111006. [PMID: 32592831 DOI: 10.1016/j.exger.2020.111006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022]
Abstract
Mental health in old age is of great concern due to the increased incidence of neurological and psychiatric diseases. With age, probability of cognitive and behavioral deficits increase and the prognosis deteriorates. Although a few in vitro studies have reported that flavonoid fisetin is beneficial for healthy aging, its effect on deteriorating mental health with aging in vivo is very limited and poorly understood. The brain aging is physiologically characterized by electroencephalograph (EEG) wave frequency, power, and distribution. Brain oscillatory waves from neural tissue get altered by various sensory-cognitive inputs. Besides, the fast-wave α(8-12 Hz)- and β(12-28 Hz)-oscillations are associated with coordination and indeed deal with complex behavioral performances. Therefore, the effect of fisetin supplementation on age-associated EEG mean cortical spectral power in α- and β-oscillations, multi-unit activity (MUA) count were studied in vivo which was not addressed so far. Besides, age-associated cognitive and behavioral alterations were also studied. The relative spectral power of α and β declined along with the MUA count in aged rats compared to young. However, supplementing fisetin for four weeks has improved relative α-power, β-power, and MUA count in aged rats. Also, fisetin supplemented aged rats showed significantly improved cognitive and behavioral performances than aged controls. These findings demonstrated the relative cortical spectral power in α-, β-oscillations, and MUA count change in aged rats and that some of these changes and behavioral alterations may be partly as a result of oxidative stress, which was prevented significantly in fisetin supplemented aged rats. Thus, fisetin boosted mental health in the aged animals.
Collapse
Affiliation(s)
- Jharana Das
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Rameshwar Singh
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Stanzin Ladol
- Department of Zoology, Central University of Jammu, Jammu and Kashmir 181143, India.
| | - Sasmita Kumari Nayak
- Department of Instrumentation and Electronics, College of Engineering and Technology, Bhubaneswar 751003, India
| | - Deepak Sharma
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
547
|
Martel J, Ojcius DM, Wu CY, Peng HH, Voisin L, Perfettini JL, Ko YF, Young JD. Emerging use of senolytics and senomorphics against aging and chronic diseases. Med Res Rev 2020; 40:2114-2131. [PMID: 32578904 DOI: 10.1002/med.21702] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 06/04/2020] [Accepted: 06/12/2020] [Indexed: 12/20/2022]
Abstract
Senescence is a state of cell cycle arrest that plays an important role in embryogenesis, wound healing and protection against cancer. Senescent cells also accumulate during aging and contribute to the development of age-related disorders and chronic diseases, such as atherosclerosis, type 2 diabetes, osteoarthritis, idiopathic pulmonary fibrosis, and liver disease. Molecules that induce apoptosis of senescent cells, such as dasatinib, quercetin, and fisetin, produce health benefits and extend lifespan in animal models. We describe here the mechanism of action of senolytics and senomorphics, many of which are derived from plants and fungi. We also discuss the possibility of using such compounds to delay aging and treat chronic diseases in humans.
Collapse
Affiliation(s)
- Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, California
| | - Cheng-Yeu Wu
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Research Center of Bacterial Pathogenesis, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Hsin-Hsin Peng
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Laboratory Animal Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China
| | - Laurent Voisin
- Institut Gustave Roussy, INSERM U1030, Université Paris-Sud, Villejuif, France
| | - Jean-Luc Perfettini
- Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, California.,Institut Gustave Roussy, INSERM U1030, Université Paris-Sud, Villejuif, France
| | - Yun-Fei Ko
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Chang Gung Biotechnology Corporation, Taipei, Taiwan, Republic of China.,Biochemical Engineering Research Center, Ming Chi University of Technology, New Taipei City, Taiwan, Republic of China
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Chang Gung Biotechnology Corporation, Taipei, Taiwan, Republic of China.,Biochemical Engineering Research Center, Ming Chi University of Technology, New Taipei City, Taiwan, Republic of China
| |
Collapse
|
548
|
Proshkina E, Shaposhnikov M, Moskalev A. Genome-Protecting Compounds as Potential Geroprotectors. Int J Mol Sci 2020; 21:E4484. [PMID: 32599754 PMCID: PMC7350017 DOI: 10.3390/ijms21124484] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
Throughout life, organisms are exposed to various exogenous and endogenous factors that cause DNA damages and somatic mutations provoking genomic instability. At a young age, compensatory mechanisms of genome protection are activated to prevent phenotypic and functional changes. However, the increasing stress and age-related deterioration in the functioning of these mechanisms result in damage accumulation, overcoming the functional threshold. This leads to aging and the development of age-related diseases. There are several ways to counteract these changes: 1) prevention of DNA damage through stimulation of antioxidant and detoxification systems, as well as transition metal chelation; 2) regulation of DNA methylation, chromatin structure, non-coding RNA activity and prevention of nuclear architecture alterations; 3) improving DNA damage response and repair; 4) selective removal of damaged non-functional and senescent cells. In the article, we have reviewed data about the effects of various trace elements, vitamins, polyphenols, terpenes, and other phytochemicals, as well as a number of synthetic pharmacological substances in these ways. Most of the compounds demonstrate the geroprotective potential and increase the lifespan in model organisms. However, their genome-protecting effects are non-selective and often are conditioned by hormesis. Consequently, the development of selective drugs targeting genome protection is an advanced direction.
