Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

Cellular Senescence in Disease
Cellular Senescence in Disease
Cellular Senescence in Disease
Ebook969 pages9 hours

Cellular Senescence in Disease

Rating: 0 out of 5 stars

()

Read preview

About this ebook

Winner of the 2023 PROSE award in Biomedicine and Neuroscience!

 

Research in the field of senescence has boomed recently due to the gradual realization that senescent cells are associated with a significant number of diseases. The genetic or pharmacological elimination of senescent cells can cause widespread benefits and improves outcomes for most of those diseases. Cellular Senescence in Diseases presents an updated review of the role of cellular senescence in multiple pathologies. Focus is given to those diseases where the implication of senescence has been more extensively documented, such as (cancer, lung and liver diseases, diabetes, Neurodegenerative diseases and others).

The Editors recruited a group of worldwide experts in each individual pathology to review the role of cellular senescence in each one of them, aiming at identifying potential therapeutic pathways. The first two chapters provide an overview of the cellular senescence principles. Next, the chapters are divided into specific diseases. Cancer, including premalignant lesions (OIS), Advanced disease (TIS), and Metastasis are covered. The following condition covered is Lund diseases, including IPF, COPD, and Pulmonary Hypertension. Next Liver Diseases are covered, including Fibrosis and Cirrhosis, and Fatty liver disease. Next there is coverage for Kidney implications, including fibrosis and transplantation. Vascular diseases are covered next including infarction and hear fibrosis, and atherosclerosis. Both diabetes types 1 and 2 are covered next. Following chapters cover Obesity, Sarcopenia, and Bone and Cartilage disorders, respectively. Neurodegenerative diseases are covered next, focusing on Alzheimer and Parkinson. The next chapter discusses accumulation of senescent cell in tissues during aging. The two final chapters cover current developments and conclusions.

Cellular Senescence in Diseases is designed for researchers and clinicians with a focus on the cellular mechanisms of diseases.  All chapters cover current experimental therapeutic approaches to eliminate or cancel the pathological effects of senescent cells.  Pharmaceutical scientists may also benefit from the contents of the book in the exploration of novel therapeutic opportunities.

  • 2023 PROSE Awards - Winner: Category: Biomedicine and Neuroscience: Association of American Publishers
  • Provides a thorough introduction to Cellular Senescence
  • Covers all major pathologies for which cellular senescence has shown evidence of involvement
  • Focuses on possible therapeutic pathways
  • Edited and authored by worldwide experts
LanguageEnglish
Release dateNov 27, 2021
ISBN9780128225158
Cellular Senescence in Disease

Related to Cellular Senescence in Disease

Related ebooks

Biology For You

View More

Related articles

Reviews for Cellular Senescence in Disease

Rating: 0 out of 5 stars
0 ratings

0 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    Cellular Senescence in Disease - Manuel Serrano

    Cellular Senescence in Disease

    Editors

    Manuel Serrano

    Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain

    Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain

    Daniel Muñoz-Espín

    CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom

    Table of Contents

    Cover image

    Title page

    Copyright

    Contributors

    Foreword

    Part 1. Fundamentals

    Chapter 1. Cellular senescence: from old to new testament

    Old testament—genesis of senescence

    New testament—from mechanisms and roles of senescence to therapies

    Part 2. Cellular senescence in disease states

    Chapter 2. Premalignant lesions and cellular senescence

    Introduction

    Cellular senescence in premalignant lesions: evidence in various organs

    Future perspectives

    Chapter 3. Lung aging and senescence in health and disease

    Introduction

    Normal lung development and aging

    A brief introduction to COPD and IPF

    Abnormal hallmarks of lung aging in COPD and IPF

    Future treatment targeting lung senescence

    Conclusions

    Supported

    Abbreviations

    Chapter 4. Cell senescence in pulmonary hypertension

    Introduction

    Pulmonary hypertension, a non-aging-related proliferative vascular disorder at the crossroads of vascular disease and cancer

    General considerations about aging of the systemic and pulmonary vascular systems

    Considerations about constitutive cells of pulmonary vessels and the specificity of the pulmonary vasculature

    Potential mechanisms accounting for cell senescence in PH and PAH

    Role for senescent cells in PH and PAH

    Conclusion

    Chapter 5. Liver diseases fibrosis and cirrhosis

    Liver structure and function

    Cellular senescence

    Liver diseases—epidemiology and clinical aspects

    Senescence during aging of the healthy liver

    Senescence in acute liver injury

    Senescence in chronic liver disease

    Role of senescence in hepatic dysfunction

    Evolutionary role of senescence in the liver

    Senescence during hepatic carcinogenesis

    Summary and closing comments

    Chapter 6. Cellular senescence during aging and chronic liver diseases: mechanisms and therapeutic opportunities

    Introduction

    Cellular senescence in the liver

    Mechanisms contributing to cellular senescence in liver

    Therapies: senolytic and senostatic drugs

    Conclusions and outstanding questions

    List of abbreviations

    Chapter 7. Kidney diseases: fibrosis

    Introduction

    Fibrosis is a common feature of unresolved kidney damage and kidney aging

    Glomerulosclerosis, vascular sclerosis, tubulointerstitial fibrosis

    ECM in renal homeostasis, injury, and repair

    Cellular senescence in renal aging, AKI, and CKD

    Different forms and different timing of cell-cycle arrest may have detrimental or beneficial effects in AKI and CKD

    TIF and cellular senescence

    Senescence and the cellular origin of TIF

    Senescence-associated secretory phenotype

    Transforming growth factor-β (TGF-β)

    WNT/β-catenin signaling

    The renin-angiotensin system (RAS)

    The antiaging factor Klotho

    Inflammation, innate immunity, and TIF

    Senescence and TIF—physiology and pathology

    Chapter 8. Kidney diseases: transplantation

    Introduction

    Factors determining donor organ quality and their association with senescence

    Impact of cellular senescence on transplant-related injuries and transplant outcome

