Cell Senescence Methods and Protocols 1st Edition Andrew R. J. Young
Cell Senescence Methods and Protocols 1st Edition Andrew R. J. Young
Cell Senescence Methods and Protocols 1st Edition Andrew R. J. Young
Cell Senescence Methods and Protocols 1st Edition Andrew R. J. Young
1. Cell Senescence Methods and Protocols 1st
Edition Andrew R. J. Young download
https://ebookgate.com/product/cell-senescence-methods-and-
protocols-1st-edition-andrew-r-j-young/
Get Instant Ebook Downloads – Browse at https://ebookgate.com
2. Get Your Digital Files Instantly: PDF, ePub, MOBI and More
Quick Digital Downloads: PDF, ePub, MOBI and Other Formats
Cell Cell Interactions Methods and Protocols Methods in
Molecular Biology Vol 341 1st Edition Sean P. Colgan
https://ebookgate.com/product/cell-cell-interactions-methods-and-
protocols-methods-in-molecular-biology-vol-341-1st-edition-sean-
p-colgan/
Plant Cell Morphogenesis Methods and Protocols 1st
Edition Aleš Soukup
https://ebookgate.com/product/plant-cell-morphogenesis-methods-
and-protocols-1st-edition-ales-soukup/
Stem Cell Migration Methods and Protocols 1st Edition
Claire Magnon
https://ebookgate.com/product/stem-cell-migration-methods-and-
protocols-1st-edition-claire-magnon/
T Cell Trafficking Methods and Protocols 1st Edition
Alf Hamann (Auth.)
https://ebookgate.com/product/t-cell-trafficking-methods-and-
protocols-1st-edition-alf-hamann-auth/
3. Live Cell Imaging Methods and Protocols 1st Edition
Martin Oheim (Auth.)
https://ebookgate.com/product/live-cell-imaging-methods-and-
protocols-1st-edition-martin-oheim-auth-2/
Neural Cell Transplantation Methods and Protocols 1st
Edition Mahendra S. Rao
https://ebookgate.com/product/neural-cell-transplantation-
methods-and-protocols-1st-edition-mahendra-s-rao/
Cell Cycle Synchronization Methods and Protocols 1st
Edition Gaspar Banfalvi (Auth.)
https://ebookgate.com/product/cell-cycle-synchronization-methods-
and-protocols-1st-edition-gaspar-banfalvi-auth/
Cancer Cell Culture Methods and Protocols 1st Edition
Simon P. Langdon
https://ebookgate.com/product/cancer-cell-culture-methods-and-
protocols-1st-edition-simon-p-langdon/
Embryonic Stem Cell Immunobiology Methods and Protocols
1st Edition Naoki Ichiryu
https://ebookgate.com/product/embryonic-stem-cell-immunobiology-
methods-and-protocols-1st-edition-naoki-ichiryu/
4. ME T H O D S I N MO L E C U L A R BI O L O G Y ™
Series Editor
John M. Walker
School of Life Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
For further volumes:
http://www.springer.com/series/7651
6. Cell Senescence
Methods and Protocols
Edited by
Lorenzo Galluzzi, IlioVitale, Oliver Kepp,
and Guido Kroemer
InstitutGustaveRoussy(IGR),INSERMU848,
Villejuif,France
8. v
v
Preface
In Vitro and In Vivo Cell Senescence
Cell senescence, i.e., the process whereby cells permanently lose the possibility to prolifer-
ate without undergoing cell death, can be observed in vitro as well as in vivo, and occurs in
a plethora of distinct model organisms. In both cases, cell senescence can be physiological,
constituting a safeguard mechanism against cells that have accumulated potentially danger-
ous genetic alterations, or can be triggered by exogenous perturbations, such as the admin-
istration of DNA-damaging agents at low doses. This book provides a detailed description
of the most common techniques for the investigation of cell senescence, in model organ-
isms encompassing bacteria (Escherichia coli), fungi (Saccharomyces cerevisiae and Podospora
anserina), worms (Caenorhabditis elegans), flies (Drosophila melanogaster), zebrafish (Danio
rerio), and mammalian cells. The techniques presented in this book not only cover the
study of all the biochemical and functional manifestations of senescence at the cellular level
but also include protocols for population analysis and high-throughput approaches in suit-
able model organisms, as described by worldwide renowned experts of the field.
Chapter Organization
The book is composed of three types of chapters. Four review chapters open the book to
provide a solid theoretical background on cell senescence, its morphological and biochemi-
cal manifestations and its pathophysiological relevance. Twenty-three protocol chapters
follow, detailing the methods to investigate the morphological and biochemical features of
senescence at the cellular level, in cultured mammalian cells. Finally, seven protocol chap-
ters provide techniques for the study of cell senescence in lower model organisms, including
methods for population studies. Each of these 30 protocols starts with an Abstract and
includes four major sections: Introduction, Materials, Methods, and Notes. The “Abstract”
presents an overview of the technique(s) detailed in the chapter. The “Introduction” pro-
vides a short theoretical view of the procedure and of its applications. “Materials” recapitu-
late the buffers, reagents, solutions, disposables, and equipments necessary to carry out the
protocol(s). “Methods” describe step-by-step how the technique(s) must be carried out.
Finally, the “Notes” section, which is the hallmark of Methods in Molecular Biology series,
indicates not only the sources of problems and how to identify and overcome them, but
also safety information, alternative procedures, and hints for the correct interpretation of
experimental results.
9. vi
Brief Content of the Chapters
Chapter 1 provides an overview on cell senescence and its dynamic links with autophagy, an
important cytoprotective mechanism. Chapters 2 and 3 discuss the regulation of cell senes-
cence by critical signaling molecules such as the mammalian target of rapamycin (mTOR)
and p53. Chapter 4 summarizes the morphological and biochemical markers that have been
associated with cell senescence. In Chapters 5–23, protocols for the investigation of senes-
cence-associated alterations in cultured cells are provided, including the following: mor-
phological features (Chapter 5), cell cycle blockage (Chapter 6), cell cycle-arresting proteins
(Chapter 7), senescence-associated β-galactosidase (Chapters 8 and 9), senescence-associ-
ated secretory phenotype and chemokyne signaling (Chapters 10 and 11), senescence-
associated heterochromatin foci (Chapter 12), DNA damage (Chapter 13), telomerase
activity and telomere length (Chapters 14 and 15), alterations of the nuclear envelope
(Chapter 16), multiple markers of oxidative stress (Chapters 17–20), BRAF, sirtuin, and
p66SHC
signaling during senescence (Chapters 21–23). In Chapters 24–27, protocols for
the study of cell senescence in global terms are detailed, including a method for the study
of metabolomic alterations (Chapter 24), a technique to apply genome-wide RNAi
approaches to cell senescence research (Chapter 25), and multiparametric strategies
(Chapters 26 and 27). Finally, in Chapters 28–34, protocols applicable to lower model
organisms are described, encompassing techniques to assess senescence in Escherichia coli
(Chapter 28), Podospora anserina (Chapter 29), Saccharomyces cerevisiae (Chapter 30),
Caenorhabditis elegans (Chapters 31 and 32), Drosophila melanogaster (Chapter 33), and
Danio rerio (Chapter 34).
Potential Audience of This Book
In the first instance, this book will be of interest not only for undergraduate and graduate
students but also for more experienced scientists who are approaching the study of cell
senescence. In addition, the audience of this book encompasses:
Libraries of universities and public biological/biomedical research institutions.
●
Scientists interested in molecular and cell biology, biochemistry, pharmacology, genet-
●
ics, systems biology, medicine, public health, and in life sciences in general.
Specialists and experts in model organisms including bacteria, fungi, worms, flies, and
●
mammals.
Medical oncologists and scientists working in oncology.
●
Pharmaceutical companies and developers of new drugs.
●
Villejuif, France Lorenzo Galluzzi
Ilio Vitale
Oliver Kepp
Guido Kroemer
Preface
10. vii
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
1 Cell Senescence as Both a Dynamic and a Static Phenotype. . . . . . . . . . . . . . . . . . 1
Andrew R.J. Young, Masako Narita, and Masashi Narita
2 Senescence Regulation by mTOR. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Vjekoslav Dulic
3 Senescence Regulation by the p53 Protein Family. . . . . . . . . . . . . . . . . . . . . . . . . 37
Yingjuan Qian and Xinbin Chen
4 Markers of Cellular Senescence. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
Amancio Carnero
5 Biomarkers of Cell Senescence Assessed by Imaging Cytometry . . . . . . . . . . . . . . 83
Hong Zhao and Zbigniew Darzynkiewicz
6 Cytofluorometric Assessment of Cell Cycle Progression . . . . . . . . . . . . . . . . . . . . 93
Ilio Vitale, Mohamed Jemaà, Lorenzo Galluzzi,
Didier Metivier, Maria Castedo, and Guido Kroemer
7 Quantification of Cell Cycle-Arresting Proteins. . . . . . . . . . . . . . . . . . . . . . . . . . . 121
Oliver Kepp, Isabelle Martins, Laurie Menger, Mickaël Michaud,
Sandy Adjemian, Abdul Qader Sukkurwala, Lorenzo Galluzzi,
and Guido Kroemer
8 Colorimetric Detection of Senescence-Associated b Galactosidase . . . . . . . . . . . . . 143
Koji Itahana, Yoko Itahana, and Goberdhan P. Dimri
9 Chemiluminescent Detection of Senescence-Associated b Galactosidase . . . . . . . . 157
Vinicius Bassaneze, Ayumi Aurea Miyakawa, and José Eduardo Krieger
10 Detection of the Senescence-Associated Secretory Phenotype (SASP) . . . . . . . . . . 165
Francis Rodier
11 Unbiased Characterization of the Senescence-Associated Secretome
Using SILAC-Based Quantitative Proteomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175
Juan Carlos Acosta, Ambrosius P. Snijders, and Jesús Gil
12 Detection of Senescence-Associated Heterochromatin Foci (SAHF) . . . . . . . . . . . 185
Katherine M. Aird and Rugang Zhang
13 Monitoring DNA Damage During Cell Senescence . . . . . . . . . . . . . . . . . . . . . . . 197
Glyn Nelson and Thomas von Zglinicki
14 Assessment and Quantification of Telomerase Enzyme Activity . . . . . . . . . . . . . . . 215
Michelle F. Maritz, Laura A. Richards, and Karen L. MacKenzie
15 Methods for the Assessment of Telomere Status . . . . . . . . . . . . . . . . . . . . . . . . . . 233
Asako J. Nakamura
11. viii Contents
16 Detection of Nuclear Envelope Alterations in Senescence . . . . . . . . . . . . . . . . . . . 243
Clea Bárcena, Fernando G. Osorio, and José Maria Pérez Freije
17 Measuring Reactive Oxygen Species in Senescent Cells . . . . . . . . . . . . . . . . . . . . . 253
João F. Passos, Satomi Miwa, and Thomas von Zglinicki
18 Quantification of Protein Carbonylation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
Nancy B. Wehr and Rodney L. Levine
19 Assays for the Measurement of Lipid Peroxidation . . . . . . . . . . . . . . . . . . . . . . . . 283
Ana Cipak Gasparovic, Morana Jaganjac, Branka Mihaljevic,
Suzana Borovic Sunjic, and Neven Zarkovic
20 Raman Spectroscopy for the Detection of AGEs/ALEs . . . . . . . . . . . . . . . . . . . . 297
J. Renwick Beattie, John J. McGarvey, and Alan W. Stitt
21 Monitoring Oncogenic B-RAF-Induced Senescence in Melanocytes . . . . . . . . . . . 313
Sieu L. Tran and Helen Rizos
22 Methods to Investigate the Role of SIRT1 in Endothelial Senescence . . . . . . . . . . 327
Bo Bai and Yu Wang
23 Monitoring Nutrient Signaling Through the Longevity Protein p66SHC1
. . . . . . . . 341
Sofia Chiatamone Ranieri and Giovambattista Pani
24 Profiling the Metabolic Signature of Senescence . . . . . . . . . . . . . . . . . . . . . . . . . . 355
Florian M. Geier, Silke Fuchs, Gabriel Valbuena,
Armand M. Leroi, and Jacob G. Bundy
25 Genome-Wide RNAi Screening to Identify Regulators of
Oncogene-Induced Cellular Senescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 373
Narendra Wajapeyee, Sara K. Deibler, and Michael R. Green
26 An Integrated Approach for Monitoring Cell Senescence . . . . . . . . . . . . . . . . . . . 383
Tatiana V. Pospelova, Zhanna V. Chitikova, and Valery A. Pospelov
27 Robust Multiparametric Assessment of Cellular Senescence. . . . . . . . . . . . . . . . . . 409
Clara Correia-Melo, Diana Jurk, and João F. Passos
28 Assessing Chronological Aging in Bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 421
Stavros Gonidakis and Valter D. Longo
29 Assessing Organismal Aging in the Filamentous Fungus Podospora anserina . . . . . 439
Heinz D. Osiewacz, Andrea Hamann, and Sandra Zintel
30 Assessing Chronological Aging in Saccharomyces cerevisiae . . . . . . . . . . . . . . . . . . 463
Jia Hu, Min Wei, Mario G. Mirisola, and Valter D. Longo
31 Assessing Aging and Senescent Decline in Caenorhabditis elegans:
Cohort Survival Analysis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 473
Eirini Lionaki and Nektarios Tavernarakis
32 High-Throughput and Longitudinal Analysis of Aging
and Senescent Decline in Caenorhabditis elegans . . . . . . . . . . . . . . . . . . . . . . . . . . 485
Eirini Lionaki and Nektarios Tavernarakis
33 Assessing Senescence in Drosophila Using Video Tracking . . . . . . . . . . . . . . . . . . 501
Reza Ardekani, Simon Tavaré, and John Tower
34 Assessing Vascular Senescence in Zebrafish . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
Sandra Donnini, Antonio Giachetti, and Marina Ziche
Index. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 533
12. ix
Contributors
JUAN CARLOS ACOSTA • Cell Proliferation Group, MRC Clinical Sciences Centre,
Imperial College, London, UK
SANDY ADJEMIAN • INSERM, U848, Villejuif, France; Institut Gustave Roussy, Villejuif,
France
KATHERINE M. AIRD • Women’s Cancer Program, Epigenetics and Progenitor
Cells Keystone Program, Fox Chase Cancer Center, Philadelphia, PA, USA
REZA ARDEKANI • Molecular and Computational Biology Program, Department of
Biological Sciences, University of Southern California, Los Angeles, CA, USA
BO BAI • Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine,
University of Hong Kong, Hong Kong, China
CLEA BÁRCENA • Departamento de Bioquímica y Biología Molecular,
Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo, Spain
VINICIUS BASSANEZE • Laboratory of Genetics and Molecular Cardiology/LIM 13,
Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil
J. RENWICK BEATTIE • Centre for Vision and Vascular Science, School of Medicine and
Dentistry, Queen’s University, Belfast, UK
JACOB G. BUNDY • Biomolecular Medicine, Department of Surgery and Cancer,
Imperial College, London, UK
AMANCIO CARNERO • Instituto de Biomedicina, Hospital Universitario Virgen del Rocio,
Sevilla, Spain
MARIA CASTEDO • INSERM, U848, Villejuif, France; Institut Gustave Roussy, Villejuif,
France
XINBIN CHEN • Comparative Oncology Laboratory, University of California, Davis, CA, USA
ZHANNA V. CHITIKOVA • Institute of Cytology, Russian Academy of Sciences,
St. Petersburg, Russia; St. Petersburg State University, St. Petersburg, Russia
CLARA CORREIA-MELO • Ageing Research Laboratories, Centre for Integrated Systems
Biology of Ageing and Nutrition, Institute for Ageing and Health, Newcastle University,
Newcastle upon Tyne, UK
ZBIGNIEW DARZYNKIEWICZ • Department of Pathology, Brander Cancer Research Institute,
New York Medical College, Valhalla, NY, USA
SARA K. DEIBLER • Howard Hughes Medical Institute, Chevy Chase, MD, USA; Programs
in Gene Function and Expression and Molecular Medicine, University of Massachusetts
Medical School, Worcester, MA, USA
GOBERDHAN P. DIMRI • Department of Biochemistry and Molecular Biology, The George
Washington University Medical Center, Washington, DC, USA
SANDRA DONNINI • Department of Biotechnology, University of Siena, Siena, Italy
VJEKOSLAV DULIC • Institut de Génétique Moléculaire, Montpellier, France;
CNRS, UMR5535, Montpellier, France; Université Montpellier 1, Montpellier, France
JOSÉ MARIA PÉREZ FREIJE • Departamento de Bioquímica y Biología Molecular, Instituto
Universitario de Oncología, Universidad de Oviedo, Oviedo, Spain
SILKE FUCHS • Cell and Molecular Biology, Department of Life Sciences, Imperial College,
London, UK
13. x Contributors
LORENZO GALLUZZI • Institut Gustave Roussy, Villejuif, France; Université Paris Descartes,
Sorbonne Paris Cité, Paris, France
ANA CIPAK GASPAROVIC • Rudjer Boskovic Institute, Zagreb, Croatia
FLORIAN M. GEIER • Biomolecular Medicine, Department of Surgery and Cancer,
Imperial College, London, UK
ANTONIO GIACHETTI • Department of Biotechnology, University of Siena, Siena, Italy
JESÚS GIL • Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College,
London, UK
STAVROS GONIDAKIS • Department of Biological Sciences, Ethel Percy Andrus Gerontology
Center, University of Southern California, Los Angeles, CA, USA
MICHAEL R. GREEN • Howard Hughes Medical Institute, Chevy Chase, MD, USA;
Programs in Gene Function and Expression and Molecular Medicine,
University of Massachusetts Medical School, Worcester, MA, USA
ANDREA HAMANN • Faculty of Biosciences, Institute of Molecular Biosciences, Johann
Wolfgang Goethe University Frankfurt, Frankfurt, Germany; Frankfurt Cluster of
Excellence “Macromolecular Complexes”, Frankfurt, Germany
JIA HU • Ethel Percy Andrus Gerontology Center, Davis School of Gerontology,
University of Southern California, Los Angeles, CA, USA
KOJI ITAHANA • Duke-NUS Graduate Medical School Singapore, Singapore, Singapore
YOKO ITAHANA • Duke-NUS Graduate Medical School Singapore, Singapore, Singapore
MORANA JAGANJAC • Rudjer Boskovic Institute, Zagreb, Croatia
MOHAMED JEMÀA • INSERM, U848, Villejuif, France; Institut Gustave Roussy,
Villejuif, France
DIANA JURK • Ageing Research Laboratories, Centre for Integrated Systems Biology of
Ageing and Nutrition, Institute for Ageing and Health, Newcastle University,
Newcastle upon Tyne, UK
OLIVER KEPP • INSERM, U848, Villejuif, France; Institut Gustave Roussy, Villejuif, France
JOSÉ EDUARDO KRIEGER • Laboratory of Genetics and Molecular Cardiology/LIM 13,
Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil
GUIDO KROEMER • Centre de Recherche des Cordeliers, Paris, France; INSERM, U848,
Villejuif, France; Metabolomics Platform, Institut Gustave Roussy, Villejuif, France;
Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France;
Université Paris Descartes, Sorbonne Paris Cité, Paris, France
ARMAND M. LEROI • Ecology and Evolution, Department of Life Sciences, Imperial College,
London, UK
RODNEY L. LEVINE • Laboratory of Biochemistry, National Heart, Lung, and Blood
Institute, NIH, Bethesda, MD, USA
EIRINI LIONAKI • Institute of Molecular Biology and Biotechnology, Foundation for
Research and Technology, Heraklion, Greece
VALTER D. LONGO • Davis School of Gerontology, Ethel Percy Andrus Gerontology Center,
University of Southern California, Los Angeles, CA, USA
CARLOS LÓPEZ-OTÍN • Departamento de Bioquímica y Biología Molecular,
Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo, Spain
ISABELLE MARTINS • INSERM, U848, Villejuif, France; Institut Gustave Roussy, Villejuif,
France
KAREN L. MACKENZIE • Cancer Cell Development Group, Children’s Cancer Institute