Collapse
Affiliation(s)
- Ekaterina Proshkina
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (M.S.)
| | - Mikhail Shaposhnikov
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (M.S.)
| | - Alexey Moskalev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (M.S.)
- Pitirim Sorokin Syktyvkar State University, 55 Oktyabrsky prosp., 167001 Syktyvkar, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
549
|
Battram AM, Bachiller M, Martín-Antonio B. Senescence in the Development and Response to Cancer with Immunotherapy: A Double-Edged Sword. Int J Mol Sci 2020; 21:ijms21124346. [PMID: 32570952 PMCID: PMC7352478 DOI: 10.3390/ijms21124346] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence was first described as a physiological tumor cell suppressor mechanism that leads to cell growth arrest with production of the senescence-associated secretory phenotype known as SASP. The main role of SASP in physiological conditions is to attract immune cells to clear senescent cells avoiding tumor development. However, senescence can be damage-associated and, depending on the nature of these stimuli, additional types of senescence have been described. In the context of cancer, damage-associated senescence has been described as a consequence of chemotherapy treatments that were initially thought of as a tumor suppressor mechanism. However, in certain contexts, senescence after chemotherapy can promote cancer progression, especially when immune cells become senescent and cannot clear senescent tumor cells. Moreover, aging itself leads to continuous inflammaging and immunosenescence which are responsible for rewiring immune cells to become defective in their functionality. Here, we define different types of senescence, pathways that activate them, and functions of SASP in these events. Additionally, we describe the role of senescence in cancer and its treatments, including how aging and chemotherapy contribute to senescence in tumor cells, before focusing on immune cell senescence and its role in cancer. Finally, we discuss potential therapeutic interventions to reverse cell senescence.
Collapse
Affiliation(s)
- Anthony M. Battram
- Department of Hematology, Hospital Clinic, IDIBAPS, 08036 Barcelona, Spain; (A.M.B.); (M.B.)
| | - Mireia Bachiller
- Department of Hematology, Hospital Clinic, IDIBAPS, 08036 Barcelona, Spain; (A.M.B.); (M.B.)
| | - Beatriz Martín-Antonio
- Department of Hematology, Hospital Clinic, IDIBAPS, 08036 Barcelona, Spain; (A.M.B.); (M.B.)
- Department of Hematology, Hospital Clinic, IDIBAPS/Josep Carreras Leukaemia Research Institute, Carrer Rosselló 149-153, 08036 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-227-45-28; Fax: +34-93-312-94-07
| |
Collapse
|
550
|
Abstract
PURPOSE OF REVIEW Diabetic kidney disease (DKD) is the leading cause of kidney failure in the USA, representing ~ 44% of all cases of kidney failure. Advancements in both glucose management and inhibitors of the renin-angiotensin system have significantly improved prognosis for individuals with DKD, yet DKD continues to affect 30-40% of people with type 2 diabetes and is still a major predictor of mortality in this population. Thus, new interventions are required to address this significant health burden. RECENT FINDINGS One potential target for intervention is cellular senescence. Senescence permanently arrests cell division in response to genotoxic, oncogenic, or metabolic stresses-coupled to the secretion of inflammatory cytokines, chemokines, growth factors, proteases, and other molecules that can have potent local and systemic effects. This senescence-associated secretory phenotype (SASP) explains how a relatively small number of senescent cells can promote pathology, and a growing number of degenerative conditions have been found to be caused or aggravated by senescent cells. Many SASP factors are also associated with loss of kidney function. Targeted elimination of senescent cells prevents the development of several degenerative pathologies. Since senescent cells appear in the proximal tubules and podocytes of patients with DKD, they are an appealing target for intervention in these disorders. Here, we review the current literature linking senescence to DKD and speculate on the likely routes to intervention in a clinical setting.
Collapse
Affiliation(s)
- Christopher D Wiley
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| |
Collapse
|