    Rejuvenating and protecting kidney transplants

    Chapter 9. Vascular diseases: atherosclerosis and atherosclerotic cardiovascular diseases

    Introduction—a man is as old as his arteries

    Features of cellular senescence in vascular cells

    Evidence of cellular senescence in atherosclerosis

    Factors involved in vascular senescence associated with the pathophysiology of atherosclerosis

    Therapeutics targeting cellular senescence for atherosclerosis

    Conclusions (limitations and perspectives)

    Chapter 10. Diabetes: senescence in type 1 diabetes

    Introduction

    Senescent beta cell accumulation as a novel pathogenic mechanism in T1D

    Outstanding questions and future directions

    Conclusions and outlook

    Chapter 11. Senescence in obesity: causes and consequences

    Introduction

    Adipose tissue function in obesity

    Senescent cells accumulate in multiple tissues in obesity

    Causes of cellular senescence in obesity

    Threshold theory of senescent cell burden

    Implications of senescence in obesity: downstream effects

    Strategies to target obesity-related senescent cells

    Chapter 12. A framework for addressing senescent cell burden in the osteoarthritic knee: therapeutics, immune signaling, and the local-systemic interface

    Introduction

    Main

    Conclusion

    Competing interests

    Chapter 13. Osteoporosis and bone loss

    Osteoporosis as a public health problem

    The hallmarks of aging in bone

    The role of cellular senescence in mediating age-related bone loss

    Estrogen deficiency and cellular senescence

    The role of cellular senescence in the effects of diabetes mellitus on bone

    Cellular senescence and radiation- and chemotherapy-induced bone loss

    Role of cellular senescence in the growth plate and regulation by parathyroid hormone–related peptide (PTHrP)

    Summary and conclusions

    Chapter 14. Cellular senescence in neurodegenerative diseases

    Cellular senescence: driving force or beneficial response in neurodegeneration?

    Postmitotic senescence in brain aging and neurodegenerative diseases

    Replicative senescence in brain aging and neurodegenerative diseases

    Demyelination, oligodendrocyte lineages, and Aβ plaque propagation in AD brains

    Senolytics as AD therapy

    Combination therapy for AD using senolytics and senomorphics

    Conclusions

    Chapter 15. Cell senescence is a cause of frailty

    What is frailty?

    How is frailty assessed in humans?

    How is frailty defined and assessed in experimental animals?

    What are the possible causes of frailty?

    What is cell senescence?

    Does cell senescence cause frailty? How good is the evidence for it?

    Conclusions

    Part 3. Conclusions

    Chapter 16. Senescence as a therapeutic target: current state and future challenges

    Biological interpretations of cellular senescence

    Tissue remodeling by senescence is a two-step process

    The two steps of senescence in cancer

    The two steps of senescence in disease

    SASP-induced tissue repair versus SASP-induced tissue dysfunction

    Molecular triggers of senescence

    Triggers of senescence in vivo

    The challenge of detecting cellular senescence in clinical settings

    Potential side effects of eliminating senescent cells

    Senolytics as an anti-aging strategy

    Index

    Copyright

    Academic Press is an imprint of Elsevier

    125 London Wall, London EC2Y 5AS, United Kingdom

    525 B Street, Suite 1650, San Diego, CA 92101, United States

    50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States

    The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom

    Copyright © 2022 Elsevier Inc. All rights reserved.

    No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

    This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be noted herein).

    Notices

    Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research methods, professional practices, or medical treatment may become necessary.

    Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility.

    To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein.

    Library of Congress Cataloging-in-Publication Data

    A catalog record for this book is available from the Library of Congress

    British Library Cataloguing-in-Publication Data

    A catalogue record for this book is available from the British Library

    ISBN: 978-0-12-822514-1

    For information on all Academic Press publications visit our website at https://www.elsevier.com/books-and-journals

    Publisher: Stacy Masucci

    Acquisitions Editor: Andre G. Wolff

    Editorial Project Manager: Barbara Makinster

    Production Project Manager: Punithavathy Govindaradjane

    Cover Designer: Miles Hitchen

    Typeset by TNQ Technologies

    Contributors

    Shariq Abid,     Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France

    Serge Adnot

    Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France

    AP-HP Hôpital Henri Mondor, Département de Physiologie-Explorations Fonctionnelles, FHU SENEC, Créteil, France

    Institute for Lung Health, Justus Liebig University, Giessen, Germany

    Alvar Agusti

    Department of Pulmonology, ICR, Hospital Clinic, Barcelona, Spain

    IDIBAPS, Barcelona, Spain

    University of Barcelona, Barcelona, Spain

    CIBERES, Madrid, Spain

    Delphine Beaulieu,     Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France

    Anil Bhushan,     Department of Medicine, University of California San Francisco, San Francisco, CA, United States

    Thomas G. Bird

    Cancer Research UK Beatson Institute, Glasgow, United Kingdom

    Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom

    MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom

    Emmanuelle Born,     Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France

    Marielle Breau,     Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France

    Alexander F. Chin,     Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States

    Jennifer H. Elisseeff

    Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States

    Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States

    Zulrahman Erlangga,     Children's Hospital, Hannover Medical School, Hannover, Germany

    Konstantinos Evangelou,     Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece

    Rosa Faner

    IDIBAPS, Barcelona, Spain

    CIBERES, Madrid, Spain

    Joshua N. Farr,     From the Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, MN, United States

    Eleni Georgakopoulou,     Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece

    Estela González-Gualda,     CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom

    Vassilis G. Gorgoulis

    Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece

    Biomedical Research Foundation, Academy of Athens, Athens, Greece

    Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom

    Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece

    Faculty of Health and Medical Sciences, University of Surrey, Surrey, United Kingdom

    Elise Gray-Gaillard,     Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States