Australia, Randwick, Australia; Lowy Cancer Research Centre, University of
New South Wales, Randwick, Australia
14. xi
MICHELLE F. MARITZ • Cancer Cell Development Group, Children’s Cancer Institute
Australia, Randwick, Australia; Lowy Cancer Research Centre, University of
New South Wales, Randwick, Australia
JOHN J. MCGARVEY • School of Chemistry and Chemical Engineering, Queen’s University,
Belfast, UK
LAURIE MENGER • INSERM, U848, Villejuif, France; Institut Gustave Roussy, Villejuif,
France
DIDIER METIVIER • INSERM, U848, Villejuif, France; Institut Gustave Roussy, Villejuif,
France
MICKAËL MICHAUD • INSERM, U848, Villejuif, France; Institut Gustave Roussy, Villejuif,
France
BRANKA MIHALJEVIC • Rudjer Boskovic Institute, Zagreb, Croatia
MARIO G. MIRISOLA • Department of Medical and Forensic Biopathology e Biotechnology,
University of Palermo, Palermo, Italy
SATOMI MIWA • Ageing Research Laboratories, Centre for Integrated Systems Biology of
Ageing and Nutrition, Institute for Ageing and Health, Newcastle University,
Newcastle upon Tyne, UK
AYUMI AUREA MIYAKAWA • Laboratory of Genetics and Molecular Cardiology/LIM 13,
Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil
ASAKO J. NAKAMURA • Department of Anatomy and Cell Biology, Osaka Medical College,
Takatsuki, Japan
MASAKO NARITA • Cancer Research UK, Cambridge Research Institute, Cambridge, UK
MASASHI NARITA • Cancer Research UK, Cambridge Research Institute, Cambridge, UK
GLYN NELSON • Ageing Research Laboratories, Centre for Integrated Systems Biology of
Ageing and Nutrition, Institute for Ageing and Health, Newcastle University,
Newcastle upon Tyne, UK
HEINZ D. OSIEWACZ • Faculty of Biosciences, Institute of Molecular Biosciences, Johann
Wolfgang Goethe University Frankfurt, Frankfurt, Germany; Frankfurt Cluster of
Excellence “Macromolecular Complexes”, Frankfurt, Germany
FERNANDO G. OSORIO • Departamento de Bioquímica y Biología Molecular, Instituto
Universitario de Oncología, Universidad de Oviedo, Oviedo, Spain
GIOVAMBATTISTA PANI • Institute of General Pathology, Catholic University Medical School,
Rome, Italy
JOÃO F. PASSOS • Ageing Research Laboratories, Centre for Integrated Systems Biology of
Ageing and Nutrition, Institute for Ageing and Health, Newcastle University,
Newcastle upon Tyne, UK
VALERY A. POSPELOV • Institute of Cytology, Russian Academy of Sciences,
St. Petersburg, Russia; St. Petersburg State University, St. Petersburg, Russia
TATIANA V. POSPELOVA • Institute of Cytology, Russian Academy of Sciences,
St. Petersburg, Russia; St. Petersburg State University, St. Petersburg, Russia
YINGJUAN QIAN • Comparative Oncology Laboratory, University of California,
Davis, CA, USA
SOFIA CHIATAMONE RANIERI • Clinical Chemistry, Laboratory and Endocrinology
Unit, Departement of Laboratory Medicine, Azienda Ospedaliera ASMN,
Istituto di Ricovero e Cura a Carattere Scientifico, Reggio Emilia, Italy
LAURA A. RICHARDS • Cancer Cell Development Group, Children’s Cancer Institute
Australia, Randwick, Australia; Lowy Cancer Research Centre, University of
New South Wales, Randwick, Australia
Contributors
15. xii
HELEN RIZOS • Melanoma Institute Australia, North Sydney, Australia University of
Sydney, Sydney, Australia; Westmead Institute for Cancer Research, Westmead
Millennium Institute for Medical Research, Westmead, Australia
FRANCIS RODIER • Institut du cancer de Montréal, Centre de recherche du CHUM,
Montréal, Canada
AMBROSIUS P. SNIJDERS • Biomolecular Mass Spectrometry and Proteomics Laboratory,
MRC Clinical Sciences Centre, Imperial College, London, UK
ALAN W. STITT • Centre for Vision and Vascular Science, School of Medicine and Dentistry,
Queen’s University, Belfast, UK
ABDUL QADER SUKKURWALA • INSERM, U848, Villejuif, France; Institut Gustave Roussy,
Villejuif, France
SUZANA BOROVIC SUNJIC • Rudjer Boskovic Institute, Zagreb, Croatia
SIMON TAVARÉ • Molecular and Computational Biology Program, Department of
Biological Sciences, University of Southern California, Los Angeles, CA, USA;
Department of Oncology, University of Cambridge, Cambridge, UK; Cancer Research
UK, Cambridge Research Institute, Cambridge, UK
NEKTARIOS TAVERNARAKIS • Institute of Molecular Biology and Biotechnology,
Foundation for Research and Technology, Heraklion, Greece
JOHN TOWER • Molecular and Computational Biology Program, Department of Biological
Sciences, University of Southern California, Los Angeles, CA, USA
SIEU L. TRAN • Melanoma Institute Australia, North Sydney, Australia; University of
Sydney, Sydney, Australia; Westmead Institute for Cancer Research, Westmead
Millennium Institute for Medical Research, Westmead, Australia
GABRIEL VALBUENA • Biomolecular Medicine, Department of Surgery and Cancer,
Imperial College, London, UK
ILIO VITALE • INSERM, U848, Villejuif, France; Institut Gustave Roussy, Villejuif, France
THOMAS VON ZGLINICKI • Ageing Research Laboratories, Centre for Integrated Systems
Biology of Ageing and Nutrition, Institute for Ageing and Health, Newcastle University,
Newcastle upon Tyne, UK
NARENDRA WAJAPEYEE • Department of Pathology, Yale University School of Medicine,
New Haven, CT, USA
YU WANG • Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine,
University of Hong Kong, Hong Kong, China
NANCY B. WEHR • Laboratory of Biochemistry, National Heart, Lung, and Blood Institute,
NIH, Bethesda, MD, USA
MIN WEI • Davis School of Gerontology, Ethel Percy Andrus Gerontology Center,
University of Southern California, Los Angeles, CA, USA
ANDREW R.J. YOUNG • Cancer Research UK, Cambridge Research Institute,
Cambridge, UK
NEVEN ZARKOVIC • Rudjer Boskovic Institute, Zagreb, Croatia
RUGANG ZHANG • Women’s Cancer Program, Epigenetics and Progenitor Cells Keystone
Program, Fox Chase Cancer Center, Philadelphia, PA, USA
HONG ZHAO • Department of Pathology, Brander Cancer Research Institute,
New York Medical College, Valhalla, NY, USA
MARINA ZICHE • Department of Biotechnology, University of Siena, Siena, Italy
SANDRA ZINTEL • Faculty of Biosciences, Institute of Molecular Biosciences, Johann Wolfgang
Goethe University Frankfurt, Frankfurt, Germany; Frankfurt Cluster of Excellence
“Macromolecular Complexes”, Frankfurt, Germany
Contributors
17. 2 A.R.J. Young et al.
While it is widely accepted that both the p53 and p16/Rb
tumor suppressor pathways are involved in senescence, the precise
mechanisms underlying the phenotype are still elusive. Cells
respond to cytotoxic stimuli differently depending upon the cell
type and the strength of the stress. HDFs are relatively resistant to
apoptosis, and are highly prone to senescence. Compared to apop-
tosis, senescence is a delayed and much longer process. The con-
trast in these two stress responsive phenotypes may be due to the
way the signals are conveyed. In apoptosis, a variety of triggers can
converge to the executioner caspases through a common mecha-
nism (4). In contrast, in senescence a number of effector mecha-
nisms, each contributing to the process, have been proposed and it
is likely that these effectors collectively define the phenotype. Thus,
the senescence process can be viewed as a “matrix” composed of a
series of signaling transductions, rather than the single “vector” of
signals in apoptosis (Fig. 1). Such effector programs of senescence
include senescence associated heterochromatic foci (SAHFs) and
epigenetic gene regulation, the DNA damage response, senescence
associated secretory phenotype (SASP)/Senescence-messaging
secretome (SMS), and macroautophagy (5–16). Due to the highly
heterogeneous nature of the senescence phenotype, it has not been
possible to identify a universal marker for senescence that applies in
all cases. However, the use of a combination of various markers
often associated with effector programs of senescence has success-
fully contributed to extending the concept of senescence outside
the HDF system: a similar phenotype can be induced by various
Fig. 1. The juxtaposition of senescence and apoptosis. The contrast between these two
stress responsive phenotypes may be due to the way the signals are conveyed. In apop-
tosis, a variety of triggers can converge to the executioner caspases through a common
mechanism. In contrast, in senescence, a number of effector mechanisms, which can
each contribute to the process, have been proposed and it is likely that these effectors
collectively define the phenotype, and also that the eventual phenotype maybe different
depending on the signals and effectors that induced it. In addition, senescence is typically
a delayed and progressive process. Thus, as schematically illustrated here, the senes-
cence process can be viewed as a “matrix” composed of a series of signal transductions,
rather than the signal “vectors” of apoptosis.
18. 3
1 Dynamic Interaction Between Autophagy and Senescence
cellular stresses in different types of somatic cells, as well as different
tissues in animals (17–20). Importantly, the effector mechanisms
and markers used in these studies were mostly identified in experi-
ments using HDFs, and thus, HDFs are still the best-characterized
model system for this topic. Therefore, in this review, most descrip-
tions are derived from this system unless specifically mentioned
otherwise. We also use the term “senescence” to specifically
describe the cellular phenotype, distinct from “aging” at the
organism level.
The complexity of senescence also implies a “time axis,” by
which senescence can be understood as a delayed and progressive
process. Therefore, it would be sensible to view senescence not
only as a static endpoint, but also as a dynamic process of pheno-
typic establishment (Fig. 1). This distinction becomes more rele-
vant in acute types of senescence, such as oncogene-induced
senescence (OIS). Here, we overview the static and dynamic aspects
of senescence and discuss our current understanding of the com-
plex interactions between the effector programs during senescence,
with a particular focus on the regulation of gene expression.
It was shown in 1997 that oncogenic Ras could induce premature
senescence, in both HDFs and mouse embryonic fibroblasts
(MEFs) (21). As the paradoxical name suggests, the Ras-induced
senescence (RIS) phenotype is nonlinear. Due to the direct impact
of the abnormal mitogenic stimuli, the cells initially proliferate
more rapidly (22). This mitotic phenotype is then gradually
replaced by the senescence phenotype. Interestingly, the in vivo
counterpart of OIS is well represented by some pre-neoplastic or
benign tumors, such as melanocytic nevi, lung adenomas, and pan-
creatic intraductal neoplasias, and chemically induced skin papillo-
mas (23–28) (see ref. 20). These tumors are likely to be a
consequence of cell proliferation during this mitotic phase. Thus,
OIS/RIS may be a model for the initial steps of tumorigenesis,
which include the series of events from the first reaction of the cells
to the mitotic stress, following activation of various effector pro-
grams, and the maintenance of the phenotype. Importantly, each
effector program is typically involved not only in senescence but
also in its original, physiological, role outside of senescence. Thus,
studying the OIS/RIS system should extend our understanding
of each of these effector programs and provide insight into the
more general concepts in dynamic alterations of phenotype, such
as differentiation and transformation.
2. Oncogenic,
Ras-Induced
Senescence
19. 4 A.R.J. Young et al.
The strong stability of the phenotype, compared to the readily
reversible G0 arrest of quiescence, is a critical aspect of senescence,
implying its biological relevance in, for instance, tumor suppres-
sion and therapy (29, 30), aging (31), and the efficiency of repro-
gramming to pluripotent stem cells (32–37). Not surprisingly, one
focus of the earlier studies on senescence was on the irreversibility
of the cell cycle exit. For example, the Campisi group showed that
the refractoriness of cells to mitogens is a hallmark of senescence,
and that some growth-related genes are not responsive to growth
factors (38, 39). The same group later examined the reversibility of
senescence caused by telomere dysfunction using different types of
HDFs, with varying levels of the CDK inhibitor p16. They found
that the replicative senescence arrest could be reversed by p53
depletion in p16-low cells (such as BJ cells), whereas in p16-high
cells (such as WI38 cells) senescence could not be so easily reversed
(40). Thus, while p53 contributes to continuous cell cycle arrest
signaling, the p16/Rb pathway provides a dominant barrier to the
immortalization of human cells. A primary role for the p53 path-
way has also been shown in OIS, where it is activated by a persis-
tent DNA damage response (41). These data also reinforce the
classical view that, in human cells, it is required to abrogate both
the p53 and p16/Rb pathways in order to bypass senescence, and
that most cancers do indeed have defects in both pathways.
We and others have previously shown that HDFs exhibit dra-
matic alterations in heterochromatin during senescence (SAHF) in
a p16/Rb dependent manner, and have proposed that SAHF for-
mation is involved in the stable silencing of at least some cell cycle
genes (42, 43). Interestingly, senescent HDFs with high p16 (non-
reversible by p53 depletion) show more prominent SAHFs com-
pared to p16-low HDFs (reversible by p53 depletion), indicating a
correlation between SAHF formation and the stability of senescence
arrest (5). Furthermore, we have identified chromatin architectural
proteins, HMGA1 and HMGA2, as SAHF components. SAHF dis-
ruption by HMGA1 depletion made cellular senescence bypass
easier, reinforcing the correlation between phenotype stability and
high-order chromatin architecture (44). However, both hetero-
chromatin structure and the related epigenetic marks can also be
dynamic. Thus, it is conceivable that cells need to have additional
mechanisms to achieve the ultimate irreversibility of senescence.
The precise relationship between high-order SAHF structure
and individual gene regulation is still unclear. To investigate this
connection it would be necessary to effectively combine both imag-
ing and biochemical data for SAHF markers, which to date has not
been managed. It is tempting, however, to speculate that such a
clear segregation between euchromatin and heterochromatin in
3. Transcriptional
Regulation During
Senescence
20. 5
1 Dynamic Interaction Between Autophagy and Senescence
the static condition might allow a rather fixed gene expression
profile, giving up dynamic and complex gene expression regulation.
In this sense, SAHFs may be important not only for gene repres-
sion as originally proposed, but may also play a role in the stable
maintenance of gene activation in the euchromatic regions. This
interpretation would extend our understanding of SAHFs beyond
senescence, and rather put the concept of SAHFs in the context of
gene plasticity that is associated with distinct cellular phenotypes.
Conceivably, senescent cells may exhibit a distinct mRNA
expression profile compared to their growing or quiescent coun-
terparts (45). A flurry of papers in the late 2000s defined the func-
tionality of the plethora of factors secreted by senescent cells. This
heterogenous collection of cytokines, chemokines and other fac-
tors, termed the SASP or SMS has been shown to have autocrine
and paracrine functions (10–13, 45, 46)—the autocrine function
being to reinforce the senescence phenotype in the cell of origin
and one potential paracrine function being to induce senescence in
other cells sharing the same DNA damaging/tumorigenic microen-
vironment. Another paracrine function may be the non-serendipi-
tous promotion of the transformation of premalignant—but not of
normal—cells within the microenvironment (47, 48).
Amongtheseriesofpapersdescribingthefunctionalsignificance
of SASP/SMS, the Peeper group notably identified SASP/SMS as
a “secretome” specifically associated with OIS (10). Microarray
experiments showed a set of transcripts highly upregulated in OIS
cells, which were upregulated neither in cells that had bypassed
OIS nor in quiescent cells (10). Thus, it is tempting to speculate
that the “open” areas of chromatin generated by SAHF formation
may permit and promote the expression of the secretome as a unit.
Interestingly, SASP/SMS can also influence SAHF, in that deple-
tion of IL-6 (a central component of SASP/SMS) in OIS corre-
lated with reduced formation of SAHF as well as p15, a
senescence-related CDK inhibitor (CDKI) (10). It has been shown
that over-expression of p16, another CDKI, which is sufficient to
induce senescence and SAHFs in HDFs, fails to trigger SASP/
SMS, indicating that SAHF formation per se cannot activate the
secretome (49). However, in our time series microarray experi-
ments in RIS HDFs, upregulation of SASP/SMS components
starts during the transition phase of RIS, when SAHF formation is
not yet fully established (15). Thus, the two processes may mutu-
ally reinforce each other during the course of OIS establishment:
SASP/SMS facilitating SAHF formation (possibly in part through
p15 upregulation), while high-order chromatin reorganization
might reinforce the stable expression of SASP/SMS components
in the late stages of RIS.
Thus, it is useful to think of OIS as a whole process, not just a
static phenotype, and, along those lines, we recently identified
another effector mechanism that affects SASP/SMS during the
21. 6 A.R.J. Young et al.
RIS-transition, namely macroautophagy (referred to hereafter as
autophagy). As well as transcriptional effects, SASP/SMS is also
facilitated at the posttranscription level.
To study the process and the dynamic features of the senescence
phenotype, 4OHT-inducible RIS is very useful. In the 4OHT RIS
system in HDFs, it typically takes around 7 days to establish the
senescence phenotype (Fig. 2) (15). In this system, much of the
dynamic transcriptional alteration occurs during the RIS transition
(15). However, if rapid protein turnover were to happen at the
same time it would make achieving gene expression changes far
more efficient, and perhaps allow emergent cell remodeling.
Consistent with this idea, autophagy is highly active during the
RIS transition (15).
Autophagy is, essentially, the “trash collector” of the cell, or
rather the “recycling collection truck.” Eukaryotic cells have two
main known mechanisms for protein degradation: the proteasome
and autophagy. The proteasome takes only one protein at a time
and targets are signaled for selection, whereas autophagy can col-
lect cellular components en masse (although, degrees of specificity
are emerging for autophagy too (50)). In autophagy, vesicles form
within the cytoplasm and can be seen to have enclosed cytoplasmic
content. That cytoplasmic content is then trafficked along micro-
tubules to the lysosome. Lysosomes contain the degradative
enzymes of the cell: lipases, proteases, DNase, etc. (51).
Autophagosomes then fuse with lysosomes, the product of which
is termed an autolysosome, and pass their content inside for degra-
dation and recycling. That content is then actively released from
the lysosome (52), and thus, presumably a gradient of metabolites
emanates from the lysosome.
4. Autophagy
and SASP/SMS
in Senescence
Fig. 2. A schematic view of oncogenic Ras-induced senescence (RIS) in human diploid
fibroblasts (HDFs). RIS can be acutely induced by adding 4OHT to HDFs stably expressing
ER:Ras, mediated by retrovirus-mediated gene transfer. The whole process is typically
completed in ~7 days.
22. 7
1 Dynamic Interaction Between Autophagy and Senescence
The emerging dogma is thus that autophagy represents the
“guardian” of the proteome (53), which could be broadened out
to the macromolecule-ome. As macromolecules become damaged,
for instance through oxidation, they become misfunctional. Hence
constant degradation and resynthesis of macromolecules is required,
and is especially required in dire times of cellular stress, such as
oxidative or metabolic stress. In addition to that role, however, we
have shown that autophagy can have quite unforeseen conse-
quences for the cell, impacting on cell fate, in that it can facilitate
the establishment of senescence both cell autonomously and non-
cell-autonomously through its role in the production of SASP/
SMS in RIS, and beyond.
As mentioned in the introduction, the characteristics of senescent
cells are that they typically have enlarged cell and nuclear bodies,
prominent nucleoli, as well as numerous cytoplasmic vacuoles. Part
of this vacuolation may come from enlargement of the ER (54) but
a good part comes from the accumulation of autolysosomes (15).
Upon the induction of Ras, the HDFs at first proliferate more
often, or in other words they go through a “proliferative burst,”
and then gradually proliferation declines as they approach the “per-
manent” proliferative arrested state of senescence. It is during the
transition phase (from this proliferative phase to the senescent,
proliferative arrest) that autophagy becomes active. Inhibition of
autophagy causes the downregulation of SASP/SMS, such as IL6
and IL8, at the posttranscriptional level. Presumably autophagy
plays a role in delivering the “raw material,” the proteins for deg-
radation, for their reuse in the synthesis of these SASP/SMS fac-
tors, thus supporting the translation of the upregulated SASP/
SMS transcripts.
A similar positive role for autophagy in senescence associated
secretory protein synthesis has recently been observed in senescent
HDFs induced by oxidative stress (55). The authors showed that
H2
O2
induced senescence with activated autophagy, and that the
inhibition of autophagy not only delayed senescence but also
reduced the secretory phenotype. Furthermore, autophagy seems
to affect SASP/SMS outside fibroblasts. Sasaki et al. (56) showed
that autophagy can mediate cellular senescence in primary biliary
cirrhosis (PBC). PBC is a liver-specific autoimmune disease that
eventually leads to the extensive loss of small bile ducts. The authors
had previously demonstrated cellular senescence in the damaged
small bile ducts and suggested that it may be involved in the patho-
genesis of progressive bile duct loss in PBC (57). They went on to
look for autophagy in their bank of biopsied or surgically resected
human liver specimens. Immunohistochemistry staining for
autophagy and lysosomal markers showed large vesicles in PBC,
chronic viral hepatitis, nonalcoholic steatohepatitis and extrahe-
patic biliary obstruction. By far the highest level of these vesicles
4.1. Autophagy in RIS
23. 8 A.R.J. Young et al.
was seen in PBC. The autophagy positive small bile ducts also
stained for markers of senescence. Moving into cultured biliary
epithelial cells (BECs), Sasaki et al. showed that if autophagy was
pharmacologically inhibited then the extent of senescence was
significantly decreased, as was the BEC SASP/SMS.
These studies support the idea that active protein turnover can
promote the en masse production of secretory proteins during senes-
cence. However, it is also possible that autophagy modulates the
senescence phenotype through its ability to maintain the quality of
macromolecules. Gamerdinger et al. identified activated autophagy
during replicative senescence in HDFs as well as in the neurons of
aged rodent brains (14, 58). The authors showed that BAG3, a co-
chaperone, together with other chaperons, Hsp70 and HspB8, pro-
moted the targeting of misfolded and aggregation prone proteins to
autophagosomes. Thus, damaged proteins accumulated during
senescence or aging can be eliminated by autophagy. Of note, rep-
licative senescence is also associated with activation of a secretome
(49), although how autophagy affects SASP/SMS in the context of
replicative senescence remains to be tested. Therefore, autophagy
would seem to have, at least, two distinct levels of influence on
senescence; through its specific role in SASP/SMS and also through
its general activity in the quality control of macromolecules. How
can we reconcile these roles, or does this suggest another contro-
versy, as in the association between autophagy and cell death: do
cells become senescent due to autophagy or despite autophagy? The
potentially mixed roles for autophagy in senescence might reflect
the complex effector networks of senescence mentioned above
(Fig. 1). The collective outcome of the combinatorial consequences
of those effector programs would shift within the range of the senes-
cence phenotype depending on the situation. Therefore, the domi-
nant effect of autophagy might be different between RIS (a highly
dynamic process) and replicative senescence (a slow and adaptive
process). Consistent with this idea, we have recently identified an
additional mechanism occurring during RIS by which autophagy
can support SASP/SMS, which involves a TOR-autophagy spatial
coupling compartment (TASCC) (15).
TASCC—more defined mechanistic insight into the relationship
between autophagy and senescence. Autophagy and protein synthesis
are typically regulated in opposite directions. For example, when
cells are starved, autophagy is activated while protein synthesis is
suppressed, allowing the degradation products (such as amino
acids) to be used as an alternative energy source. This reciprocal
regulation of anabolic and catabolic processes is achieved through
target of rapamycin (TOR), the master regulator of protein synthe-
sis (59). TOR coordinately senses cellular nutrient and energy sta-
tus as well as growth factor signals. TOR then integrates those
signals and “decides” whether sufficient metabolites and energy
are available to synthesize protein (60). TOR is a protein kinase
24. 9
1 Dynamic Interaction Between Autophagy and Senescence
and, if conditions are right, it in turn phosphorylates its substrates,
such as eIF4E binding protein (4EBP) and S6 kinase (S6K), and
facilitates protein synthesis (59).
However, it is possible that the simultaneous activation of pro-
tein degradation and synthesis may allow cells to “regenerate”
themselves very efficiently, and this seems to be occurring during
RIS, where both autophagy and protein synthesis are active (15).
In addition, it has also been shown that Rapamycin, which inhibits
TOR, can divert senescence into quiescence; thus, mTOR is
required for senescence maintenance (61). One potential explana-
tion for the apparently paradoxical simultaneous activation of pro-
tein degradation and protein synthesis may be the spatial separation
of these processes within the cell. By immunostaining, we identified
a unique cellular compartment, the TASCC, in which both mam-
malian TOR (mTOR) and autolysosomes (the end stage of
autophagy) are enriched in the vicinity of the rER-Golgi apparatus
(GA). Aside from growth factor signaling, mTOR can also be acti-
vated by amino acids (the ultimate end product of autophagic pro-
tein degradation). The inhibitory point where mTOR acts on
autophagy is the initial step of autophagosome formation; thus, it
is possible that the physical coupling of these two opposing pro-
cesses causes them to reinforce each other. Indeed, we have shown
that amino acids derived from autolysosomes are required for
mTOR localization to the TASCC, and that early stage autophago-
somes are only detected outside of the TASCC. Our data is highly
consistent with emerging evidence from the Sabatini group, indi-
cating that mTOR is recruited to, and activated at, the lysosomal
surface, where Rheb resides, and that this process is dependent on
Rag GTPases, which also localize to lysosomes (62, 63).
Furthermore, the TASCC is consistently surrounded by the
trans-side of the GA. The rER-GA is the place where both secre-
tory proteins as well as lysosomal and other membrane compo-
nents are synthesized and processed. Thus, the encompassing of
the TASCC by the rER-GA raises an interesting possibility: that
the TASCC, together with the associated rER-GA, may form a
protein “factory” for both the SASP/SMS to be secreted, and lys-
osomal and other membrane proteins (which would, in turn, rein-
force TASCC formation itself). This could be tested using the
technique of fluorescent noncanonical amino acid tagging
(FUNCAT) to specifically visualize newly synthesized proteins in
situ (FUNCAT was developed by the Schuman laboratory (64–
66), and is now licensed to Invitrogen). This system is essentially a
more modern alternative to radioactive, e.g., 35
S, methionine met-
abolic labeling. Pulse–chase experiments with the “tagged” amino
acid revealed that as early as 3 min the nascent proteins are enriched
around the GA, and subsequently move to the GA, and about 2 h
later are detected in the TASCC. In addition, by immunolabelling,
IL6 and IL8 were specifically detected in the TASCC as well as the
25. 10 A.R.J. Young et al.
surrounding GA, suggesting that the same compartment facilitates
the synthesis of proteins for both the degradation machinery and
for SASP/SMS. Consistently, inhibition of Rag GTPase activity
during RIS blocked mTOR enrichment to the TASCC and reduced
the synthesis of SASP/SMS components (15). Although direct
evidence for the regulation of protein synthesis within the TASCC
by mTOR remains to be shown, the TASCC demonstrates addi-
tional complexity in terms of its functional associations between
SASP/SMS, senescence and autophagy.
Importantly, we also found that the TASCC was in close asso-
ciation with the GA in other cellular conditions, such as mac-
rophage-like differentiation in culture, during which cells acutely
produce abundant IL8 (15). In addition, TASCC-like compart-
ments were detected specifically in the glomerular podocytes of
mouse kidneys (15). Podocytes are highly enriched for constitutive
autophagy as well as prominent GA, and produce abundant secre-
tory proteins to support the constant turnover of the glomerular
basement membrane (67, 68). The TASCC may thus be associated
more generally with the acute, abundant production of secretory
proteins, rather than senescence per se. Consistent with this inter-
pretation, replicative senescent HDFs do not show a prominent
TASCC (Masako Narita & Masashi Narita, unpublished data), sug-
gesting that the rate or amount of synthesis of SASP/SMS compo-
nents in this adaptive condition may not require such self-reinforcing
machinery. Although the exact molecular basis for the necessity of
the TASCC in facilitating the production of secretory proteins in
some cases but not in others remains to be elucidated, the TASCC
might provide a mechanism to modulate the downstream effects of
activated autophagy.
Taken together, the studies outlined in this review reinforce the
higher-order network of the different effector programs of senes-
cence and its complexity, in which the same effector might even
produce different outcomes through the different aspects—
dynamic or static—of senescence. Furthermore when it is consid-
ered, as mentioned in Subheading 3, that SASP can affect SAHF,
then the new role for autophagy in the regulation of SASP/SMS
and its spatial association with mTOR implies a functional coop-
eration between nuclear (transcription) and cytoplasmic pheno-
types (protein turnover) toward the expression of a subset of genes.
While we only focus on the limited aspects of gene regulation in
RIS in this review, there are other mechanisms that are also involved
in the phenotype and we expect, in this model system, additional
unexpected interactions between them, as well as new effector
mechanisms to be identified in the future.
5. Conclusions
26. 11
1 Dynamic Interaction Between Autophagy and Senescence
Acknowledgments
Our work is supported by Cancer Research UK.
References
1. Hayflick L (1961) The serial cultivation of
human diploid cell strains. Exp Cell Res 25:
585–621
2. Shay JW, Wright WE (2000) Hayflick, his limit,
and cellular ageing. Nat Rev Mol Cell Biol
1:72–76
3. Harley CB, Futcher AB, Greider CW (1990)
Telomeres shorten during ageing of human
fibroblasts. Nature 345:458–460
4. Strasser A, Cory S, Adams JM (2011)
Deciphering the rules of programmed cell
death to improve therapy of cancer and other
diseases. EMBO J 30:3667–3683
5. Narita M, Nũnez S, Heard E, Narita M, Lin
AW, Hearn SA, Spector DL, Hannon GJ, Lowe
SW (2003) Rb-mediated heterochromatin for-
mation and silencing of E2F target genes dur-
ing cellular senescence. Cell 113:703–716
6. Zhang R, Poustovoitov MV, Ye X, Santos HA,
ChenW,DaganzoSM,ErzbergerJP,Serebriiskii
IG, Canutescu AA, Dunbrack RL, Pehrson JR,
Berger JM, Kaufman PD, Adams PD (2005)
Formation of MacroH2A-containing senes-
cence-associated heterochromatin foci and
senescence driven by ASF1a and HIRA. Dev
Cell 8:19–30
7. Di Micco R, Fumagalli M, Cicalese A, Piccinin
S, Gasparini P, Luise C, Schurra C, Garre M,
Giovanni Nuciforo P, Bensimon A, Maestro R,
Giuseppe Pelicci P, D’adda Di Fagagna F
(2006) Oncogene-induced senescence is a
DNA damage response triggered by DNA
hyper-replication. Nature 444:638–642
8. Bartkova J, Rezaei N, Liontos M, Karakaidos P,
Kletsas D, Issaeva N, Vassiliou L-VF, Kolettas
E, Niforou K, Zoumpourlis VC, Takaoka M,
Nakagawa H, Tort F, Fugger K, Johansson F,
Sehested M, Andersen CL, Dyrskjot L, Ørntoft
T, Lukas J, Kittas C, Helleday T, Halazonetis
TD, Bartek J, Gorgoulis VG (2006) Oncogene-
induced senescence is part of the tumorigenesis
barrier imposed by DNA damage checkpoints.
Nature 444:633–637
9. Mallette FA, Gaumont-Leclerc M-F, Ferbeyre
G (2007) The DNA damage signaling pathway
is a critical mediator of oncogene-induced
senescence. Genes Dev 21:43–48
10. Kuilman T, Michaloglou C, Vredeveld LCW,
Douma S, van Doorn R, Desmet CJ, Aarden LA,
Mooi WJ, Peeper DS (2008) Oncogene-induced
senescence relayed by an interleukin-dependent
inflammatory network. Cell 133:1019–1031
11. Acosta JC, O’Loghlen A, Banito A, Guijarro
MV, Augert A, Raguz S, Fumagalli M, Da
Costa M, Brown C, Popov N, Takatsu Y,
Melamed J, D’adda Di Fagagna F, Bernard D,
Hernando E, Gil J (2008) Chemokine signal-
ing via the CXCR2 receptor reinforces senes-
cence. Cell 133:1006–1018
12. Wajapeyee N, Serra RW, Zhu X, Mahalingam
M, Green MR (2008) Oncogenic BRAF
induces senescence and apoptosis through
pathways mediated by the secreted protein
IGFBP7. Cell 132:363–374
13. Coppé J-P, Patil CK, Rodier F, Sun Y, Muñoz
DP, Goldstein J, Nelson PS, Desprez P-Y,
Campisi J (2008) Senescence-associated secre-
tory phenotypes reveal cell-nonautonomous
functions of oncogenic RAS and the p53 tumor
suppressor. PLoS Biol 6:2853–2868
14. Gamerdinger M, Hajieva P, Kaya AM, Wolfrum
U, Hartl FU, Behl C (2009) Protein quality
control during aging involves recruitment of
the macroautophagy pathway by BAG3. EMBO
J 28:889–901
15. Young ARJ, Narita M, Ferreira M, Kirschner K,
Sadaie M, Darot JFJ, Tavaré S, Arakawa S,
Shimizu S, Watt FM, Narita M (2009)
Autophagy mediates the mitotic senescence
transition. Genes Dev 23:798–803
16. Narita M, Young ARJ, Arakawa S, Samarajiwa
SA, Nakashima T, Yoshida S, Hong S, Berry
LS, Reichelt S, Ferreira M, Tavaré S, Inoki K,
Shimizu S, Narita M (2011) Spatial coupling of
mTOR and autophagy augments secretory
phenotypes. Science 332(6032):966–970
17. Romanov SR, Kozakiewicz BK, Holst CR,
Stampfer MR, Haupt LM, Tlsty TD (2001)
Normal human mammary epithelial cells spon-
taneously escape senescence and acquire
genomic changes. Nature 409:633–637
18. Lin AW, Lowe SW (2001) Oncogenic ras acti-
vates the ARF-p53 pathway to suppress epithe-
lial cell transformation. Proc Natl Acad Sci U S
A 98:5025–5030
19. Shay JW, Roninson IB (2004) Hallmarks of
senescence in carcinogenesis and cancer ther-
apy. Oncogene 23:2919–2933
20. Narita M, Lowe SW (2005) Senescence comes
of age. Nat Med 11:920–922
27. 12 A.R.J. Young et al.
21. Serrano M, Lin AW, McCurrach ME, Beach D,
Lowe SW (1997) Oncogenic ras provokes pre-
mature cell senescence associated with accumu-
lation of p53 and p16INK4a. Cell 88:593–602
22. Lin AW, Barradas M, Stone JC, Van Aelst L,
Serrano M, Lowe SW (1998) Premature senes-
cence involving p53 and p16 is activated in
response to constitutive MEK/MAPK mito-
genic signaling. Genes Dev 12:3008–3019
23. Lazzerini Denchi E, Attwooll C, Pasini D,
Helin K (2005) Deregulated E2F activity
induces hyperplasia and senescence-like fea-
tures in the mouse pituitary gland. Mol Cell
Biol 25:2660–2672
24. Braig M, Lee S, Loddenkemper C, Rudolph C,
Peters AH, Schlegelberger B, Stein H, Dörken
B, Jenuwein T, Schmitt CA (2005) Oncogene-
induced senescence as an initial barrier in lym-
phoma development. Nature 436:660–665
25. Chen Z, Trotman LC, Shaffer D, Lin H-K,
Dotan ZA, Niki M, Koutcher JA, Scher HI,
Ludwig T, Gerald W, Cordon-Cardo C, Paolo
Pandolfi P (2005) Crucial role of p53-depen-
dent cellular senescence in suppression of Pten-
deficient tumorigenesis. Nature 436:725–730
26. Collado M, Gil J, Efeyan A, Guerra C,
Schuhmacher AJ, Barradas M, Benguría A,
Zaballos A, Flores JM, Barbacid M, Beach D,
Serrano M (2005) Tumour biology: senescence
in premalignant tumours. Nature 436:642
27. Michaloglou C, Vredeveld LCW, Soengas MS,
Denoyelle C, Kuilman T, Van Der Horst
CMAM, Majoor DM, Shay JW, Mooi WJ,
Peeper DS (2005) BRAFE600-associated
senescence-like cell cycle arrest of human naevi.
Nature 436:720–724
28. DeNicola GM, Karreth FA, Humpton TJ,
Gopinathan A, Wei C, Frese K, Mangal D, Yu
KH, Yeo CJ, Calhoun ES, Scrimieri F, Winter
JM, Hruban RH, Iacobuzio-Donahue C, Kern
SE, Blair IA, Tuveson DA (2011) Oncogene-
induced Nrf2 transcription promotes ROS
detoxification and tumorigenesis. Nature 475:
106–109
29. Schmitt CA, Fridman JS, Yang M, Lee S,
Baranov E, Hoffman RM, Lowe SW (2002) A
senescence program controlled by p53 and
p16INK4a contributes to the outcome of can-
cer therapy. Cell 109:335–346
30. te Poele RH, Okorokov AL, Jardine L,
Cummings J, Joel SP (2002) DNA damage is
able to induce senescence in tumor cells in vitro
and in vivo. Cancer Res 62:1876–1883
31. Sharpless NE, Depinho RA (2007) How stem
cells age and why this makes us grow old. Nat
Rev Mol Cell Biol 8:703–713
32. Hong H, Takahashi K, Ichisaka T, Aoi T,
Kanagawa O, Nakagawa M, Okita K, Yamanaka S
(2009) Suppression of induced pluripotent
stem cell generation by the p53-p21 pathway.