    Jin Han,     Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States

    Fernanda Hernandez-Gonzalez

    Department of Pulmonology, ICR, Hospital Clinic, Barcelona, Spain

    Institut de Recerca Biomedica, Barcelona, Spain

    IDIBAPS, Barcelona, Spain

    Amal Houssaini

    Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France

    Institute for Lung Health, Justus Liebig University, Giessen, Germany

    Michael D. Jensen,     Endocrine Research Unit, Rochester, MN, United States

    Diana Jurk

    Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States

    Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States

    Goro Katsuumi

    Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan

    Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan

    Sundeep Khosla,     From the Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, MN, United States

    Christos Kiourtis

    Cancer Research UK Beatson Institute, Glasgow, United Kingdom

    Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom

    James L. Kirkland,     Robert and Arlene Kogod Center on Aging, Rochester, MN, United States

    Marta Kovatcheva,     Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain

    Larissa Lipskaia

    Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France

    AP-HP Hôpital Henri Mondor, Département de Physiologie-Explorations Fonctionnelles, FHU SENEC, Créteil, France

    Jose Alberto López-Domínguez,     Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain

    David Macías,     CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom

    Elisabeth Marcos,     Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France

    Mate Maus,     Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain

    Anette Melk,     Children's Hospital, Hannover Medical School, Hannover, Germany

    Kathleen Meyer,     Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain

    John Michel,     Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States

    Tohru Minamino

    Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan

    Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan

    Satomi Miwa,     Newcastle University Biosciences Institute, Newcastle upon Tyne, United Kingdom

    David G. Monroe,     From the Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, MN, United States

    Daniel Muñoz-Espín,     CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom

    Hui-Ling Ou,     CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom

    Allyson K. Palmer,     Robert and Arlene Kogod Center on Aging, Rochester, MN, United States

    Nayuta Saito,     Institut de Recerca Biomedica, Barcelona, Spain

    Roland Schmitt,     Hannover Medical School Department of Nephrology and Hypertension, Hannover, Germany

    Jacobo Sellares

    Department of Pulmonology, ICR, Hospital Clinic, Barcelona, Spain

    IDIBAPS, Barcelona, Spain

    University of Barcelona, Barcelona, Spain

    CIBERES, Madrid, Spain

    Manuel Serrano

    Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain

    Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain

    Myong-Hee Sung,     Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States

    Tamara Tchkonia,     Robert and Arlene Kogod Center on Aging, Rochester, MN, United States

    Peter J. Thompson

    Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada

    Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada

    Thomas von Zglinicki

    Newcastle University Biosciences Institute, Newcastle upon Tyne, United Kingdom

    Ageing Biology Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom

    Tengfei Wan,     Newcastle University Biosciences Institute, Newcastle upon Tyne, United Kingdom

    Peisu Zhang,     Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States

    Foreword

    The concepts of aging, disease, and immortality have been intermingled since the early 1900s, when Carrel proposed that cells were immortal, despite the fact that organisms were mortal. ¹ Of course, much of that misconception was dispelled by Hayflick and colleagues in the 1960s ² , ³ , with the first formal description of what is now known as cellular senescence. This book captures our current understanding of this complex cellular phenomenon, as well as new exciting ideas that now mark the field with the promise of novel interventions into aging. Not quite immortality … but hope of a healthy life, relatively unscarred by the chronic disabilities that so often plague the elderly.

    Cellular senescence is a multifaceted cell fate. Vertebrate cells adopt this fate in response to both endogenous and exogenous stressors, as well as evolutionarily selected physiological signals. Our understanding of cellular senescence, its deleterious effects, and its beneficial effects has virtually exploded in recent decades. The implications of this increase in knowledge is not only documented here, but, importantly, its implications for human health are explored in detail.

    Whether and to what extent interventions that modify or eliminate cellular senescence will have an impact on life span remains a big unknown. But there is rising evidence, deftly described here, that cellular senescence does indeed contribute to many of the chronic diseases of aging, and that an increase in health span—years of healthy life—may well be on the horizon as a result of our still-evolving understanding of cellular senescence.

    This book is especially suitable for those who practice biogerontology—whether as physicians dedicated to treating aged patients or as basic scientists interested in translational aspects of cellular senescence. It provides a wealth of knowledge from a wealth of leaders in the field. And it should also entice those curious about the aging process and the promises of new fact-based interventions.

    Judith Campisi

    Professor, The Buck Institute

    Senior Scientist, Lawrence Berkeley National Laboratory

    References

    1. . Carrel A. On the permanent life of tissues outside of the organism.  J Exp Med . 1912;15:516–528.

    2. . Hayflick L. The limited in vitro lifetime of human diploid cell strains.  Exp Cell Res . 1965;37:614–636.

    3. . Hayflick L, Moorhead P.S. The serial cultivation of human diploid cell strains.  Exp Cell Res . 1961;25:585–621.

    Part 1

    Fundamentals

    Outline

    Chapter 1. Cellular senescence: from old to new testament

    Chapter 1: Cellular senescence

    from old to new testament

    Estela González-Gualda, Hui-Ling Ou, David Macías, and Daniel Muñoz-Espín     CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom

    Abstract

    Senescence is a cellular state in response to irreparable damage discovered 60 years ago and characterized by the implementation of a stable cell cycle arrest and a potent secretory phenotype affecting the nearby tissue. From its early days to date, our perception and definition of cellular senescence and its roles (see Box 1.1) have been subjected to constant and significant changes. The complexity of this process is exemplified by its close association with both physiological and pathological states in living organisms. Senescence plays a fundamental role in tissue repair, the maintenance of homeostasis, tumor suppression, and even embryonic development, but, in sharp contrast, it can also contribute to the progression and pathological manifestations of multiple chronic medical conditions, and has been convincingly shown to be a crucial hallmark of aging. Senescence is therefore a defining feature of a variety of age-related disorders, including cancer, and targeted elimination of these cells has recently emerged as a promising therapeutic approach to ameliorate tissue damage and promote repair and regeneration. This introductory chapter will take the reader from the origins of cellular senescence to the landmark discoveries made by its Prophets over the past decades (Fig. 1.1), which have transformed the field of senescence into a new research paradigm that may change our vision of aging and disease, and yield novel therapeutic modalities for chronic—so far untreatable—disorders.