Nature 460:1132–1135
33. Kawamura T, Suzuki J, Wang YV, Menendez S,
Morera LB, Raya A, Wahl GM, Belmonte JCI
(2009) Linking the p53 tumour suppressor
pathway to somatic cell reprogramming. Nature
460:1140–1144
34. Marión RM, Strati K, Li H, Murga M, Blanco
R, Ortega S, Fernandez-Capetillo O, Serrano
M, Blasco MA (2009) A p53-mediated DNA
damage response limits reprogramming to
ensure iPS cell genomic integrity. Nature 460:
1149–1153
35. Li H, Collado M, Villasante A, Strati K, Ortega
S, Cañamero M, Blasco MA, Serrano M (2009)
The Ink4/Arf locus is a barrier for iPS cell
reprogramming. Nature 460:1136–1139
36. Krizhanovsky V, Lowe SW (2009) Stem cells:
the promises and perils of p53. Nature
460:1085–1086
37. Banito A, Rashid ST, Acosta JC, Li S, Pereira
CF, Geti I, Pinho S, Silva JC, Azuara V, Walsh
M, Vallier L, Gil J (2009) Senescence impairs
successful reprogramming to pluripotent stem
cells. Genes Dev 23:2134–2139
38. Seshadri T, Campisi J (1989) Growth-factor-
inducible gene expression in senescent human
fibroblasts. Exp Gerontol 24:515–522
39. Seshadri T, Campisi J (1990) Repression of
c-fos transcription and an altered genetic pro-
gram in senescent human fibroblasts. Science
247:205–209
40. Beauséjour CM, Krtolica A, Galimi F, Narita
M, Lowe SW, Yaswen P, Campisi J (2003)
Reversal of human cellular senescence: roles of
the p53 and p16 pathways. EMBO J 22:
4212–4222
41. D’adda Di Fagagna F (2008) Living on a break:
cellular senescence as a DNA-damage response.
Nat Rev Cancer 8:512–522
42. Narita M (2007) Cellular senescence and chro-
matin organisation. Br J Cancer 96:686–691
43. Adams PD (2007) Remodeling of chromatin
structure in senescent cells and its potential
impact on tumor suppression and aging. Gene
397:84–93
44. Narita M, Narita M, Krizhanovsky V, Nuñez S,
Chicas A, Hearn SA, Myers MP, Lowe SW
(2006) A novel role for high-mobility group a
proteins in cellular senescence and heterochro-
matin formation. Cell 126:503–514
45. Shelton DN, Chang E, Whittier PS, Choi D,
Funk WD (1999) Microarray analysis of repli-
cative senescence. Curr Biol 9:939–945
46. Young ARJ, Narita M (2009) SASP reflects
senescence. EMBO Rep 10:228–230
28. 13
1 Dynamic Interaction Between Autophagy and Senescence
47. Krtolica A, Parrinello S, Lockett S, Desprez PY,
Campisi J (2001) Senescent fibroblasts pro-
mote epithelial cell growth and tumorigenesis:
a link between cancer and aging. Proc Natl
Acad Sci U S A 98:12072–12077
48. Campisi J, D’adda Di Fagagna F (2007)
Cellular senescence: when bad things happen to
good cells. Nat Rev Mol Cell Biol 8:729–740
49. Rodier F, Coppé J-P, Patil CK, Hoeijmakers
WAM, Muñoz DP, Raza SR, Freund A,
Campeau E, Davalos AR, Campisi J (2009)
Persistent DNA damage signalling triggers
senescence-associated inflammatory cytokine
secretion. Nature 11:973–979
50. Kraft C, Peter M, Hofmann K (2010) Selective
autophagy: ubiquitin-mediated recognition
and beyond. Nature 12:836–841
51. Holtzman E (1989) Lysosomes. Springer, New
York, p 439
52. Mancini GM, Havelaar AC, Verheijen FW
(2000) Lysosomal transport disorders. J Inherit
Metab Dis 23:278–292
53. Ryan KM (2011) p53 and autophagy in cancer:
guardian of the genome meets guardian of the
proteome. Eur J Cancer 47:44–50
54. Denoyelle C, Abou-Rjaily G, Bezrookove V,
Verhaegen M, Johnson TM, Fullen DR, Pointer
JN, Gruber SB, Su LD, Nikiforov MA, Kaufman
RJ, Bastian BC, Soengas MS (2006) Anti-
oncogenic role of the endoplasmic reticulum
differentially activated by mutations in the
MAPK pathway. Nat Cell Biol 8:1053–1063
55. Luo Y, Zou P, Zou J, Wang J, Zhou D, Liu L
(2011) Autophagy regulates ROS-induced cel-
lular senescence via p21 in a p38 MAPKα
dependent manner. Exp Gerontol 46:860–867
56. Sasaki M, Miyakoshi M, Sato Y, Nakanuma Y
(2010) Autophagy mediates the process of cel-
lular senescence characterizing bile duct dam-
ages in primary biliary cirrhosis. Lab Invest
90:835–843
57. Sasaki M, Ikeda H, Haga H, Manabe T,
Nakanuma Y (2005) Frequent cellular senes-
cence in small bile ducts in primary biliary cir-
rhosis: a possible role in bile duct loss. J Pathol
205:451–459
58. Gamerdinger M, Kaya AM, Wolfrum U,
Clement AM, Behl C (2011) BAG3 mediates
chaperone-based aggresome-targeting and
selective autophagy of misfolded proteins.
EMBO Rep 12:149–156
59. Wullschleger S, Loewith R, Hall MN (2006)
TOR signaling in growth and metabolism. Cell
124:471–484
60. Sengupta S, Peterson TR, Sabatini DM (2010)
Regulation of the mTOR complex 1 pathway
by nutrients, growth factors, and stress. Mol
Cell 40:310–322
61. Korotchkina LG, Leontieva OV, Bukreeva EI,
Demidenko ZN, Gudkov AV, Blagosklonny
MV (2010) The choice between p53-induced
senescence and quiescence is determined in
part by the mTOR pathway. Aging (Albany
NY) 2:344–352
62. Sancak Y, Peterson TR, Shaul YD, Lindquist RA,
Thoreen CC, Bar-Peled L, Sabatini DM (2008)
The Rag GTPases bind raptor and mediate
amino acid signaling to mTORC1. Science
320:1496–1501
63. Sancak Y, Bar-Peled L, Zoncu R, Markhard AL,
Nada S, Sabatini DM (2010) Ragulator-Rag
complex targets mTORC1 to the lysosomal
surface and is necessary for its activation by
amino acids. Cell 141:290–303
64. Dieterich DC, Link AJ, Graumann J, Tirrell DA,
Schuman EM (2006) Selective identification of
newly synthesized proteins in mammalian cells
using bioorthogonal noncanonical amino acid
tagging (BONCAT). Proc Natl Acad Sci U S A
103:9482–9487
65. Dieterich DC, Lee JJ, Link AJ, Graumann J,
Tirrell DA, Schuman EM (2007) Labeling,
detection and identification of newly synthe-
sized proteomes with bioorthogonal non-
canonical amino-acid tagging. Nat Protoc 2:
532–540
66. Dieterich DC, Hodas JJL, Gouzer G, Shadrin IY,
Ngo JT, Triller A, Tirrell DA, Schuman EM
(2010) In situ visualization and dynamics of
newly synthesized proteins in rat hippocampal
neurons. Nat Neurosci 13:897–905
67. Hartleben B, Gödel M, Meyer-Schwesinger C,
Liu S, Ulrich T, Köbler S, Wiech T, Grahammer
F, Arnold SJ, Lindenmeyer MT, Cohen CD,
Pavenstädt H, Kerjaschki D, Mizushima N,
Shaw AS, Walz G, Huber TB (2010) Autophagy
influences glomerular disease susceptibility and
maintains podocyte homeostasis in aging mice.
J Clin Invest 120:1084–1096
68. Pavenstädt H, Kriz W, Kretzler M (2003) Cell
biology of the glomerular podocyte. Physiol
Rev 83:253–307
30. 16 V. Dulic
PDK1 3-Phosphoinositide-dependent kinase 1
PI3K Phosphoinositide 3-kinase
PIKK Phosphoinositide 3-kinase (PI3K)-related protein serine/threonine kinase family
PML Promyelocytic leukemia protein
PTEN Phosphatase and tensin homologue
pRb Retinoblastoma protein
RHEB Ras homologue enriched in brain
SAHF Senescence-associated heterochromatin foci
SASP Senescence-associated secretory phenotype
SGK Serum- and glucocorticoid-regulated kinase
TSC1/2 Tuberous sclerosis 1,2 GTPase
TGFb Transforming growth factor b
Cellular senescence was originally described by Leonard Hayflick,
who showed that, after a finite number of population doublings
(mitotic clock), primary human cells irreversibly cease to prolifer-
ate in vitro (1). This so-called Hayflick limit was later shown to be
provoked, at least in part, by telomere erosion (2), the gradual
loss of DNA at the ends of chromosomes, a consequence of the
“replication problem” predicted by Olovnikov in the early 1970—
hence “replicative senescence” (3). It has been therefore proposed
that telomere erosion could activate a DNA damage response
(DDR) and cause G1 cell cycle arrest similar to the one elicited by
ionizing radiation (4). Moreover, following an excellent intuition,
Hayflick associated cellular senescence with both cancer and ageing—
with cancer, because cells must acquire certain characteristics of
tumor cells to escape senescence, and with ageing, because accu-
mulation of senescent cells could contribute to the global deterio-
ration of an organism. We now know that these original hypotheses
were fundamentally right, and since then, senescence, from a “tis-
sue culture artifact,” became an ever-expanding field of intense
research largely due to its tumor suppression potential and contri-
bution to age-related pathologies. In addition to telomere dys-
function, senescence can be induced by a large variety of stress
stimuli, including strong mitogenic signals generated by hyper-
activation of certain oncogenes (Ras, Raf, Myc), irreparable DNA
damage, or oxidative stress. This invariably inhibits cyclin-
dependent kinases (CDK), key cell division regulators, leading to
the cell cycle arrest that requires activities of two major tumor
suppressors, p53 and pRb (Fig. 1).
As a number of excellent recent reviews extensively covered
this topic (5–7), we shall here briefly outline the aspects of cellular
senescence and the pathways implicated in its implementation that
are relevant for this chapter.
1. Introduction
31. 17
2 mTOR, Metabolism and Cell Senescence
Senescence is a viable and metabolically active state, which is char-
acterized by a virtually permanent (irreversible) cessation of cell
division. It is associated with dramatic changes in cell morphology
(large flat cells), metabolism, gene expression and secretion patterns
(senescence-associated secretory phenotype or SASP), originally
described by Campisi and colleagues (8). Senescence has recently
been shown to play a crucial role in age-related pathologies associ-
ated with accumulation of senescent cells (9, 10). Both in vitro and
in vivo, senescent cells can be detected by virtue of β-galactosidase
activity (β-gal) (11), a lysosomal enzyme, which probably does not
play a direct role in senescence (12), but serves as a reliable biomarker
2. How is
Permanent
Cell Cycle Arrest
Achieved?
Fig. 1. Molecular pathways leading to senescence-associated irreversible cell cycle arrest.
Pro-senescence stimuli activate DNA damage response pathway (DDR) leading to perma-
nent inactivation of cyclin-dependent kinases (CDK) that control DNA replication and mito-
sis (K1/2). In addition, together with Cdk4/6 (K4/6), they keep inactive tumor suppressor
pRb (and related pocket proteins p107 and p130). Genotoxic stress and activated onco-
genes induce expression of CDK inhibitors p21Waf1/Cip1
(p21) and p16Ink4A
(p16) that block cell
cycle progression and pRb inactivation. Active pRb blocks expression of genes controlling
cell cycle (in part by sequestering E2F family of transcription factors) and contributes to
chromatin reorganization in the form of SAHF or PML nuclear bodies.The pathways leading
to induction of p16 are not entirely elucidated but, in the case of OIS (oncogene-induced
senescence), it has been shown that demethylase JMJD3 blocks repression of Ink4A locus
by PcG protein Bmi1.Arf (p19ARF
), another product of this locus, plays a major role in senes-
cence in mice, while p16 is more important in humans. Figure adapted from ref. 119.
32. 18 V. Dulic
(see Chapter 4 and Fig. 2). In addition, senescent cells often exhibit
dramatic rearrangement of chromatin structure, in the forms of
PML (promyelocytic leukemia protein) nuclear bodies (13, 14) and
senescence-associated heterochromatin foci (SAHF) (15). These
heterochromatin structures, whose formation requires active pRb,
are thought to play an important role in conferring irreversibility of
the cell cycle arrest by repressing the genes controlling cell prolif-
eration (Figs. 1 and 2). However, SAHFs are not observed in all cell
types and in all forms of senescence (15), and recent work associates
SAHF formation with oncogene induced senescence and repression
of DNA damage signaling (16).
The pro-senescence stimuli all converge on one major pathway
to permanently arrest the cell cycle (Fig. 1). The senescence pro-
gram is initiated by inactivation of CDK (17), the key cell cycle
regulators. CDKs play two distinct roles. First, they control the
onset and progression of DNA replication and mitosis, thus
enabling and orchestrating the cell cycle. Second, CDKs, particu-
larly Cdk4/6 associated with D-type cyclins, phosphorylate, thus
keeping inactive, the retinoblastoma (pRb) tumor suppressor and
related “pocket-proteins” (p107, p130). This is essential for cell
cycle progression, since active (hypo-phosphorylated) pocket pro-
teins drive cell cycle exit by sequestering E2F family transcription
factors and by repressing the genes required for cell division (18).
Fig. 2. Senescence versus quiescence. Quiescence (or G0) is reversible and hypotrophic cell cycle exit that occurs in G1
prior to restriction (R) point. It is mediated preferentially by p27-dependent CDK inactivation and cyclin D1 downregulation.
Some results suggest that genes expressed in quiescent cells can block senescence (HES1). By contrast, senescence is
irreversible and hypertrophic cell cycle exit that takes place either in G1 or G2, depending on the activated checkpoint. Cell
cycle arrest is mediated by p16/p21-dependent CDK inactivation (Fig. 1) and senescent cells contain high levels of G1
cyclins (cyclin D and cyclin E) associated with inactive CDK.While the cytoplasm could be frequently stained for β-galacto-
sidase (see Chapters 4, 8 and 9), senescent nuclei are often large and sometimes binuclear (due to endoreplication or
defective cytokinesis) and could contain heterochromatin in the forms of SAHF (see Chapter 12) or PML bodies. Both events
require active pRb family pocket proteins but in senescence only pRb is essential (ref. 19). GF growth factors.
33. 19
2 mTOR, Metabolism and Cell Senescence
Despite often-overlapping functions between different members of
the pocket protein family, pRb was shown to play a unique and
nonredundant role in senescence by repressing E2F target genes
involved in DNA replication (19) (Fig. 1). Recent data suggest
that this process requires a recruitment of promoter-bound pRb
and E2Fs to PML nuclear bodies (14). More recently, p130/E2F4
complex was also implicated in senescence by repressing, via PML,
T-box protein 2 (TBX2) frequently overexpressed in cancer (20).
The presence of diverse genotoxic stresses that induce senescence
activates a DDR network, controlled by ATM/ATR kinases, which
blocks CDK activation via CDC25 family phosphatases, thereby
temporarily halting cell cycle progression (Fig. 1). While not
required for this transient arrest, p53 and its transcriptional target
p21Waf1/Cip1/Sdi1
(p21), a CDK inhibitor (CKI), are essential parts of
irreversible cell cycle arrest by the senescence program (21, 22).
Permanent CDK inactivation by p21 stably blocks DNA synthesis
and mitotic entry and, via activation of pRb, drives exit from the
cell cycle (19, 23) (Fig. 1). Another CKI, p16Ink4A
(p16), which
targets specifically pRb kinases Cdk4/6, also plays an important
role in senescence and, along with β-gal, it is often used as a bio-
marker both in vitro and in vivo (9, 24). However, unlike p21, p16
does not seem to be involved in the cell cycle arrest associated with
replicative senescence (21) and its late induction, which occurs
after p21-mediated cell cycle arrest, might promote a stabilization
of senescent state (25, 26). Nevertheless, p16 is a key regulator of
oncogene-induced senescence (OIS) (27) and recent work in a
mouse model strongly implicated this inhibitor in acquisition of
age-related pathologies (9). Although p16 is widely considered as
an essential part of the senescence program, the pathways regulat-
ing its induction in response to senescence-promoting stimuli are
still not entirely elucidated. It has been suggested that, in the case
of OIS, activation of histone demethylase JMJD3 suppresses the
repression of the INK4A-ARF locus by Polycomb group proteins
(PcG), Bmi1; (5). Another protein encoded by this locus is tumor
suppressor p19ARF
(ARF) that plays a prominent role in senescence
in mice but not in humans (27). ARF stabilizes p53 by inhibiting
MDM2 and, via its effector p21 blocks cell cycle. Thus, ultimate
targets of both products of the INK4A-ARF locus are CDKs that
inactivate pRb, and their dysregulation invariably compromises
genome integrity (Fig. 1).
What distinguishes senescence from another non-proliferating
state, quiescence, also referred to as G0 or G0/G1 phase (28)? In
contrast to senescence, quiescence is a reversible cell cycle arrest,
3. Quiescence
Versus
Senescence
34. 20 V. Dulic
which is induced by the absence of mitogens or growth factors,
nutrient starvation, or increasing cell density (confluence) (Fig. 2).
Typical examples of quiescent cells are lymphocytes, whose activation
is part of the immune response, adult stem cells, or dermal
fibroblasts, which actively participate in wound healing (29). In
general, quiescence is characterized by low metabolism and protein
synthesis, lack of cellular growth, and, the absence of global het-
erochromatin structures such as PML bodies (30) or SAHF (15).
The latter feature is probably responsible for the reversible nature
of quiescence. However, much against the prevailing view, recent
results showed that fibroblasts rendered quiescent by contact inhi-
bition exhibit comparable metabolic activity to actively proliferat-
ing cells. In addition to promote recycling of damaged
macromolecules via autophagy, high metabolic activity might serve
for biosynthesis and secretion of extracellular matrix proteins (31).
Importantly, while downregulating expression of genes involved in
cell division, quiescent cells upregulate genes, such as HES1, that
inhibit senescence, differentiation and apoptosis (32, 33). Unlike
senescence, which can occur either in the G1 or G2 phase of the
cell cycle (34), depending on when the damage is detected and the
efficiency of the checkpoints (23, 35), quiescence essentially takes
place in G1, prior to the restriction (R) checkpoint (36). A teleo-
logical explanation for this observation could be given by the uni-
directionality of the cell cycle—G0 state always precedes DNA
replication (S phase). Therefore, if cells became quiescent in the
G2 phase, cell cycle entry upon stimulation would result in genome
reduplication giving rise to undesirable tetraploidy.
Like senescence, quiescence is characterized by CDK inactivation,
the absence of S-phase- and Mitosis-promoting cyclins and the pres-
ence of hypo-phosphorylated pocket proteins. However, unlike
senescence, where CDKs are inhibited by p21 or/and p16, the
major CKI involved in quiescent arrest is p27Kip1
(p27) (37–39).
Unlike p21, p27 induction is independent of the p53 pathway (39)
and its levels/activity is primarily regulated by translation, phos-
phorylation, and Skp2-mediated degradation (40). Although p53
and p21 were also implicated in the cell cycle arrest following
growth factor removal (41, 42), p53 integrity does not seem to be
essential for quiescence (43) (Fig. 2). However, a redundancy of
p27 with p21 might explain why contact inhibition is not impaired
in p27−/−
cells (44). Interestingly, the expression of D-type
cyclins, regulatory subunits of Cdk4/6 kinases and key signal inte-
grators, strikingly differs between quiescence and senescence (Fig. 2).
Downregulation of cyclin Ds is a hallmark of quiescence and their
rapid mitogen-dependent induction is invariably associated with
cell cycle entry (42, 45, 46). In sharp contrast, D cyclins are stabi-
lized (cyclin D1) and even overexpressed (cyclin D2) in senescent
cells (17, 35, 43, 47). In conjunction with the large-cell phenotype
that is observed in senescent cells, these results suggest that the
35. 21
2 mTOR, Metabolism and Cell Senescence
mitogenic pathways involved in cell growth are active in senescent
cells (see below) and might even have a positive role in senescence
regulation. Indeed, several recent publications have connected the
“hypertrophic” phenotype of senescent cells with activity of
mTOR, a master cell growth regulator (48), although this topic is
still controversial (49).
Originally identified in budding yeast mutants conferring resis-
tance to a potent antifungal metabolite (50), the target of rapamy-
cin (TOR) is at the core of a vast signaling pathway regulating cell
growth and metabolism in virtually all eukaryotes. The complexity
of this pathway, which couples energy and nutrient abundance to
the execution of cellular growth and division, relies on the fact that
TOR simultaneously senses energy (ATP state), nutrients, stress, as
well as growth factors (51, 52). In addition to its growth-related
functions, the TOR pathway is also wired to the regulators of cell
cycle machinery such as G1 cyclins (D-type and E-type) control-
ling G1/S-phase progression (see below and Fig. 3). It is therefore
not surprising that TOR is essential for many developmental and
physiological processes (53) while deregulation in its signaling has
been implicated in a wide variety of diseases (52). mTOR (“m”
stands for mammalian or, more recently, mechanistic), which
belongs to the phosphoinositide 3-kinase (PI3K)-related protein
serine/threonine kinase family (PIKK), forms two distinct multi-
protein complexes, having distinctive physical structures and
functions: mTOR complex 1 (mTORC1), which is sensitive to
rapamycin owing to association with the peptidyl-prolyl cis/trans
isomerase FKBP12 (50), and mTORC2, which is not sensitive to
rapamycin in most cases. Their assembly, substrate specificity, and
regulation are defined by specific accessory proteins: Raptor (regu-
latory associated protein of mTOR) being the most prominent for
mTORC1, and Rictor (rapamycin-insensitive companion of
mTOR), which is specific for mTORC2. Since the regulatory
inputs and the cellular actions of mTOR are far too complex for
the scope of this chapter (and are reviewed in ref. 53, 54), I outline
only some “essentials” focusing mainly on mTORC1, the better
characterized of the two.
mTORC1, which is activated by nutrients, growth factors, and
cellular energy status, regulates temporal aspects of cellular growth
including protein synthesis, ribosome biogenesis, lipid synthesis,
nutrient import, and autophagy. Best-known substrates of
mTORC1 are p70 ribosomal protein S6 kinases (S6K1 and S6K2)
and the eIF4E binding proteins (4E-BP1 and 4E-BP2) by which it
controls protein synthesis. The positive and negative control of
4. The mTOR
Pathway
Essentials
36. 22 V. Dulic
mTORC1 in response to intracellular and extracellular stimuli is
mediated by the phosphorylation and subsequent inhibition of
Tuberous sclerosis 1 (TSC1, hamartin)/TSC2 (tuberin) complex
(Fig. 3). TSC2 subunit, a hub for a variety of cues impinging on
mTORC1, serves as GTPase activating protein (GAP), which itself
inhibits the small GTPase Rheb, a direct mTORC1 activator (51,
53, 55). TSC2 activity is regulated by several kinases: negatively, by
Akt and ERK1/2 (activate mTORC1) and positively, by AMPK
and GSK3β (inhibit mTORC1). Importantly, the Wnt pathway
also regulates TSC1/TSC2, via inhibition of GSK3β (53) (see
Fig. 4). mTORC2 is thought to regulate mainly cytoskeleton orga-
nization and cell survival by phosphorylating of AGC kinase family
members, including Akt, SGK1, and PKC (52, 53) (Fig. 3).
Although little is known about upstream regulators of mTORC2
(56), recent reports showing that its activation requires association
with ribosomes suggest that it is active only in growing cells (57).
Fig. 3. Interactions between mTOR signaling, p53 and cell cycle regulators.While mTORC1 controls cell growth/division and
metabolism,TORC2 mainly controls cell shape and activates Akt.TSC1/TSC2 complex plays a pivotal role as a hub for both
positive and negative cues that control mTORC1 activity via signaling kinases (see ref. 53). mTOR controls cell division by
regulating synthesis of cyclin D and activity/localization of CDK inhibitor p27. While in the nucleus, p27 inhibits CDKs (cD-
K4/K6 and cE-K2) thereby blocking cell cycle progression (like in quiescence). Growth factors stimulate mTOR and ERK1/2
that, via Akt, RSK and SGK, mediate p27 phosphorylation leading to its cytoplasmic sequestration. Cytoplasmic p27 pro-
motes proliferation by stabilizing cyclin D-Cdk4/6 assembly and cell motility by inhibiting RhoA/ROCK (see ref. 71). In
response to DNA damage or other stresses, p53 negatively regulates mTORC1 by activating AMPK and PTEN and inducing
TSC2. Constitutive activation of mTOR (asterisk) could induce p53 translation (see ref. 86). Figure adapted from ref. 119.
GF growth factors (insulin, IGF1); Sesn, sestrin.
37. 23
2 mTOR, Metabolism and Cell Senescence
As eluded above, mTOR pathway is also implicated in cell division,
in part by controlling the synthesis of D-type cyclins (58, 59),
major regulators of G1-S phase progression, whose induction in
response to growth factors is regulated by the Ras-ERK1/2 path-
way (60, 61) (Fig. 3). Recent data show that mTORC1 controls
cell proliferation, but not growth, via its targets 4E-BPs, by stimu-
lating the translation of proteins involved in cell cycle progression,
including cyclin D3 (62). In addition, in the fission yeast
model, TOR was also shown to control mitotic entry in response
to nutrients. In this system, both rapamycin (by inhibiting TOR)
and poor nitrogen sources stimulate the stress MAPK pathway
(Spc1/Sty1), which, by activating Polo kinase and Cdc2 (Cdk1
ortholog, the main mitotic regulator) accelerated mitosis with
reduced cell size as consequence (63). While no equivalent path-
way was yet reported in other models, these observations are con-
sistent with earlier results that linked mTOR signaling with cell-size
control (64). Nevertheless, despite general downregulation of
5. mTOR
and Cell Cycle
Fig. 4. Possible roles of p53 and mTOR in senescence. Genotoxic stress (via p53–p21) and activated oncogenes (via p16)
induce senescence by inactivating CDK and activating pRb (see Fig. 1). Classically, p53 and activated oncogenes induce
autophagy by inhibiting mTOR pathway (grey box).Autophagy is implicated in senescence by stimulating specific secretory
phenotype (SASP) that also promotes senescence. However, recent data suggest that mTOR might positively regulate
senescence (grey dotted lines) as in the case of persistent Wnt1 signaling or PTEN knockdown. Pro-senescence mTOR
activity could be also activated via oncogenic stress—by inducing autophagy, or via low p53 levels (grey dotted cross)—by
promoting cell growth and D-type cyclin synthesis. Hypertrophy of senescent cells could be explained by deregulated cell
signaling (not responding to extracellular cues) resulting in permanent mTOR activation. This is in agreement with results
showing that in senescent cells cell growth and cell division are uncoupled events. For more details regarding mTOR regu-
lation, see Fig. 3 and ref. 53. Figure adapted from ref. 119. GF growth factors.
38. 24 V. Dulic
protein translation in mitosis (65), several recent papers suggest
that mTORC1 might also control protein synthesis in mitosis by
regulating expression of human Cdk1, which phosphorylates
eukaryotic elongation factor 2 (eEF2); (66). Moreover, it seems
that, at G2/M transition, Cdk1 might also regulate mTORC1
activity by phosphorylating Raptor (67, 68).
The mTOR pathway also positively controls cell cycle by
regulating the activity and subcellular localization of the CKI
p27, negative regulator of G1/S progression (Fig. 3). In the
presence of growth factors, Akt (activated by mTORC2), RSK
(activated by ERK1/2), and SKG (activated by mTORC1,
mTORC2 and Pdk1) phosphorylate p27, resulting in its cyto-
plasmic retention (69–71). This not only prevents p27 to exert
its nuclear role as CDK inhibitor, by inactivating CDKs control-
ling DNA replication (as cyclin E-Cdk2), but also promote cell
proliferation by stabilizing assembly of cyclin D-CDK4/6 com-
plexes and increasing cell motility, via inhibition of RhoA signal-
ing (71) (Fig. 3). In addition, mTORC2 regulates cell
proliferation via AKT-mediated inhibition of forkhead box pro-
tein O (FOXO) transcription factors playing a key role in pro-
moting apoptosis (72). Importantly, the FOXO family members
were also shown to block cell cycle by inducing p27 (73) or
repressing D-type cyclins (74).
In the light of increasing evidence that altered metabolism is not
only a common feature of many cancer cells but that it can also
greatly contribute to malignant transformation (75), the discov-
ery that p53 also regulates metabolic pathways and interacts with
mTOR/AKT pathways is not that surprising (reviewed in refs.
76, 77) (Fig. 3 and 4). Indeed, it has been shown that, in response
to diverse stress signals, activation of p53 directly or indirectly
inhibits mTOR activity, thus regulating its downstream targets,
including those involved in the activation of autophagy, a recently
described tumor suppression mechanism that is involved in senes-
cence (54, 78) (see also Chapter 3). The role of autophagy in
senescence is discussed in more detail in the chapter by M. Narita
(see Chapter 1). In addition to its housekeeping role in the main-
tenance of energy homeostasis, autophagy, which is negatively
regulated by mTOR, is also induced by a variety of stress stimuli,
such as nutrient depletion (when mTOR is inhibited), playing a
vital role in preserving cellular viability through the degradation
of cellular proteins and organelles (53, 79).
6. TOR, p53, and
Genotoxic Stress
39. 25
2 mTOR, Metabolism and Cell Senescence
The mechanism by which p53 negatively regulates mTOR
involves, in part, the activation of AMPK and requires its target
TSC1/TSC2 complex (Fig. 3), both of which respond to energy
deprivation in cells (53, 80). In response to genotoxic stress caused
by DNA damage, p53 inhibits mTORC1 pathway, by phosphory-
lating Sestrin 1 and 2 that activate AMPK (81), as well as the
IGF-1/Akt pathway, by inducing transcription of PTEN (82)
(Fig. 3). In addition, p53 can repress mTORC1 by inducing
directly TSC2 (83) or AMPK, which suppresses translation by acti-
vating TSC2 (80) or by directly inhibiting mTORC1 (54, 84).
However, it has been reported that AMPK might also control the
p53 activity, like in the case of glucose starvation, which induces
the transient AMPK–mediated phosphorylation and activation of
p53 leading to reversible cell cycle arrest (85, 86). There are sev-
eral examples showing that conversely, mTOR can also control
p53. Upon glucose removal, mTOR negatively regulates p53 by
activating PP2A (87). However, some results suggest that deregu-
lated (constitutive) mTOR activity might, in response to nutrient
or genomic stress, induce p53 translation, which in conjunction
with activation by AMPK, induces apoptosis (88). This implies
that, at least in certain biological contexts, mTOR downregula-
tion/inactivation is required to prevent cell death caused by stress
stimuli. Thus, p53 and mTOR signaling networks can cross-talk
and coordinately regulate cell growth, proliferation and death.
The mTOR pathway has been shown to have opposing roles on
cellular senescence. On the one hand, by inhibiting autophagy (78,
86, 89), mTOR is considered as a negative regulator of senescence
(Fig. 4), which is in agreement with its widely accepted negative
role in organismal ageing (90). Moreover, as mentioned above,
mTOR is inhibited by p53, a bona fide pro-senescence positive reg-
ulator (Figs. 3 and 4). On the other hand, the hypertrophic pheno-
type of senescent cells (Fig. 2) and increased production of secretory
proteins (SASP; Fig. 4), which requires high metabolic activity, are
in apparent contradiction with cellular shrinkage that is usually
associated with autophagy and mTOR inhibition (as in quiescence).
Indeed, in the case of oncogene or DNA damage-induced senes-
cence, the onset of both senescence and autophagy correlates with
inhibition of mTORC1 and mTORC2 activity (91). This might
suggest a negative feedback between mTOR and senescence. These
findings are also in agreement with earlier observations that acti-
vation of p53 by genotoxic stress inhibits mTOR pathway by acti-
vating both AMPK and PTEN (Fig. 3) (81, 86). Likewise, persistent
7. mTOR’ s Role
in Senescence:
Repressor
or Inducer?
40. 26 V. Dulic
AMPK activation leads to senescence in mouse embryo fibroblasts
(MEF) in the presence of functional p53 (85), which is consistent
with earlier observation that AMP levels strongly increase in senes-
cent cells (92).
However, it is also possible that mTOR activation via different
stimuli could be involved in the induction of senescence. This pro-
senescence role of mTOR was first uncovered by manipulation of
its upstream regulators or downstream targets. Pandolfi and col-
leagues found that overexpression of an mTOR target eIF-4E
induced senescence both in vitro and in vivo (93). More recently,
activation of the PI3K/Akt and mTOR pathways by PTEN knock-
down has been shown to induce senescence by promoting accu-
mulation of p53 and p21 (94). Similarly, in an elegant in vivo skin
model, Gutkind and colleagues showed that continuous mTOR
activation induced by persistent Wnt1 signaling (via inhibiting
GSK3; (95)) promoted epithelial stem cell exhaustion by provok-
ing senescence (96). Importantly, senescence and hair loss were
abolished by the presence of the mTOR inhibitor rapamycin sug-
gesting that mTOR stimulation is a direct consequence of Wnt1
expression (96) (Fig. 4). These results connect persistent mitogen
stimulation and mTOR activation with senescence. Moreover,
they are in agreement with accumulated evidence connecting
accumulation of senescent cells with ageing (7, 9). Indeed,
increased mTOR activity was also observed in hematopoietic stem
cells from old mice (97), whereas mTOR inhibition by rapamycin
could extend lifespan both in invertebrates (98, 99) and in mice
(97, 100, 101).
In the context of OIS, mTOR promotes lysosome biogenesis,
which is required for autophagy and senescence-specific secretory
phenotype, in a newly identified cytoplasmic compartment (termed
TOR-autophagy spatial coupling compartment or TASCC) (102).
While these results clearly indicate that mTOR has pro-senescence
functions (Fig. 4), they are surprising in light of earlier report, con-
necting the induction of senescence by oncogenic ras with mTOR
downregulation (91). However, in this previous report it was
clearly shown that increased phosphorylation of both mTORC1
and mTORC2 substrates (S6K and FoxO3a, respectively) coin-
cided more with the onset of cycle arrest (and senescence) than
with ras-induced cell cycle entry, as proposed by the authors.
Indeed, transient activation of mTOR was also observed in cells in
which senescence was triggered by the genotoxic agent etoposide.
Therefore, while the results showing that appearance of late senes-
cence and autophagy markers coincided with downregulation of
mTOR activity, overall this work supports a positive role of mTOR
pathway in the induction of autophagy at the onset of senescence.
Although mainly focused on autophagy-related roles of mTOR,
the above results are consistent with the model proposed by
Blagosklonny and coworkers, whose recent experiments suggest
41. 27
2 mTOR, Metabolism and Cell Senescence
that cellular growth and persistent mTOR activity might be
implicated in the onset of the senescence program. Consistent with
their finding that serum stimulation is required for both the senes-
cent phenotype (large flat cells) and irreversibility of the cell cycle
arrest induced by overexpression of the CDK inhibitor p21 or
DNA damage, they observed that a key mTOR target, S6K, is
strongly phosphorylated in senescent cells (103). Moreover, in
agreement with our earlier observation (17), senescent cells also
accumulated another mTOR target, cyclin D1, but the biological
significance of its presence is not clear (see below). Interestingly,
while rapamycin, like serum starvation, prevented S6K phosphory-
lation and diminished the senescent marker β-gal staining, it failed
to completely abolish large flat cell morphology and to inhibit
cyclin D1 induction. While these results imply that mTORC1 is
not essential for these events, it would be interesting to test whether
they are controlled by mTORC2, which is less sensitive to rapamy-
cin and which is implicated in both cell shape and, via its target
Akt, in cyclin D1 stability (Figs. 3 and 4). In addition, rapamycin
prevents the onset of senescence if added simultaneously with
senescence inducers (p21 induction, genotoxic agents), but fails to
revert the already established senescence (104).
Taken together, the above results suggest that (transient?)
mTOR activity might positively control senescence but it is still
unclear how DDR network controls activity of this pathway.