    Keywords

    Cell cycle arrest; Oncogene; Senescence; Senotherapy; Telomere; Tumor suppression

    Old testament—genesis of senescence

    The process of cellular senescence was described for the first time in a seminal study by Hayflick and Moorhead (1961). The authors cultured normal human diploid fibroblasts and, after serial passages in vitro, they realized that the cells entered a state of irreversible growth arrest, in contrast to cancer cells, which were able to proliferate indefinitely in culture. This ground-breaking study was the starting point of an entire research field and led the authors to conjecture a visionary hypothesis at the time: senescence was linked to organismal aging. Importantly, the authors also posed the idea that certain cellular factors may be lost or diluted through consecutive cellular divisions, thereby restricting the proliferative capacity of normal cells.

    BOX 1.1

    The 10 Commandments Of Cellular Senescence

    This work laid the foundations for the concept of replicative senescence, and established the maximum number of potential cell doublings (around 50 divisions), which was coined the Hayflick limit (Hayflick, 1965). Through their observations, the authors speculated that the finite lifetime of diploid cell strains in vitro may be an expression of aging or senescence at the cellular level, and that the ultimate accrual of nondividing cells could be the result of accumulated damage to a single or multiple cellular targets.

    For many years, the field of senescence stood largely abeyant, and the intrinsic molecular mechanisms remained unknown. During this time, the onset of cellular  senescence continued to be considered as one of the main cellular theories of aging, together with other cellular (e.g., free radical), evolutionary (e.g., mutation accumulation), and molecular (e.g., error-catastrophe) theories of aging (Martin, 1980). At this stage, despite reported in cell cultures, senescence had not formally been demonstrated in vivo. In 1979, impairment of cell proliferation was found in ex vivo cultures of skin fibroblasts derived from skin biopsy samples obtained from old individuals as part of a longitudinal study on human aging. Importantly, such halt in proliferation was not observed in young individuals, which suggested a connection between senescence and aging (Schneider et al., 1979). Further evidence of reduced clonogenic potential was generated with preadipocytes obtained from old rat adipose tissue (29 months old vs. 3 months old) in culture (Kirkland et al., 1990). These studies therefore represent the first evidence of senescence occurring in living organisms. However, the assessment of the senescent phenotype was restricted to the proliferative capacity of the cells in culture, given that the hallmarks of senescence (Fig. 1.2) and mechanistic insights of this program (Fig. 1.3) had not been discovered and described thus far, and therefore no reliable biomarkers were available at the time.

    Figure 1.1 Landmark discoveries of cellular senescence.Timeline depicting the discovery of cellular senescence and the main findings over the past decades to date.

    Figure 1.2 Hallmarks of cellular senescence.Senescence is a program that is elicited in response to various stimuli, including damage and stress. Depending on the trigger, it can be classified into different types: replicative senescence, which occurs when telomeres shorten or become dysfunctional after sequential replications; damage-induced senescence, which can be further subdivided into therapy-induced senescence or oxidative-stress senescence, for instance, and oncogene-induced senescence, which takes places upon the aberrant activation of an oncogene or the loss of a tumor suppressor. Despite being elicited by different insults, several traits and mechanisms are generally preserved among the different types, and they constitute the so-called hallmarks of senescence. These include a stable proliferative arrest at the G1 phase of the cell cycle, driven by the activation and cooperation of different proteins involved in the p21CIP1/p53 and p16INK4a/Rb pathways. Senescent cells also present epigenetic changes and chromatin reorganization, which includes the formation of well-established structures named SAHFs (Senescence-Associated Heterochromatin Foci) and DNA-SCARS (DNA Segments with Chromatin Alterations Reinforcing Senescence), as well as the disruption of the nuclear membrane by the downregulation of Lamin B1 expression and the presence of more than one nucleus in the cell (multinucleation). In addition, senescent cells are characterized by metabolic changes and the accumulation of macromolecular damage, which results in higher ROS levels and dysfunctional mitochondria. An additional hallmark is the well-described resistance to apoptosis, which results from the upregulation and activation of prosurvival signaling pathways. Senescent cells also display an increased lysosomal compartment and the overexpression of SA-β-gal (Senescence-Associated β-galactosidase), an enzymatic activity commonly used for the detection of senescence in vitro and in tissues. The senescent program also features the implementation of a strong paracrine secretion of factors to the surrounding tissue, termed SASP (Senescence-Associated Secretory Phenotype). Further to these traits, senescent cells in vitro generally adopt a flattened and enlarged cellular morphology, probably derived from the adoption of the different structural and metabolic changes. Finally, senescent cells have more recently been described to induce the expression of what is now surging as the senescent surfaceome. While all these elements are strongly associated to the senescent phenotype, they may not always be present or detected in senescent cells, as the majority are not exclusive nor indispensable for the implementation of the senescent program, except for the cell cycle arrest. As a consensus in the field, the presence of more than two of the hallmarks described above are generally sufficient to confirm the induction of cellular senescence in vitro and in vivo.