How one can reconcile the data suggesting a positive role of mTOR
in senescence with those showing that p53 inhibits the mTOR
pathway (76, 77)? In the work of Narita’s team the possible inter-
actions between p53 and the mTOR pathway were not examined,
because in OIS cell cycle arrest is mainly mediated by the CDK
inhibitor p16, which is not regulated by p53 (91) (Fig. 1). In the
quest to decipher the respective roles of mTOR and p53 in the
senescence program, Blagosklonny and coworkers took advantage
of nutlin-3A, a specific small-molecule MDM2 antagonist, which
strongly induces p53 expression thereby triggering its transcrip-
tional activity in a non-genotoxic manner (105). Surprisingly, while
previous work has shown that nutlin-3A induces senescence in
mouse fibroblasts (106), in human fibrosarcoma cells HT1080
and WI-38 human fibroblasts, nutlin-3A induces reversible cell
cycle exit resembling quiescence (107). By exploiting a HT1080-
derived cell line in which p21 is expressed from an inducible
promoter, Demidenko et al. showed that nutlin-3A or p53 overex-
pression could “convert” p21-induced senescence into a quies-
cence-like state (108). Nutlin-3A was, however, much less efficient
8. Connections
Between mTOR
and p53 in
Senescence—
Lessons from
Nutlin-3A
42. 28 V. Dulic
in preventing senescence in the presence of DNA damage by H2
O2
,
which also stimulates the phosphorylation of mTOR (108). In
addition, both nutlin-3A and rapamycin suppress senescence with-
out interfering with cell cycle arrest, which led the authors to sug-
gest that high p53 levels preclude the onset of senescence by
inhibitingmTORandinducingquiescence(108,109).Accordingly,
knockdown of TSC2, a negative mTORC1 regulator (Fig. 3), par-
tially compromises nutlin-3-induced quiescence, resulting in accu-
mulation of senescent cells (110). In agreement with the positive
role of mTOR in senescence, in cell lines in which nutlin-3A did
induce senescence (termed “senescence-prone”), mTOR was not
inhibited, whereas the quiescence was “restored” in the presence
of rapamycin (110). Unfortunately, these studies did not provide
insight into molecular mechanisms explaining differences between
“senescence-prone” and “quiescence-prone” cells. In agreement
with the above experiments, when exposed to etoposide, serum-
deprived or rapamycin-treated fibroblasts (or epithelial cells) failed
to become senescent despite p21 induction, as drug removal, con-
comitant with serum addition, enabled proliferation (111). In con-
trast, serum addition in the presence of etoposide induced
senescence, presumably by activating mTOR. These results sup-
port the hypothesis that quiescence (or mTOR inhibition) com-
promises senescence. However, it is not clear why the checkpoints
were not activated once cells exposed to etoposide reentered the
cell cycle (assuming that the DNA damage had not been repaired)
and how the cells got rid of high p21 levels.
This work, however, did not address the role of the CKI p27
in p53-induced quiescence. This point is relevant for understand-
ing the mechanism that induces quiescence, since mTOR inhibi-
tion by rapamycin or p53 overexpression might also block SGK/
Akt-mediated p27 phosphorylation and cytoplasmic localization
leading to its nuclear accumulation and activation (70, 71) (Fig. 3).
One could imagine that, in the presence of rapamycin or absence
of serum, p27-dependent CDK inactivation together with cyclin
D1 down-regulation could also contribute to quiescence.
Reactivation of mTOR (by serum addition) would revert this pro-
cess, enabling cell cycle entry and progression, probably by degrad-
ing p27 and p21 and inducing G1 cyclins (D- and E-type).
Overall, the results of Blagosklonny’s team are consistent with
the idea that mTOR positively regulates senescence, which might
be because p53 levels are not sufficient to inhibit mTOR-dependent
cell growth. According to their hypothesis, the “true role” of p53
(when highly expressed) would be to induce quiescence by inhibit-
ing mTOR, which in turn would block the onset of senescence.
Consequently, senescence occurs in situations where “the condi-
tions for quiescence are not met” (low p53 levels) and where p53
“fails” to suppress the mTOR pathway (108, 112). This hypothesis
is consistent with the data showing that quiescence prevents
43. 29
2 mTOR, Metabolism and Cell Senescence
p21-induced senescence or inappropriate differentiation due to
expression of the transcriptional repressor HES1, playing a key role
in the reversibility of this non-proliferative state (33) (Fig. 2).
There are, however, several caveats regarding the use of nutlin-
3A as a tool to study the role of p53 and mTOR in senescence. For
example, strong p53 induction by nutlin-3A is not usually observed
in either quiescence or senescence and it is not clear which biologi-
cal event produces equivalent amounts of p53. As a matter of fact,
primary fibroblasts expressing the HPV16-E6 (hereafter E6) onco-
gene that degrades p53 (113) can become quiescent in the absence
of serum or confluence, whereas strong p21 induction in senescent
cells occurs in the absence of significant increases in p53 protein
levels (43). Therefore, the hypothesis that high p53 levels induce
quiescence while low p53 levels lead to senescence (108, 109)
should be verified in other experimental models. For example, our
recent results showed that irreversible cell cycle arrest by genotoxic
drugs in non-transformed human cells is independent on the
degree of p53 activation (or levels) or even p21 levels but rather on
the efficiency of p21 to inhibit different CDKs (Lossaint, 2011).
Finally, as mentioned above, this model is in apparent contradic-
tion with the results showing that in other cell lines nutlin-3A does
not induce quiescence but instead elicits endoreduplication, giving
rise to tetraploid cells (114), senescence—due to persistent p21
expression (106, 115), or even apoptosis (116). One explanation
could be that nutlin-3A might have other targets than p53 (pRb?)
or that its effects might be dependent on the experimental proto-
col and cell type.
The above-mentioned work mostly addressed the role of mTOR in
premature (induced) senescence but less is known regarding its
implication in replicative senescence. Several earlier observations,
however, are consistent with the notion that the mTOR pathway
might be active in senescent cells and that it is not controlled by
p53 or affected by its status. In fact, in human fibroblasts aged
in vitro a suppression of the p53/p21 pathway compromises cell
cycle arrest, senescence-specific cyclin D2 induction (see below), as
well as formation of SAHF, but it does not prevent large flat cell
morphology or cyclin D1 accumulation (43). This implies that
these two sets of key events associated with senescence, i.e. irre-
versible cell cycle arrest and cell growth, might be uncoupled,
which can also explain the senescent phenotype (Fig. 4). In addi-
tion, some data suggest that mitogenic pathways in senescent cells
might be also deregulated. Firstly, in sharp contrast to early passage
fibroblasts, serum withdrawal in senescent fibroblasts does not
9. mTOR and
Senescence:
Constitutive
Activation or
Deregulation?
44. 30 V. Dulic
affect cyclin D1 mRNA or cyclin E1 expression or protein synthesis.
The CDKs associated with these cyclins are, however, inhibited by
p21, explaining the absence of DNA replication (17, 26) (Fig. 1).
Interestingly, in agreement with other publications (35, 117), late
senescence is associated with strong accumulation of cyclin D2,
whose levels are not affected by serum withdrawal. The presence of
G1 cyclins, and especially of cyclin D2, in senescent cells is intrigu-
ing and it is not clear whether they might play a role in senescence
or they merely reflect deregulated mTOR activity. One possibility
is that cyclin D1 is part of the DNA repair machinery, as suggested
recently (118). Secondly, late-passage p53-deficient fibroblasts
(E6) failed to become quiescent upon serum withdrawal, which is
documented by both the absence of cyclin D1 or cyclin A down-
regulation and p27 induction (or activation). Consequently, CDKs
controlling DNA replication were highly active even in the absence
of serum. However, these cells failed to proliferate due to deleteri-
ous effects of DNA damage resulting in aberrant mitoses, endorep-
lication, or cell death (43). It is therefore possible that, concomitant
with increased population doubling, gradual deregulation of the
mTOR pathway contributes to the senescent phenotype. In the
absence of the p53/pRb safeguard system, this deregulation pro-
vides a fertile ground for tumorigenesis.
In conclusion, the role of the mTOR pathway in senescence is
still controversial, partly due to the various models and experimen-
tal designs employed in different studies. Apart from the studies
manipulating upstream mTOR regulators (PTEN, Wnt1) that
reveal pro-senescence mTOR-functions, most of the work has
focused on the autophagy-related aspects of senescence, which
appeared to exclude mTOR as an important regulator of senes-
cence. Few researchers, however, considered a hypertrophic phe-
notype of senescent cells and elevated levels of D-type cyclins as an
evidence of mTOR activity. Future work will show whether or not
this activity is actually required for the onset of senescence, as some
investigators proposed, or it is merely the result of senescence-
associated deregulation of the mTOR pathway. If the former is the
case, it will be interesting to learn whether, and if so, how p53
and/or pRb networks contribute to its induction.
Acknowledgments
I am grateful to Drs Anne Brunet (Stanford University, Palo Alto,
USA) and Jacques Piette (CRBM, Montpellier, France) for their
insightful comments.
45. 31
2 mTOR, Metabolism and Cell Senescence
References
1. Hayflick L (1965) The limited in vitro life-
time of human diploid cell strains. Exp Cell
Res 37:614–636
2. Harley CB, Futcher AB, Greider CW (1990)
Telomeres shorten during ageing of human
fibroblasts. Nature 345:458–460
3. Olovnikov AM (1973) A theory of margino-
tomy. The incomplete copying of template
margin in enzymic synthesis of polynucle-
otides and biological significance of the phe-
nomenon. J Theor Biol 41:181–190
4. Stein GH, Dulic V (1995) Origins of G1
arrest in senescent human fibroblasts.
Bioessays 17:537–543
5. Kuilman T, Michaloglou C, Mooi WJ, Peeper
DS (2010) The essence of senescence. Genes
Dev 24:2463–2479
6. Adams PD (2009) Healing and hurting:
molecular mechanisms, functions, and pathol-
ogies of cellular senescence. Mol Cell 36:
2–14
7. Rodier F, Campisi J (2011) Four faces of cel-
lular senescence. J Cell Biol 192:547–556
8. Rodier F, Coppe JP, Patil CK, Hoeijmakers
WA, Munoz DP, Raza SR, Freund A, Campeau
E, Davalos AR, Campisi J (2009) Persistent
DNA damage signalling triggers senescence-
associated inflammatory cytokine secretion.
Nat Cell Biol 11:973–979
9. Baker DJ, Wijshake T, Tchkonia T,
LeBrasseur NK, Childs BG, van de Sluis B,
Kirkland JL, van Deursen JM (2011)
Clearance of p16Ink4a-positive senescent
cells delays ageing-associated disorders.
Nature 479:232–236
10. Kang TW, Yevsa T, Woller N, Hoenicke L,
Wuestefeld T, Dauch D, Hohmeyer A, Gereke
M, Rudalska R, Potapova A, Iken M, Vucur
M, Weiss S, Heikenwalder M, Khan S, Gil J,
Bruder D, Manns M, Schirmacher P, Tacke F,
Ott M, Luedde T, Longerich T, Kubicka S,
Zender L (2011) Senescence surveillance of
pre-malignant hepatocytes limits liver cancer
development. Nature 479:547–551
11. Dimri GP, Lee X, Basile G, Acosta M, Scott
G, Roskelley C, Medrano EE, Linskens M,
Rubelj I, Pereira-Smith O et al (1995) A bio-
marker that identifies senescent human cells
in culture and in aging skin in vivo. Proc Natl
Acad Sci U S A 92:9363–9367
12. Lee BY, Han JA, Im JS, Morrone A, Johung
K, Goodwin EC, Kleijer WJ, DiMaio D,
Hwang ES (2006) Senescence-associated beta-
galactosidase is lysosomal beta-galactosidase.
Aging Cell 5:187–195
13. Bernardi R, Pandolfi PP (2007) Structure,
dynamics and functions of promyelocytic leu-
kaemia nuclear bodies. Nat Rev Mol Cell Biol
8:1006–1016
14. Vernier M, Bourdeau V, Gaumont-Leclerc MF,
Moiseeva O, Begin V, Saad F, Mes-Masson AM,
Ferbeyre G (2011) Regulation of E2Fs and
senescence by PML nuclear bodies. Genes
Dev 25:41–50
15. Narita M, Nunez S, Heard E, Narita M, Lin AW,
Hearn SA, Spector DL, Hannon GJ, Lowe SW
(2003) Rb-mediated heterochromatin forma-
tion and silencing of E2F target genes during
cellular senescence. Cell 113:703–716
16. Di Micco R, Sulli G, Dobreva M, Liontos M,
Botrugno OA, Gargiulo G, Dal Zuffo R,
Matti V, d’Ario G, Montani E, Mercurio C,
Hahn WC, Gorgoulis V, Minucci S, d’Adda di
Fagagna F (2011) Interplay between onco-
gene-induced DNA damage response and
heterochromatin in senescence and cancer.
Nat Cell Biol 13:292–302
17. Dulic V, Drullinger LF, Lees E, Reed SI,
Stein GH (1993) Altered regulation of G1
cyclins in senescent human diploid fibroblasts:
accumulation of inactive cyclin E-Cdk2 and
cyclin D1-Cdk2 complexes. Proc Natl Acad
Sci U S A 90:11034–11038
18. Burkhart DL, Sage J (2008) Cellular mecha-
nisms of tumour suppression by the retino-
blastoma gene. Nat Rev Cancer 8:671–682
19. Chicas A, Wang X, Zhang C, McCurrach M,
Zhao Z, Mert O, Dickins RA, Narita M,
Zhang M, Lowe SW (2010) Dissecting the
unique role of the retinoblastoma tumor sup-
pressor during cellular senescence. Cancer
Cell 17:376–387
20. Martin N, Benhamed M, Nacerddine K,
Demarque MD, van Lohuizen M, Dejean A,
Bischof O (2012) Physical and functional
interaction between PML and TBX2 in the
establishment of cellular senescence. EMBO J
31:95–109
21. Herbig U, Jobling WA, Chen BP, Chen DJ,
Sedivy JM (2004) Telomere shortening trig-
gers senescence of human cells through a
pathway involving ATM, p53, and p21(CIP1),
but not p16(INK4a). Mol Cell 14:501–513
22. Vousden KH, Prives C (2009) Blinded by the
light: the growing complexity of p53. Cell
137:413–431
23. Baus F, Gire V, Fisher D, Piette J, Dulic V
(2003) Permanent cell cycle exit in G2 phase
after DNA damage in normal human
fibroblasts. EMBO J 22:3992–4002
46. 32 V. Dulic
24. Krishnamurthy J, Torrice C, Ramsey MR,
Kovalev GI, Al-Regaiey K, Su L, Sharpless NE
(2004) Ink4a/Arf expression is a biomarker
of aging. J Clin Invest 114:1299–1307
25. Alcorta DA, Xiong Y, Phelps D, Hannon G,
Beach D, Barrett JC (1996) Involvement of
the cyclin-dependent kinase inhibitor p16
(INK4a) in replicative senescence of normal
human fibroblasts. Proc Natl Acad Sci U S A
93:13742–13747
26. Stein GH, Drullinger LF, Soulard A, Dulic V
(1999) Differential roles for cyclin-dependent
kinase inhibitors p21 and p16 in the mecha-
nisms of senescence and differentiation in
human fibroblasts. Mol Cell Biol 19:
2109–2117
27. Gil J, Peters G (2006) Regulation of the
INK4b-ARF-INK4a tumour suppressor locus:
all for one or one for all. Nat Rev Mol Cell
Biol 7:667–677
28. Coller HA (2011) Cell biology. The essence
of quiescence. Science 334:1074–1075
29. Coller HA (2007) What’s taking so long?
S-phase entry from quiescence versus prolif-
eration. Nat Rev Mol Cell Biol 8:667–670
30. Ferbeyre G, de Stanchina E, Querido E,
Baptiste N, Prives C, Lowe SW (2000) PML
is induced by oncogenic ras and promotes
premature senescence. Genes Dev 14:
2015–2027
31. Lemons JM, Feng XJ, Bennett BD, Legesse-
Miller A, Johnson EL, Raitman I, Pollina EA,
Rabitz HA, Rabinowitz JD, Coller HA (2010)
Quiescent fibroblasts exhibit high metabolic
activity. PLoS Biol 8:e1000514
32. Coller HA, Sang L, Roberts JM (2006) A
new description of cellular quiescence. PLoS
Biol 4:e83
33. Sang L, Coller HA, Roberts JM (2008)
Control of the reversibility of cellular quies-
cence by the transcriptional repressor HES1.
Science 321:1095–1100
34. Blagosklonny MV (2006) Cell senescence:
hypertrophic arrest beyond the restriction
point. J Cell Physiol 209:592–597
35. Shelton DN, Chang E, Whittier PS, Choi D,
Funk WD (1999) Microarray analysis of repli-
cative senescence. Curr Biol 9:939–945
36. Pardee AB (1989) G1 events and regulation
of cell proliferation. Science 246:603–608
37. Polyak K, Kato JY, Solomon MJ, Sherr CJ,
Massague J, Roberts JM, Koff A (1994)
p27Kip1, a cyclin-Cdk inhibitor, links trans-
forming growth factor-beta and contact inhi-
bition to cell cycle arrest. Genes Dev 8:9–22
38. Fero ML, Rivkin M, Tasch M, Porter P,
Carow CE, Firpo E, Polyak K, Tsai LH,
Broudy V, Perlmutter RM, Kaushansky K,
Roberts JM (1996) A syndrome of multior-
gan hyperplasia with features of gigantism,
tumorigenesis, and female sterility in
p27(Kip1)-deficient mice. Cell 85:733–744
39. Sherr CJ, Roberts JM (1999) CDK inhibitors:
positive and negative regulators of G1-phase
progression. Genes Dev 13:1501–1512
40. Chu IM, Hengst L, Slingerland JM (2008)
The Cdk inhibitor p27 in human cancer:
prognostic potential and relevance to antican-
cer therapy. Nat Rev Cancer 8:253–267
41. Itahana K, Dimri GP, Hara E, Itahana Y, Zou
Y, Desprez PY, Campisi J (2002) A role for
p53 in maintaining and establishing the qui-
escence growth arrest in human cells. J Biol
Chem 277:18206–18214
42. Pajalunga D, Mazzola A, Salzano AM, Biferi
MG, De Luca G, Crescenzi M (2007) Critical
requirement for cell cycle inhibitors in sus-
taining nonproliferative states. J Cell Biol
176:807–818
43. Dulic V, Beney GE, Frebourg G, Drullinger LF,
Stein GH (2000) Uncoupling between phe-
notypic senescence and cell cycle arrest in
aging p21-deficient fibroblasts. Mol Cell Biol
20:6741–6754
44. Nakayama K, Ishida N, Shirane M, Inomata
A, Inoue T, Shishido N, Horii I, Loh DY
(1996) Mice lacking p27(Kip1) display
increased body size, multiple organ hyperpla-
sia, retinal dysplasia, and pituitary tumors.
Cell 85:707–720
45. Matsushime H, Roussel MF, Ashmun RA,
Sherr CJ (1991) Colony-stimulating factor 1
regulates novel cyclins during the G1 phase of
the cell cycle. Cell 65:701–713
46. Baldin V, Lukas J, Marcote MJ, Pagano M,
Draetta G (1993) Cyclin D1 is a nuclear pro-
tein required for cell cycle progression in G1.
Genes Dev 7:812–821
47. Atadja P, Wong H, Veillete C, Riabowol K
(1995) Overexpression of cyclin D1 blocks
proliferation of normal diploid fibroblasts.
Exp Cell Res 217:205–216
48. Pani G (2011) From growing to secreting:
new roles for mTOR in aging cells. Cell Cycle
10:2450–2453
49. Blagosklonny MV (2011) Cell cycle arrest is
not senescence. Aging (Albany NY) 3:94–101
50. Heitman J, Movva NR, Hall MN (1991)
Targets for cell cycle arrest by the immuno-
suppressant rapamycin in yeast. Science
253:905–909
51. Wullschleger S, Loewith R, Hall MN (2006)
TOR signaling in growth and metabolism.