    New testament—from mechanisms and roles of senescence to therapies

    Replicative senescence and telomeres

    Landmark studies performed in the late 1980s contemplated the possibility that the finite doubling capacity of normal mammalian cells could be due to a loss of telomeric DNA and the eventual degradation or deletion of essential genomic sequences (Cooke & Smith, 1986), and it was found that a mutation resulting in defective telomere elongation led to senescence in yeast (Lundblad & Szostak, 1989). Definitive evidence that the amount and length of telomeric DNA decreases as a function of serial passage in culture was obtained with human fibroblasts (Harley et al., 1990), thereby proposing the causality of the gradual loss of telomeric DNA in the implementation of the senescent program (Fig. 1.3A). This scenario was radically different from that of immortal or cancer cells, where the loss of telomeric DNA can be balanced by telomere elongation through the activation of telomerase, a ribonucleoprotein with DNA polymerase activity (Greider & Blackburn, 1985), detected in HeLa cells (Morin, 1989). The above observations were confirmed when retinal epithelial cells and foreskin fibroblasts were transfected with vectors driving the ectopic expression of the human telomerase catalytic subunit, which resulted in elongated telomeres and eventually led to the bypass of senescence and the immortalization of these cellular types (Bodnar et al., 1998). This experiment formally established a causal link between telomere shortening and cellular senescence in vitro, and introduced the possibility of manipulating differentiated cells to be maintained in a phenotypically healthy and youthful state.

    Oncogene-induced senescence and tumor suppression

    The findings on telomere shortening as an underlying mechanism for replicative senescence led the scientific community to hypothesize in the early 1990s that senescence could act as a tumor suppression response, thereby preventing cellular transformation and unlimited proliferation. Ground-breaking studies pointed at the p53/p21 and p16INK4/pRb axes to have a fundamental role in regulating how oncogenic signaling triggers senescence (Fig. 1.3A). p16INK4 is encoded by the INK4a/ARF locus and was firstly isolated and shown to inhibit the cyclin D/CDK4 complex, preventing retinoblastoma protein (Rb) phosphorylation and cell cycle progression (Serrano et al., 1993). On the other hand, p21 was shown to inhibit the activity of most members of the cyclin/CDK family in a p53-dependent manner (Xiong et al., 1993). In a follow-up study, the authors demonstrated that p16INK4 expression inhibited oncogenic ras-induced proliferation and cellular transformation (Serrano et al., 1995). Like the tumor suppressor p53, the cell cycle-inhibitor p16INK4 is mutated in numerous cancer types and, accordingly, p16-deficient mice were highly prone to develop spontaneous and carcinogen-induced tumors (Serrano et al., 1996).

    Figure 1.3 Molecular mechanisms of cellular senescence. (A) Cell cycle arrest: Despite the multiple triggers of cellular senescence, most of the molecular cascades ultimately converge in the activation of p53 (encoded by TP53) and the cyclin-dependent kinase (CDK) inhibitors p16 (also known as INK4A; encoded by CDKN2A), p15 (also known as INK4B; encoded by CDKN2B), p21 (also known as WAF1; encoded by CDKN1A), and p27 (encoded by CDKN1B). These, in turn, result in the inhibition of CDK1, CDK2, CDK4, and CDK6 which prevents the phosphorylation of the retinoblastoma protein (RB), leading to the suppression of S-phase genes and an ensuing stable cell cycle arrest. DNA damaging triggers activate the DNA-damage response (DDR) pathway. The main mediators of the DDR are the DNA damage kinases ATM, ATR, which phosphorylate and activate the p53/p21 pathway. Aging and epigenetic derepression of the Ink4a/ARF locus also lead to the activation of cell cycle inhibitors p16 and p21. ROS lead to the activation of the MAPK signaling pathway and its downstream effector p38. The aberrant expression of oncogenes or the loss of tumor suppressors lead to p53 activation through the Ras-Raf-MEK-ERK or AKT-mTOR signaling pathways, and TGFβ, an important factor of the SASP, leads to p15, p21, and p27 upregulation via SMAD signaling. Other triggers such as developmental cues and polyploidy activate the AKT, SMAD, and/or Ras-Rad-MEK-ERK pathway for p21 upregulation, while processes such as cell fusion signal through the DDR for p53 activation. (B) SASP: A similar scenario occurs for the implementation of the SASP. SASP implementation is orchestrated by the activation of the transcription factors (TFs) NF-κB and C/EBPβ through a variety of upstream signaling pathways in response to different senescence stimuli. DNA-damaging agents, ROS and OIS, generally activate the expression of SASP TFs via the AKT and/or the Ras-Raf-MEK-ERK axis. Additionally, DNA fragments are also known to trigger the activation of the cGAS/STING signaling, resulting in the activation of the IRF3 TF and subsequent transcription of Type 1 IFN. OIS-derived SASP is dynamic and can also be orchestrated by NOTCH signaling, a process that restrains the inflammatory secretion by inhibiting C/EBPβ at initial stages, and allows the activation of SASP-related super enhancers through NF-κB later on. Finally, accumulating increased levels of TFs reinforce the senescent phenotype through autocrine and paracrine signaling. SASP-derived inflammatory chemokines such as IL-6 and IL-8 promote epigenetic modifications reinforcing the cell cycle arrest through the JAK/STAT cascade, while IL-1α stimulates the activity of NF-κB and C/EBPβ promoting a positive feedback loop on the secretion of other cytokines. OIS, oncogene-induced senescence; TS, tumor suppressor. ROS, reactive oxygen species; SASP, senescence-associated secretory phenotype. 

    Figure adapted from our previous work Páez-Ribes et al. EMBO Mol Med, 2019. https://creativecommons.org/licenses/by/4.0/.

    These and other observations concluded with the discovery that oncogenic ras provokes premature senescence accompanied by the accumulation of p53 and p16INK4 (Serrano et al., 1997) (Fig. 1.3A), a process independent of telomere length, which led to the emergent concept of oncogene-induced senescence (OIS). Conceptually, this seminal study placed senescence as a crucial tumor suppressive mechanism, together with apoptosis, and was the inception for the research community to later suggest the possibility of promoting senescence induction as a barrier against cancer. Consistent with these results, oncogenic ras was found to promote senescence through activation of the MAPK cascade subsequently limiting the consequences of mitogenic signaling (Lin et al., 1998).

    In 2005, a formal demonstration of the concept of OIS in living organisms, including both human and mice, was obtained through a variety of preclinical models and patient tissue samples. Remarkably, senescence was found to be a defining feature of multiple types of premalignant lesions, while malignant and advanced cancer states were characterized by bypassing the senescent program. This is the case of human naevi (Michaloglou et al., 2005), lung adenomas (Collado et al., 2005), hyperplasia in the pituitary gland (Lazzerini Denchi et al., 2005), and initial stages of lymphoma (Braig et al., 2005) and prostate cancer (Chen et al., 2005). Last but not least, further to its role as a barrier against early tumorigenesis, restoration of cellular senescence through the reactivation of endogenous p53 was described to result in tumor clearance and regression in advanced liver carcinoma (Xue et al., 2007), as well as sarcoma (Ventura et al., 2007) and lymphomas (Martins et al., 2006; Ventura et al., 2007). Altogether, the above studies confirmed senescence as a key tumor suppressive mechanism through the halt of the proliferation of precancerous cells, leading to tumor arrest and even regression. Intriguingly, these results also supported the idea to treat human cancers by pharmacologically active agents that could reactivate p53 and induce cellular senescence.

    Damage-induced senescence

    We now know that cellular senescence can be triggered by multiple stimuli, including a variety of sources of damage and stress resulting in DNA damage. It is important to note that mutations and other types of DNA damage cannot only contribute to the onset of senescence, but also to cancer initiation and progression. Further, organismal aging has been largely reported to be accompanied by relevant levels of genomic alterations, which, altogether, emphasizes the interplay between senescence, cancer, and aging. Importantly, early studies demonstrated that the infliction of a variety of forms of DNA damage had the capacity to trigger senescence in cell cultures, mouse models, and human tissues. These sources of damage included genotoxic and cytotoxic drugs as well as radiotherapy (Roberson et al., 2005; Schmitt et al., 2002; te Poele et al., 2002). Of note, telomeric damage was found to trigger senescence through the implementation of DNA-damage response (DDR) involving apical kinases ATM and ATR and transducer checkpoint kinases CHK1 and CHK2 (d'Adda di Fagagna et al., 2003) (Fig. 1.3A). Also, the rate of telomere shortening is accelerated by oxidative damage, leading to replicative senescence (von Zglinicki et al., 2003).

    Hallmarks of cellular senescence

    The above studies showed that senescence is a very heterogenous response triggered by different stressors and hence the assessment of bona fide senescent cells was dependent on the identification of a more universal biomarker of senescence, in particular, one suitable for detection in cell cultures and in vivo tissue samples. Although still imperfect, the most widely used biomarker of senescent cells is the senescence-associated β-galactosidase (SA-β-gal) activity (Dimri et al., 1995), which is present in most of the forms of senescence and cellular types described so far. Senescent cells are featured by an enlarged or aberrant morphology and by the significant accumulation of lysosomal content. Lysosomal β-galactosidase is encoded by GLB1, and this activity is typically measured at the acidic pH 4.5. However, senescence-associated lysosomal β-galactosidase is detectable at the restrictive pH 6.0 in a colorimetric reaction using the chromogenic substrate 5-bromo-4-chloro-3-indoyl β-D-galactopyranoside (X-gal), which yields an insoluble blue dye when cleaved by lysosomal β-galactosidase. A limitation of this biomarker is that SA-β-gal activity can also be detected in some cellular types/states unrelated to senescence and that this enzymatic reaction requires fresh tissues, so it is not suitable for archived material. Therefore, a formal assessment of cellular senescence is usually performed with a collection of hallmarks and biomarkers of senescent phenotypes (see Fig. 1.2). Despite these limitations, the identification of SA-β-gal activity was crucial for the development of the field.

    SASP

    Another key feature of senescent cells is the implementation of a complex and very active proinflammatory paracrine response, the so-called senescence-associated secretory phenotype (SASP), consisting of a cocktail of cytokines, chemokines, proteases, growth factors, and other tissue-remodeling factors (see Fig. 1.2). Remarkably, the underlying mechanisms and drivers governing the SASP are different and usually independent from those pathways controlling the implementation of a stable cell cycle arrest. Although the acronym of SASP was not initially introduced until 2008, preceding works already illustrated this concept. An early study that performed microarray analyses with fibroblasts, epithelial and endothelial cells showed evidence that replicative senescence triggers mRNA expression patterns mimicking inflammatory wound repair processes (Shelton et al., 1999). This finding was expanded by a pioneering study showing that senescent fibroblasts can promote epithelial cell growth and tumorigenesis of nearby cells by secreting a collection of soluble and insoluble tumor-promoting factors (Krtolica et al., 2001).

    Later, in 2008, the secretory response of senescent cells and its underlying signaling pathways ballooned with the publication of three comprehensive studies (see Fig. 1.3B showing the main SASP drivers and underlying signaling pathways). The SASP was dissected more comprehensively and shown to induce epithelial-mesenchymal transitions (EMT) in recipient cells by a paracrine mechanism largely dependent on interleukin (IL)-6 and IL-8, which were amplified by cell-nonautonomous functions of oncogenic ras and functional loss of p53 tumor suppressor (Coppe et al., 2008). OIS was therefore linked to the activation of an inflammatory transcriptome, a process that requires the cooperation of transcription factor C/EBPβ with IL-6 to amplify the activation of the inflammatory network, including IL-8 expression (Kuilman et al., 2008). Of note, the effects of the SASP were also found to be autocrine, thereby reinforcing the senescent arrest through the secretion of multiple CXCR2-binding chemokines in a molecular program regulated by the NF-κB and C/EBPβ transcription factors, which cooperate to induce CXCR2 expression in senescent lesions (Acosta et al., 2008). Further unbiased screenings identified multiple SASP components that are controlled by inflammasome-mediated IL-1 signaling, including TGF-β family ligands, VEGF, CCL2, and CCL20. This secretory program had the ability to cause paracrine senescence and impact tumor suppression (Acosta et al., 2013). The senescence inflammatory secretome was further reported to modulate the immune system response, and it was found to be crucial for senescence surveillance of premalignant hepatocytes, limiting liver cancer development (Kang et al., 2011). Of note, this study denoted that senescence surveillance driven by the SASP secretome represents an important extrinsic component of the anticancer barrier, which emphasizes a central role of the senescent program in tumor suppression.