Cell 124:471–484
47. 33
2 mTOR, Metabolism and Cell Senescence
52. Dazert E, Hall MN (2011) mTOR signaling
in disease. Curr Opin Cell Biol 23:744–755
53. Zoncu R, Efeyan A, Sabatini DM (2011)
mTOR: from growth signal integration to
cancer, diabetes and ageing. Nat Rev Mol Cell
Biol 12:21–35
54. Sengupta S, Peterson TR, Sabatini DM
(2010) Regulation of the mTOR complex 1
pathway by nutrients, growth factors, and
stress. Mol Cell 40:310–322
55. Conn CS, Qian SB (2011) mTOR signaling
in protein homeostasis: less is more? Cell
Cycle 10:1940–1947
56. Oh WJ, Jacinto E (2011) mTOR complex 2
signaling and functions. Cell Cycle 10:
2305–2316
57. Zinzalla V, Stracka D, Oppliger W, Hall MN
(2011) Activation of mTORC2 by association
with the ribosome. Cell 144:757–768
58. Averous J, Fonseca BD, Proud CG (2008)
Regulation of cyclin D1 expression by
mTORC1 signaling requires eukaryotic initi-
ation factor 4E-binding protein 1. Oncogene
27:1106–1113
59. Balcazar N, Sathyamurthy A, Elghazi L,
Gould A, Weiss A, Shiojima I, Walsh K,
Bernal-Mizrachi E (2009) mTORC1 activa-
tion regulates beta-cell mass and proliferation
by modulation of cyclin D2 synthesis and sta-
bility. J Biol Chem 284:7832–7842
60. Lavoie JN, L’Allemain G, Brunet A, Muller
R, Pouyssegur J (1996) Cyclin D1 expression
is regulated positively by the p42/p44MAPK
and negatively by the p38/HOGMAPK path-
way. J Biol Chem 271:20608–20616
61. Hitomi M, Stacey DW (1999) Cyclin D1 pro-
duction in cycling cells depends on ras in a cell-
cycle- specific manner. Curr Biol 9:1075–1084
62. Dowling RJ, Topisirovic I, Alain T, Bidinosti
M, Fonseca BD, Petroulakis E, Wang X,
Larsson O, Selvaraj A, Liu Y, Kozma SC,
Thomas G, Sonenberg N (2010) mTORC1-
mediated cell proliferation, but not cell
growth, controlled by the 4E-BPs. Science
328:1172–1176
63. Petersen J, Nurse P (2007) TOR signalling
regulates mitotic commitment through the
stress MAP kinase pathway and the Polo and
Cdc2 kinases. Nat Cell Biol 9:1263–1272
64. Fingar DC, Salama S, Tsou C, Harlow E,
Blenis J (2002) Mammalian cell size is con-
trolled by mTOR and its downstream targets
S6K1 and 4EBP1/eIF4E. Genes Dev 16:
1472–1487
65. Pyronnet S, Dostie J, Sonenberg N (2001)
Suppression of cap-dependent translation in
mitosis. Genes Dev 15:2083–2093
66. Smith EM, Proud CG (2008) cdc2-cyclin B
regulates eEF2 kinase activity in a cell cycle-
and amino acid-dependent manner. EMBO J
27:1005–1016
67. Ramirez-Valle F, Badura ML, Braunstein S,
Narasimhan M, Schneider RJ (2010) Mitotic
raptor promotes mTORC1 activity, G(2)/M
cell cycle progression, and internal ribosome
entry site-mediated mRNA translation. Mol
Cell Biol 30:3151–3164
68. Gwinn DM, Asara JM, Shaw RJ (2010)
Raptor is phosphorylated by cdc2 during
mitosis. PLoS One 5:e9197
69. Hong F, Larrea MD, Doughty C, Kwiatkowski
DJ, Squillace R, Slingerland JM (2008)
mTOR-raptor binds and activates SGK1 to
regulate p27 phosphorylation. Mol Cell 30:
701–711
70. Toker A (2008) mTOR and Akt signaling in
cancer: SGK cycles in. Mol Cell 31:6–8
71. Wander SA, Zhao D, Slingerland JM (2011)
p27: a barometer of signaling deregulation
and potential predictor of response to tar-
geted therapies. Clin Cancer Res 17:12–18
72. Guertin DA, Stevens DM, Thoreen CC,
Burds AA, Kalaany NY, Moffat J, Brown M,
Fitzgerald KJ, Sabatini DM (2006) Ablation
in mice of the mTORC components raptor,
rictor, or mLST8 reveals that mTORC2 is
required for signaling to Akt-FOXO and
PKCalpha, but not S6K1. Dev Cell 11:
859–871
73. Medema RH, Kops GJ, Bos JL, Burgering BM
(2000) AFX-like Forkhead transcription fac-
tors mediate cell-cycle regulation by Ras and
PKB through p27kip1. Nature 404:782–787
74. Schmidt M, Fernandez de Mattos S, van der
Horst A, Klompmaker R, Kops GJ, Lam EW,
Burgering BM, Medema RH (2002) Cell
cycle inhibition by FoxO forkhead transcrip-
tion factors involves downregulation of cyclin
D. Mol Cell Biol 22:7842–7852
75. Kroemer G, Pouyssegur J (2008) Tumor cell
metabolism: cancer’s Achilles’ heel. Cancer
Cell 13:472–482
76. Vousden KH, Ryan KM (2009) p53 and
metabolism. Nat Rev Cancer 9:691–700
77. Feng Z (2010) p53 regulation of the IGF-1/
AKT/mTOR pathways and the endosomal
compartment. Cold Spring Harb Perspect
Biol 2:a001057
78. Young AR, Narita M (2010) Connecting
autophagy to senescence in pathophysiology.
Curr Opin Cell Biol 22:234–240
79. Kroemer G, Marino G, Levine B (2010)
Autophagy and the integrated stress response.
Mol Cell 40:280–293
48. 34 V. Dulic
80. Inoki K, Zhu T, Guan KL (2003) TSC2
mediates cellular energy response to control
cell growth and survival. Cell 115:577–590
81. Budanov AV, Karin M (2008) p53 target
genes sestrin1 and sestrin2 connect genotoxic
stress and mTOR signaling. Cell 134:
451–460
82. Feng Z, Hu W, de Stanchina E, Teresky AK,
Jin S, Lowe S, Levine AJ (2007) The regula-
tion of AMPK beta1, TSC2, and PTEN
expression by p53: stress, cell and tissue
specificity, and the role of these gene products
in modulating the IGF-1-AKT-mTOR path-
ways. Cancer Res 67:3043–3053
83. Feng Z, Zhang H, Levine AJ, Jin S (2005)
The coordinate regulation of the p53 and
mTOR pathways in cells. Proc Natl Acad Sci
U S A 102:8204–8209
84. Gwinn DM, Shackelford DB, Egan DF,
Mihaylova MM, Mery A, Vasquez DS, Turk
BE, Shaw RJ (2008) AMPK phosphorylation
of raptor mediates a metabolic checkpoint.
Mol Cell 30:214–226
85. Jones RG, Plas DR, Kubek S, Buzzai M, Mu
J, Xu Y, Birnbaum MJ, Thompson CB (2005)
AMP-activated protein kinase induces a p53-
dependent metabolic checkpoint. Mol Cell
18:283–293
86. Feng Z, Levine AJ (2010) The regulation of
energy metabolism and the IGF-1/mTOR
pathways by the p53 protein. Trends Cell Biol
20:427–434
87. Kong M, Fox CJ, Mu J, Solt L, Xu A, Cinalli
RM, Birnbaum MJ, Lindsten T, Thompson
CB (2004) The PP2A-associated protein
alpha4 is an essential inhibitor of apoptosis.
Science 306:695–698
88. Lee CH, Inoki K, Karbowniczek M,
Petroulakis E, Sonenberg N, Henske EP,
Guan KL (2007) Constitutive mTOR activa-
tion in TSC mutants sensitizes cells to energy
starvation and genomic damage via p53.
EMBO J 26:4812–4823
89. Kim J, Kundu M, Viollet B, Guan KL (2011)
AMPK and mTOR regulate autophagy
through direct phosphorylation of Ulk1. Nat
Cell Biol 13:132–141
90. Selman C, Tullet JM, Wieser D, Irvine E,
Lingard SJ, Choudhury AI, Claret M,
Al-Qassab H, Carmignac D, Ramadani F,
Woods A, Robinson IC, Schuster E, Batterham
RL, Kozma SC, Thomas G, Carling D,
Okkenhaug K, Thornton JM, Partridge L,
Gems D, Withers DJ (2009) Ribosomal pro-
tein S6 kinase 1 signaling regulates mamma-
lian life span. Science 326:140–144
91. Young AR, Narita M, Ferreira M, Kirschner K,
Sadaie M, Darot JF, Tavare S, Arakawa S,
Shimizu S, Watt FM, Narita M (2009)
Autophagy mediates the mitotic senescence
transition. Genes Dev 23:798–803
92. Zwerschke W, Mazurek S, Stockl P, Hutter E,
Eigenbrodt E, Jansen-Durr P (2003)
Metabolic analysis of senescent human
fibroblasts reveals a role for AMP in cellular
senescence. Biochem J 376:403–411
93. Ruggero D, Montanaro L, Ma L, Xu W,
Londei P, Cordon-Cardo C, Pandolfi PP
(2004) The translation factor eIF-4E pro-
motes tumor formation and cooperates with
c-Myc in lymphomagenesis. Nat Med 10:
484–486
94. Astle MV, Hannan KM, Ng PY, Lee RS,
George AJ, Hsu AK, Haupt Y, Hannan RD,
Pearson RB (2011) AKT induces senescence
in human cells via mTORC1 and p53 in the
absence of DNA damage: implications for tar-
geting mTOR during malignancy. Oncogene
31:1949–1962
95. Inoki K, Ouyang H, Zhu T, Lindvall C, Wang
Y, Zhang X, Yang Q, Bennett C, Harada Y,
Stankunas K, Wang CY, He X, MacDougald
OA, You M, Williams BO, Guan KL (2006)
TSC2 integrates Wnt and energy signals via a
coordinated phosphorylation by AMPK and
GSK3 to regulate cell growth. Cell 126:
955–968
96. Castilho RM, Squarize CH, Chodosh LA,
Williams BO, Gutkind JS (2009) mTOR
mediates Wnt-induced epidermal stem cell
exhaustion and aging. Cell Stem Cell
5:279–289
97. Chen C, Liu Y, Liu Y, Zheng P (2009) mTOR
regulation and therapeutic rejuvenation of
aging hematopoietic stem cells. Sci Signal
2:ra75
98. Vellai T, Takacs-Vellai K, Zhang Y, Kovacs
AL, Orosz L, Muller F (2003) Genetics:
influence of TOR kinase on lifespan in C.
elegans. Nature 426:620
99. Kaeberlein M, Powers RW 3rd, Steffen KK,
Westman EA, Hu D, Dang N, Kerr EO,
Kirkland KT, Fields S, Kennedy BK (2005)
Regulation of yeast replicative life span by
TOR and Sch9 in response to nutrients.
Science 310:1193–1196
100. Harrison DE, Strong R, Sharp ZD, Nelson
JF, Astle CM, Flurkey K, Nadon NL,
Wilkinson JE, Frenkel K, Carter CS, Pahor
M, Javors MA, Fernandez E, Miller RA
(2009) Rapamycin fed late in life extends
lifespan in genetically heterogeneous mice.
Nature 460:392–395
101. Kapahi P, Chen D, Rogers AN, Katewa SD, Li
PW, Thomas EL, Kockel L (2010) With TOR,
less is more: a key role for the conserved
50. of our history, but they are Bank holidays. The closing of our banks
is the one signal that for twenty-four hours we are free.
The multifarious aspects of the theme are most bewildering. As
Sir Henry Taylor said, “So manifold are the bearings of money upon
the lives and character of mankind, that an insight which should
search out the life of a man in his pecuniary relations would
penetrate into almost every cranny of his nature. For if we take
account of all the virtues with which money is mixed up, honesty,
justice, charity, frugality, forethought, self-sacrifice, and of their
correlative vices, it is a knowledge that goes near to cover the length
and breadth of humanity.”
It is certainly true that the amazingly extensive nature of the
subject might lead one away into perfectly relevant discussions of
almost every field of human activity; and nothing renders argument
so unsatisfactory and inconclusive as to have unlimited scope. But in
these pages the issue must be narrowed down and the question
confined so far as possible to a very brief examination of one
particular aspect of the subject, which will be created by formulating
a deliberate contention and pursuing it by argument into some of
the main channels of this perplexing problem. Even so it is likely that
deep water will be reached, but, after all, a suggestion need not be
driven to its utmost limits in all directions in order to establish its
significance.
In choosing a direct point of attack against this generally
accepted belief we shall treat the matter more or less from a
practical point of view. Without getting involved in abstract
philosophic propositions, without entering too far into the sphere of
economics and politics, without preaching high morality, though the
words and teachings of preachers must be quoted, an endeavour
will be made, by working out a definite line of reasoning, to submit
as a whole some of the simpler and perhaps more personal
considerations which have no doubt already occurred to many who
have given the subject thought and reflection. No maxims will be
laid down as to how money should be made, spent, saved, lent,
51. borrowed, invested, given or bequeathed, for the object is to strike
at the root principle and shatter the ideal which underlies all those
transactions, which colours men’s characters, influences their desires
and aspirations, creates artificial class contrasts, and contributes
largely to the general social confusion and chaos.
Briefly, then, our contention is: That no individual is capable of
possessing, spending, or administering more than a certain definite
amount of money, which can be roughly described as a full
competence, without producing positively harmful effects on himself
as well as on those affected by his actions. In other words, the “rich
man” is an impossibility in any decently organised economic State,
and the accumulation of capital in individual hands is detrimental to
the public good. That is what is meant by the saying from which the
title of this volume is taken.
It may appear at first sight to be an extreme view, because we
have got so much accustomed to believing that a great deal of good
can be done with money, and a great deal of happiness derived from
it, that to be confronted with an uncompromising negation on such a
time-honoured tradition may seem almost absurd. The argument is
purposely intended to be completely comprehensive, and a case will
be presented without exaggeration which will cover as much of the
ground as possible, dealing with typical rather than exceptional
instances by way of illustration.
We find in human nature three characteristic impulses which
serve as the mainspring and motive power in the gaining and
spending of money: the passion for acquisition, the instinct for
absolute property, and the desire to excel. No one would suggest
that the passion for acquisition can be destroyed: it is neither
possible nor desirable, but it can be prevented from running wild,
and it can be controlled, though it does not seem to have occurred
to many people that such control is expedient. The instinct for
absolute property is very much overestimated, and this arises from
the fact that we are accustomed to a system which hardly allows any
satisfactory intermediate stage between property and positive need.
52. The craving for complete possession on any considerable scale only
enters into the minds of those who covet their neighbours’
possessions. What a man wants and has every right to expect is
security in the enjoyment of his necessaries and comforts, but this is
precisely what in the vast majority of cases he does not get; and his
want remaining unsatisfied is converted into a craving for absolute
property. The desire to excel, which can undoubtedly be one of the
finest human qualities, is in itself vitiated by the measure of money,
which sets up an utterly false standard of excellence and converts
pure ambition into a desire for material pre-eminence.
However far we may travel, the problem will be continually
resolving itself into some variation of the question as to how these
impulses had best be regulated, and to what extent they have
broken out beyond their legitimate bounds. But although the causes
of the faith in money may be reduced to moral and psychological
terms, there are economic as well as moral results, and it is not the
metaphysical origins, but the practical results which must be looked
into.
At the outset we must acknowledge that our capacities of all
kinds are strictly limited, whether moral, intellectual, or physical. An
occasional saint, an occasional genius, or an occasional giant
stretches the limit beyond its normal point, but the limit still remains.
And yet we are foolish enough to believe that in regard to the
possession, expenditure, and administering of riches there is no sum
of money, however large, which we are not competent to deal with,
and we are convinced that it is quite easy and unquestionably within
the capacity of almost anyone to spend with benefit to himself and
to others sums of money greatly in excess of what can cover in the
widest sense his personal requirements. Whereas not only is it not
easy, but as inquiry will show, it is purely and positively impossible,
as impossible as it is to acquire vast knowledge with a limited brain
capacity, or to endure more than a certain amount of physical strain
with a limited muscular capacity. We are inclined also to think that
men who have money and men who make money are ipso facto
easily capable of spending the money properly, though we generally
53. make the mental reservation that if we had it ourselves we should
spend it a great deal better. But the inheritance or accumulation of
money does not imply by any means a special ability for spending it
wisely. To put it plainly, such men have not, nor have we, nor has
anyone this ability except in a very limited degree, far more limited
than is generally supposed or ever admitted.
The case against riches has been argued again and again on
religious and moral grounds for over two thousand years, from
Confucius to Tolstoy. But we are less impressed by the truth of it
now than ever we were; and we still hear it stated by high
authorities that it is a benefit to the community to contain men of
great wealth. The whole delusion arises from the indestructible
confidence in what money can do. And yet all of us see clearly
enough by the roughest and most general observation that
happiness does not increase with riches, that money indeed has very
little to do with happiness, though it has a good deal to do with
misery. But many of us are inclined to believe that our own
individual case is rather different, and that more money added to
our ample competence, and a consequent further enjoyment of
material possessions, must undoubtedly make us happier. And when
we have got the more and the desired result is not attained, we
never pause in hesitation to consider whether perhaps the more has
interfered with rather than augmented our happiness, but we are
persuaded that the reason we find ourselves still discontented is
simply that the more is not enough. Enough never comes to those
who have encouraged the longing for more. Nothing short of actual
experience can help to eradicate this belief, but there are few who
would care to embark on an experiment in the direction of less. And
yet it could quite well be demonstrated that a reduction of income,
provided always that the loss does not reduce the income below a
competence can lead to an increase in happiness—happiness being,
of course, distinguished from pleasure.
It may require a very rare philosophic resignation and an equally
rare breadth of view to refuse to be deluded into regarding the
possession of money as an absolute essential. Moreover, there are a
54. great many qualifications to be taken into account arising from
natural characteristics, habit, temperament, and tastes. But broadly
speaking, if a man has the courage to regard a reduction of income
not as a loss but a gain, if he can use the opportunity to kill the
instinctive but disturbing craving for more which unfortunately
seems engrained in us all, in fact, if he can eradicate the germ of the
disease, the limitation of his desire to satisfy transient and what are
really artificial needs will certainly increase his power of enjoyment
and his happiness. On the other hand, if he treats the lowering of his
means as a calamity, which is the usual case, lamenting his fate,
railing against fortune and encouraging the longing for gain—an
attitude of mind which is only the outcome of his unlimited faith in
the power of money—the result, naturally enough, will be despair.
But it might be shown as well that a type of man does exist,
exceptional no doubt, who, being capable of spending without hurt
to himself or to others more money than he has actually got, can
enrich his life in the broadest sense by an increase of fortune, and
may therefore become the happier for it. He is a man who is
indifferent to the enjoyment of material possessions and probably
would be regarded in the eyes of the world as the last man who was
competent to use money properly. But even he would be entirely
overwhelmed by anything like a large increase of fortune, and would
be as incapable as any one else of disposing of it without inflicting
injury.
“Could not riches be used well?” asks Jean Marie in Stevenson’s
Treasure of Franchard.