    Senescence in physiology, repair, and embryonic development

    In addition to a central function in tumor suppression, cellular senescence was also found to exert a variety of beneficial effects and even key physiological roles (Fig. 1.4). An innovative study analyzing the contribution of senescence in noncancer pathologies showed that senescent-activated stellate cells restrict liver fibrosis, a pathological condition that precedes cirrhosis, in mouse models (Krizhanovsky et al., 2008). This work then showed that senescence limits the fibrogenic response to acute organ tissue damage, and suggested reparative roles of senescence. Tissue repair functions were confirmed by a study that demonstrated that matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing (Jun & Lau, 2010). This conclusion was further supported in a subsequent work showing a key role of senescent cells in optimal wound healing through secretion of platelet-derived growth factor AA (PDGF-AA), which was identified as a SASP reparative component (Demaria et al., 2014). Altogether, these studies concluded a fundamental role of cell senescence in promoting tissue repair.

    Besides the above processes, senescence can also participate in tissue regeneration. This was first observed in salamanders, organisms where an amputated limb can be fully regenerated. In this injury model, senescence was found to be abundant around the sectioned area, and it was proposed that the effective immunosurveillance of senescent cells by macrophages after paracrine reparative signaling contributes to salamander limb regeneration (Yun et al., 2015). It should be noted that physiological or beneficial roles of senescence, including tumor suppression, tissue repair, regeneration, or developmental senescence, are usually accompanied by an efficient SASP-mediated immunoclearance of senescent cells.

    The close interplay between tissue damage, senescence, and tissue repair or regeneration has been further expanded by the finding that the senescent secretome can induce plasticity, dedifferentiation processes and promote stemness in nearby cells. In a reprogrammable mouse model (Abad et al., 2013), senescence was demonstrated to be in close proximity to reprogrammed cells upon the in vivo-induced expression of the Yamanaka factors, Oct4, Sox2, Klf4, and cMYC (OSKM) in the context of tissue injury and aging (Mosteiro et al., 2016). OSKM-induced senescence created a permissive environment for reprogramming and repair, a process involving the INK4a/ARF locus and IL-6. Additional studies confirmed that SASP factors in response to OIS or injury, including IL-6, induce cellular plasticity and regeneration in different tissues such as the skin and liver (Ritschka et al., 2017) and the skeletal muscle (Chiche et al., 2017), exhibiting increased expression of stem cell markers.

    Figure 1.4 Antagonistic roles of cellular senescence in physiology and pathology.The SASP has been reported to play dual roles in physiology and disease. Senescent cells can implement a particular combination of factors that orchestrate crucial developmental processes during embryogenesis, tissue repair, and regeneration, and drive immune cell recruitment to promote immunosurveillance and tumor suppression. Importantly, the SASP can also induce cellular senescence in nearby cells, as well as it has also been reported to reinforce the senescent state and cell cycle arrest in an autocrine manner. Conversely, in chronic conditions, under persistent damage or stress and during aging, senescent cells accumulate and are not effectively cleared by the immune system. In such scenarios, the SASP has been shown to promote immune evasion, tissue dysfunction, chronic inflammation, and even drive tumor promotion and metastasis.

    An unexpected finding was the physiological role of cellular senescence reported in multiple embryonic structures during mammalian and vertebrate's development, including in humans and mice (Muñoz-Espín et al., 2013; Storer et al., 2013). In these studies, senescence was shown to promote tissue remodeling and organogenesis (e.g., apical ectoderm ridge and the closing neural tube), driving the elimination of transient embryonic structures (e.g., mesonephros), and maintaining the cellular balance (e.g., endolymphatic sac). In contrast to OIS, damage-induced senescence, or replicative senescence, developmental senescence is a programmed process and not a response to stress or stochastic damage. Developmentally-programmed senescence was found to be driven by p21 in a p53-dependent manner and correlated with the upregulation of different developmental pathways. Other examples of physiological senescence include placental syncytiotrophoblasts (Chuprin et al., 2013) and normal megakaryocytes (Besancenot et al., 2010), which undergo senescence during their maturation and differentiation programs.

    In conclusion, cellular senescence can not only play a variety of beneficial and physiological roles in the adult, instructing the nearby tissue to promote repair and regeneration, but it also serves as a crucial mechanism during embryonic development. This finding opened up the possibility that cellular senescence emerged as a developmental tissue remodeling process during evolution, and that it was later adapted as a reparative response to damage in the adult organism.

    Senescence in aging and age-related disorders

    Despite the above beneficial aspects of senescence, this cellular response can also drive detrimental processes (Fig. 1.4). This usually happens in the context of chronic disorders or during aging, when senescent cells are not effectively cleared by the immune system and accumulate in tissues in response to persistent and irreparable damage. When first discovered by Hayflick and Moorhead (1961) cellular senescence was associated with organismal aging and proposed to contribute to its progression. Indeed, the increased senescent burden during aging has since then been documented in multiple tissues in humans, primates, and mice (Herbig & Sedivy, 2006; Jeyapalan et al., 2007; Krishnamurthy et al., 2006; Lawless et al., 2010; Wang et al., 2010). An early study showed that INK4a/ARF expression (encoding for p16INK4 and ARF), key mediator of senescence, is a biomarker and potential effector of aging (Krishnamurthy et al., 2004), providing for the first time a functional link between senescence and aging. Remarkably, this report additionally showed that a delay in senescence accumulation in different mouse models (such as through caloric restriction) correlates with an increased healthspan and lifespan, suggesting for the first time that targeting senescent cells could ameliorate some age-related pathological manifestations. In 2011, this hypothesis was confirmed in a seminal study that represented the origin of senotherapies (see Section below). In this work, the authors developed a transgenic mouse model, namely INK-ATTAC that allowed the pharmacogenetic ablation of p16INK4-positive cells upon administration of a drug (Baker et al., 2011), which was examined in a BubR1 progeroid mouse background (Baker et al., 2008). Interestingly, life-long removal of p16INK4-positive cells in tissues, like skeletal muscle, the eye, and adipose tissue, delayed the onset of associated age-related pathologies (including sarcopenia, cataracts, and fat loss). These data demonstrated the causal implication of senescence as a direct driver of age-related phenotypes and that the removal of senescent cells could ameliorate or defer tissue dysfunction and promote healthspan. In a subsequent study, the same authors explored the consequences of senescence eradication in naturally aged INK-ATTAC mice (Baker et al., 2016). Importantly, the clearance of p16INK4-positive cells delayed tumorigenesis (lymphomas, sarcomas, and carcinomas) and attenuated age-related deterioration of several organs. These include attenuation of age-related lipodystrophy, amelioration of kidney dysfunction and glomerulosclerosis, as well as mitigation of cardiomyocyte hypertrophy in the heart. Of note, the ablation of senescent cells in naturally aged mice not only promoted healthspan, but also extended lifespan by up to 30%.

    All these and related observations led the scientific community to examine the role of senescence in multiple chronic age-related disorders, by using animal models and the genetic and/or the pharmacological ablation of senescent cells. A number of studies have concluded that chronic senescence can contribute to the progression and even onset of numerous pathologies, including cardiovascular diseases (e.g., atherosclerosis), neurological disorders (e.g., Alzheimer and Parkinson disease), fibrosis (e.g., idiopathic pulmonary fibrosis and kidney fibrosis), metabolic syndromes, type I and II diabetes, obesity, inflammatory diseases, sarcopenia, osteoarthritis, osteoporosis, liver steatosis, and many others (for review see Muñoz-Espín and Serrano (2014)). In addition to these diseases, unresolved (i.e., noncleared) senescence can contribute to cancer by a variety of noncell autonomous (SASP-driven) effects (for review see Faget et al. (2019)) and cell autonomous effects (for review see Lee and Schmitt (2019)). These activities are exemplified by the contribution of senescence to adverse effects of chemotherapy and tumor relapse (Demaria et al., 2017), and by the ground-breaking proposal that senescent cancer cells in response to therapy have the potential to reenter the cell cycle with an enhanced proliferative potential and the acquisition of cancer-stemness properties (Milanovic et al., 2018). Interestingly, this work contrasts with the long-standing dogma that senescence is featured by a permanent and stable cell cycle arrest, at least for the particular case of therapy-induced senescent cancer cells. Finally, it is worth mentioning that an aged, senescent immune system (immunosenescence) has the potential to drive systemic aging and organ dysfunction, thereby promoting the emergence of pathological manifestations and age-related disorders (Yousefzadeh et al., 2021).

    This section should only serve as an introduction to key discoveries, and the specific contributions of senescence to the above age-related disorders, original references, and detailed descriptions of such studies will be dissected in subsequent chapters.

    Prosenescent and antisenescent therapies

    In this section, we would like to stress that due to the antagonistic effects of senescence (Fig. 1.4), both prosenescent and antisenescent therapies can be beneficial, depending on the context. As described above, oncogene-induced senescence represents an important anticancer barrier, but eventually some cells can bypass this process, leading to malignant transformation and unlimited proliferation (i.e., cancer). Even in this case, and conceptually, senescence can still be restored in cancer cells by the appropriate stimulus (Serrano & Blasco, 2007), such as through the restoration or activation of p53 in response to radiation (Martins et al., 2006; Ventura et al., 2007; Xue et al., 2007) or via Nutlin-3a administration (Mouraret et al., 2013). In addition to radiation, there are multiple identified drugs and chemotherapeutic agents capable of efficiently inducing both senescence and apoptosis of cancer cells in patients, including etoposide, doxorubicin, cisplatin, bleomycin, palbociclib, and many others (for review see Ewald et al. (2010)). Besides the benefits reported for the treatment of numerous cancer types (see Nardella et al. (2011)), prosenescent therapies can also be useful for ongoing tissue repair processes in the context of chronic pathologies, as it is the case of preclinical strategies based on CDK4 inhibitor administration for kidney fibrosis (DiRocco et al., 2014) or IL-22 (Kong et al., 2012) and the extracellular matrix protein CCN1 (Kim et al., 2013) in liver fibrosis. Therefore, prosenescent therapies are well-established in the case of cancer and are emerging as promising approaches for the treatment of fibrotic disorders and related diseases.

    On a different note, the work published by Baker and colleagues reporting that the eradication of persistent senescent cells can ameliorate, delay, and even revert age-associated disorders (Baker et al., 2011) has transformed the research field. This article established a direct link between senescence and aging and, conceptually, provided a new therapeutic strategy for chronic diseases with poor or unavailable treatments. In the last 5   years, multiple laboratories and pharmaceutical companies in the world have focused on the design, synthesis, development, and validation of a number of senotherapies, mostly based on senolytic drugs (i.e., pharmacologically active agents selectively targeting and inducing death in senescent cells). In 2015, the first drug combination with senolytic activity, namely Dasatinib   +   Quercetin (D   +   Q), was published in a landmark study (Zhu et al., 2015). By transcriptomics analysis, the authors found increased expression of prosurvival networks in senescent cells, consistent with their reported resistance to undergo apoptosis, and employed RNA interference and computational analyses by using the STRING database to identify potential senolytic targets. Screening of drugs that target these gene products showed that Dasatinib (inhibitor of tyrosine kinases) and Quercetin (a natural flavonoid) had particular promise in clearing different types of senescent cells. The capacity of the combination of

    Enjoying the preview?
    Page 1 of 1