“In theory, yes,” replied the doctor. “But it is found in experience
that no one does so. All the world imagine they will be exceptional
when they grow wealthy; but possession is debasing, new desires
spring up, and the silly taste for ostentation eats out the heart of
pleasure.”
Money is, after all, responsibility and nothing else. We are all of
us capable of undertaking a certain amount. Some of us are capable
of undertaking a good deal. No one is capable of undertaking more
55. than a relatively limited amount. But the trouble is that most of us
think ourselves capable of undertaking far more than we properly
can. Autocrats are ceasing to exist not so much because certain
monarchs proved themselves dangerously incapable, but because
the world has learned that no conceivable human being has the
capacity to rule a country single-handed. We do not yet admit this
incapacity with regard to the autocrats over capital, although it is
equally true, and when we do so we shall find considerable difficulty
in dethroning them.
Another important inference to be deduced from the argument
here set forth is that the surplus money which no individual does or
can spend beneficially remains in his hands in stagnant
unproductivity, is deflected from other remunerative channels, and is
therefore the chief cause of the existence of some of the gravest
economic ills which we have to face in our social life. Money cannot
rest, it is an active instrument for producing good or for producing
evil. Its presence in one quarter may not produce visible evil, but its
consequent absence in another quarter will produce very visible and
very positive evil. The word consequent must be emphasised
because wealth is like water—to pump it up artificially on one side is
to lower it automatically on the other.
Money in its character of potential wealth seems also to have
this peculiar characteristic. It has no positive value in itself. The
greater part of its value is given to it by its possessor, and in
proportion as it accumulates in the hands of an individual its value is
rapidly depreciated. An electric current of a certain power will
perform certain specified functions. Decrease the power and it
ceases to produce the required effect. Increase the power tenfold or
a hundredfold and you will be no nearer achieving the desired result.
That is to say, in addition to the change in value effected by the
change in individual ownership, there is actual deterioration,
produced by accumulation, whoever the individual may be who is
responsible for that accumulation.
56. As with individuals, so with the State. National wealth, which in
the highest sense of the word means the enrichment of the lives of
the people, depends not on how large a number of incomes there
are of over ten thousand a year, but on how small a number there
are of under two hundred a year. The real riches of a nation are not
to be measured by vast calculations of commercial statistics, but by
the absence of destitution and the high level of healthy life which
the people enjoy.
But we must accept the situation as it is. The rich have got their
riches, and the problem to be considered here is not how to deprive
them of their riches, but how to prevent all men, rich and poor alike,
from confiding blindly in money, as they do at present, and from
striving towards a false ideal which spoils their highest endeavours,
blunts their moral susceptibilities, poisons their happiness, and
produces a state of social disorder which is highly prejudicial to the
common good. A just appreciation of the essential fact that money
can only be made out of people’s labour and the wear and tear of
their lives would in itself do much to prevent the growth of the spirit
which leads to these alarming contrasts in riches and poverty. But
men’s ideals and their moral outlook can only be altered in the long
run by repeatedly exposing the actual fallacies in the views they now
hold and constantly emphasising the disastrous results of the actions
for which this waste of money is responsible.
57. A
Chapter III
Definition of the limit—Those whose means are above the
limit—Income translated into terms of subsistence—The
case of the rich man—His establishments—His servants—
His luxuries—Extravagance—Vanity—Sport—Racing—
Yachting—Condemnation of excess.
MORE precise definition must be given of the limit of income
referred to in the last chapter as “a definite amount of money
which might be roughly described as a full competence.”
Every man requires, though he by no means always gets a
certain income to satisfy his own needs and those of his family. In
addition to this he can profitably spend more so as to add to his
general utility by conveniences and comforts, he can satisfy his
artistic proclivities, his desire for further knowledge, his taste for
sport or amusement, all to his own and the general benefit without
hurt or hindrance to anyone. But after allowing the broadest scope
for the satisfaction of these legitimate wants there is a definite point
beyond which he cannot safely go. That is to say, if he acquires, or if
by inheritance he finds himself burdened with money beyond this
limit it will inevitably react detrimentally on himself and on others.
And this for two reasons: firstly because he is, as a normal human
being, incapable of dealing with so great a charge, and secondly
because the money, while in his possession, is being drawn away
from other channels where there is special need for it.
So long as money encourages healthy effort a man may be sure
the limit has not been reached, the moment money tends to relax
58. effort the limit has been passed. It must be described as healthy
effort, as, of course, money-making may increase the undesirable
efforts of the speculator, the gambler, and the thief. But who is to
decide what is healthy effort? The man himself. No one else can.
And he knows to a nicety. Every man or woman has a different
standard, and the level of the limit varies in each individual case
according to ideals, capacity, and temperament. But it will not
depend at all on what is one of the strongest and often the most
excusable inducements for spending money, namely, environment,
or the conventions of the particular stratum of society to which the
man belongs. The limit for one will not be the limit for another, and
a man can only become aware that this limit exists at all by
observing very closely what actually is the effect that his money is
having on his life and character, instead of blindly accepting his
already excessive income or every increase of his fortune as a
natural and unquestionable blessing.
The main brunt of the attack must clearly fall on those whose
incomes are above the limit. They are in numbers a small minority,
but the amount they possess is incredibly large. The present income
of 1,250,000 people, assessed to income, reaches the vast sum of
£850,000,000 a year. Taking the whole population of these islands, it
is roughly estimated that there are 1½ millions who can be classed
as rich, 3½ millions comfortably off, 38 millions as poor, of whom
some 12 to 13 millions are in constant need. The existence of the
1½ millions is one of the chief causes of the condition of the 38
millions. In other words, excess above the limit causes want below
the limit. The 3½ millions “comfortably off” are most of them
occupied in trying to become identified with the select 1½ millions.
If we could estimate the amounts in income which these classes
represent the figures would be even more startling. The world has
certainly never seen larger fortunes than exist to-day, nor has it seen
more extensive and more inexcusable poverty. The average rate of
luxurious living in the small minority is higher than it has ever been,
and the dangerous and degrading effect of want on individuals and
on the general community has never been so widespread or so
59. intense. “The rich,” to use a simple term, are nearly all actuated by
the same motive. They accept what they have and what they make
as their own, to be spent on themselves, according to their own
caprice, or on others, if they are so inclined, casting an occasional
sop to some charity or philanthropic scheme as a salve to their
consciences. There are, it must be acknowledged, a few, a very few
who regard their riches as a trust and endeavour to the best of their
ability to divert the greater part of it back into remunerative
channels without exceeding a reasonable sum for their own personal
wants. But as a class they insist that efforts to alter our social
system are fruitless, disturbing and doomed to failure, the division of
the world into rich and poor being a Providential decree, and if the
rich can get service from the poor without their grumbling, that is
the most desirable arrangement that can be conceived. To this a
reply may be given in the words of Professor Marshall:
“Now at least we are setting ourselves seriously to inquire
whether it is necessary that there should be any so-called
‘lower class’ at all: that is whether there need be large
numbers of people doomed from their birth to hard work in
order to provide for others the requisites of a refined and
cultured life, while they themselves are prevented by their
poverty and toil from having any share in that life.”
The case would not be quite so bad as it is if it were only “the
requisites of a refined and cultured life” that they were made to
provide. But this point must be considered later.
In order to appreciate fully the responsibility which the
possession of riches entails, let us translate an income into terms of
actual sustenance for human beings. By this means it is possible to
arrive at a more or less positive measure. There is so much that is
relative in most human requirements that they cannot serve as a
standard or as a reliable quantity to be used in calculating any
equation. But the requirements of a human being can be measured
60. in terms of actual sustenance, because they can be estimated with
something approaching precision.
Take a man with £20,000 a year, and say we deduct even as
much as £3000 for himself and his family. With his remaining
£17,000 he has the power of furnishing 170 people with £100 a year
apiece. It is not for a moment suggested that he should do any such
thing, as he would be quite unable to select 170 worthy people, and
even if he could make the choice the 170 people, on the reception of
this private dole, would soon become unworthy. This calculation is
only taken to serve as a measure of his power. What might be the
income or, more correctly speaking, the means of existence of 170
lives, is vested in one man, who is under the impression—and no
one attempts to dispute it—that he is capable of disposing of this
sum in a way that is generally beneficial.
Now let us state the case fairly from the point of view of the rich
man, taking a reasonable and more or less representative type. He
may have £10,000, £50,000, or £100,000 a year—that only alters his
activities in scope, not in quality. Let us say he has two or three
country houses and a house in London. His “position” requires him
to keep up a certain establishment, and this means the employment
of some forty or fifty servants, grooms, gardeners, chauffeurs, etc.,
who, he readily tells you, will be thrown out of employment should
any of his money be taken from him. If we take the case of a
landlord, he will also have tenants, bailiffs, farm labourers, and
gamekeepers dependent on him. He keeps the home farm and lets
out the other farms on his estate to tenant farmers. Part of his land
is built over and brings him in substantial returns in the shape of
rent. His villages are in good order and the cottages kept in proper
repair. Some thousands of acres or so he keeps for shooting. He may
have a deer forest on one of his estates, and perhaps also a grouse
moor or a river. Whether he keeps a racing stable, a pack of hounds,
or a yacht depends on his particular fancy. He will acknowledge that
he spends a certain amount on luxuries, but that “is good for trade,”
as great numbers of people have to be employed in the manufacture
of these luxuries. He is kind to his poor relations, whom he
61. entertains and helps; and his subscription list to hospitals, charities,
and philanthropic works is a large one. He enjoys himself in an
unostentatious but suitably expensive way, and his various
responsibilities allow him to lead a life consisting of occasional
rushes of activity and prolonged intervals of leisure. He most
probably finds he can spare a certain amount of money for
speculation, with a view to adding more to the sum total of his
income. He looks forward to handing down to his children sufficient
means to make each of them independent, and meanwhile has his
boys educated in the large public schools, where they can associate
with boys who are similarly situated.
What possible harm can there be in all this? So far from being
parasitic, he counts himself as a beneficent agent in the general
industrial activity, at the same time appearing as a credit to his
society and a notably refined product of the class of which he is a
member. Above all, he is popular, and gains ostensibly the respect
and regard of his friends, his neighbours, and his dependents. A
favourable case has purposely been made for him, because if we
accuse him of self-indulgence and greed, and describe him as a
gambler, spending his substance on objects which are generally
admitted to be pernicious and unworthy, the case could not be
defended at all.
The fundamental theory which makes this man’s position
untenable has already been explained—namely, that after he has
satisfied his legitimate requirements all the surplus money he keeps
is being held back from serving urgent needs; and, moreover, the
method in which he spends the surplus is directly or indirectly
harmful to himself and others.
We call the money his as if by some miracle he had made it.
Often enough he has not helped even by the smallest exertion to
create it. The wealth has been and is being daily and hourly
produced by the exertion of numberless people who are either
employed by him or employed in furthering enterprises in which he
has invested his money. It will be said that his share as the wise
62. dispenser of capital, without which labour and enterprise could not
be set in motion, is an all-important part of the general process of
business. But he invests not to promote enterprise, but to get high
dividends; and an elaborate system has been set up in order to
tempt him to put his money into concerns that are by no means
always sound or of the smallest public utility. Capital would exist and
flow far more freely without the large capitalist. He acts as a dam to
the stream; a certain amount escapes back into the main channel,
but much more is checked and diverted into stagnant and putrefying
pools of his own creation. The free flow of blood is life-giving; the
clotting or coagulation of blood produces disease.
Let us take the various points raised by his case seriatim. Many
acutely controversial problems are opened, and it will be difficult to
detach the particular actions of the rich man without generalising, to
some extent, on the problems themselves. It is no argument against
our main contention to say that people with costly tastes have, while
gratifying them, been able to exercise powers of a high order, for,
obviously, it is in spite of their shortcomings in this respect that they
have succeeded, and not because of them. If some men with means
have done valuable public service and performed admirable work in
many different spheres of life, this they have done as men naturally
gifted with high accomplishments, not as rich men. Here we are only
concerned with their works and deeds in their latter capacity.
It does not affect our argument whether our typical example has
been brought up to regard this way of living as natural and
necessary for a man of what is called his “position” (that is to say,
the purely artificial place which a rich man is able to take up in the
community solely on account of his riches), or whether he has made
the money for himself and is simply aping the habits and customs of
those who already possess it. The distinction between the vieux
riches and the nouveaux riches is one they can fight out between
themselves. The former scoffs at the latter while all the time he is
setting him, and consciously setting him, the example he is to follow.
It is not the gaining, but the spending of the money that must
occupy our attention here.
63. Our friend’s houses are only a detail in the upkeep of his
position. They may be historic castles, sham “ancestral halls,”
modern “palatial country residences,” or “fashionable mansions” in
town. Does it ever strike the owner as, let us say, a curious
arrangement that he should have several houses of fifty to a
hundred rooms apiece while some millions of his fellow-men do not
own one room? Does he know that in England and Wales alone
507,763 people occupy one-room tenements, 48.4% of which are
classified as overcrowded, while 12,458,150 are occupying
tenements of two, three, or four rooms?
4
In any case, he would
indignantly refuse to admit that there was any remote connection
between these two sets of circumstances.
When we come to the staff necessary for the maintenance of
these large establishments we touch a problem of employment
which must be examined more closely. It is not sufficient to state
baldly that these people are employed, and that if the opening were
not available for them they would be unemployed. The immediate
result of their being discharged would no doubt in some cases be
unemployment. That is just the mischief of uneconomic
employment. If a large number were simultaneously dismissed there
might be temporary unemployment on a large scale, as it would
amount to dislocation, like the extinction of some dying industry. But
the eventual readjustment would subsequently be by that much the
stronger and better adapted to the real requirements of the
community. To employ a man in useless and unremunerative work
can be regarded in some aspects as worse than not employing him
at all. It is not intended, however—and, indeed, it would be
impossible—here to enter into a discussion on the whole problem of
unemployment, but there is undoubtedly a very great economic
waste that largely contributes to the gravity of the problem, arising
from the fact that a large number of people are being forced to
devote their labour and energy to work which is, so to speak, final
and sterile. It is precisely the same with regard to the production of
expensive luxuries. The employment of a large retinue is only
another form of the possession and enjoyment of articles of
64. excessive luxury. The employers and possessors have all
disagreeable burdens and every sordid worry lifted from them, their
smallest and their most extreme desires for pleasure met, their
special appetites satisfied, their peculiar vanities titillated, and their
artificial position safeguarded and maintained, without their giving
more than a passing thought to the mass of people required to carry
on this work. Plenty of examples might be quoted in contemporary
as well as past history to show that after generations of the
enjoyment of “the vile joys of tainting luxury” men deteriorate, both
physically and mentally.
As for the particular line of life which domestic service offers
under modern circumstances, it is not too much to say that it is, as a
rule, very demoralising, more especially for the men. And its
demoralising tendency increases in proportion to the size of the
establishment. The single general servant lives a life of hard work
but genuine service on four to eight shillings a week, often living in
friendly relations with master or mistress, and really lifting from
them the burden of necessary domestic duties which they with
limited incomes and professional work of their own cannot possibly
find time to perform; and this remains true in other small
households. In the large house the faithful old family servant, who is
more of a friend than a servitor, is rare in these days of ostentation.
The butler, on wages of fifty to sixty shillings a week, which together
with board and lodging represents from £250 to £300 a year, has a
life of leisure, ease, and excessive comfort, seldom having to exert
himself even up to his limited capacities. Male house-servants are
often chosen for their looks; their work is very light physically, they
are overfed, and being under-educated, can hardly be blamed for
becoming demoralised. These able-bodied men, whose muscles, if
not their minds, might be devoted to some really serviceable
purpose, are still increasing in numbers. Over 25,000 more male
servants have got employment in the last ten years, the total
number now being 227,995. Even deducting the single indoor
servant, the single coachman or gardener, this means a large
increase of ornamental male attendants. Female servants are
65. becoming more difficult to secure in the higher grades, because the
class of women from which they are drawn value their liberty and
are not so ready to sacrifice it for food and comforts. In fact, they
are showing signs of impatience of control, and of preferring the
risky though exhilarating struggle of independence. But still large
retinues of men and women exist solely employed in keeping up
huge houses to satisfy the vanity and minister to the comfort of a
comparatively few rich people. No work of a more hopelessly barren,
profitless, and, indeed, degrading character could be found for them.
A system of tips deprives their smallest acts of what might be an
obliging and disinterested intention. Arrangements are organised
with tradesmen to defraud the employers in what is thought a
perfectly legitimate way; the actual waste of food is appalling, and
by extras, gratuities, perquisites, commissions, and pickings a
considerable amount is added to the wages of the upper servants. In
these large establishments immorality exists more as a rule than as
an exception, but it can be kept secret, for these communities of
private servants—like everything else connected with the lives of the
rich—cannot be made the subject of investigation.
If assistance to those who need it is the object of domestic
service, it is striking to note that on the money basis, generally
speaking, the wrong people are served. Who in the community most
require and should specially have the help of servants? The old and
infirm, the weak and ill, the very young and the hard-worked.
Service under such conditions raises itself to the level of one of the
highest occupations that can be imagined. But this is not our
system. A man or woman may be ill, old or over-worked, without
being able to get the assistance of a single soul. Another man or
woman may be young and healthy and have at his or her command
a retinue of thirty servants or more, solely because they have money
and servants are forced, by economic pressure, to devote their lives
to the menial task of furbishing up the endless and complicated
appanage of wealth.
Now let us turn to the inanimate luxuries, taking into account
only indisputable luxuries—that is to say, articles of high price which
66. have no special artistic value, to which much labour has been
devoted and which are not produced to serve any legitimately useful
purpose. Luxury has been well defined as “that which creates
imaginary needs, exaggerates real wants, diverts them from their
true end, establishes a habit of prodigality in Society, and offers
through the senses a satisfaction of self-love which puffs up but
does not nourish the heart and which presents to others the picture
of happiness they can never attain.”
Bond Street catalogues abound with any quantity of examples.
Furs at one thousand guineas, fifty-guinea dressing-bags, twenty-
guinea hats, thousand-guinea tiaras, fruit and vegetables out of
season, cigars at three shillings apiece, ruinously expensive wines,
and fantastic foods of all descriptions. There is no need to
exaggerate, for all those articles can be bought for much higher
prices than those quoted. A great amount of skilled labour of a high
order goes to the production of these luxuries, and a great amount
of labour of the lowest and most cruelly sweated description is also
enlisted for their production, and incredible as it may seem, it is on
the ground that they give employment that these luxuries are
defended. It was calculated in 1884
5
that, even giving a liberal
extension of meaning to the term “necessaries” and “comforts” of
life, over six millions of manual labourers, who with their families
constitute thirteen millions of the population, were engaged in
producing what, in contradistinction to the above, must be classified
as luxuries.
A prominent statesman,
6
expressing the views of his class, said
a few years ago: “The more human wants are stimulated and
multiplied, the more widespread will be the inducement to hire.
Therefore all outcries and prejudices against the progress of wealth
and what is called luxury are nothing but outcries of prejudice
against the very sources and fountains of all employment.”
On such an argument as this the defence of luxuries generally
rests. The essence of the fallacy lies in the fact, which cannot be
repeated too often, that labour spent on such articles is
67. unremunerative and unproductive, because its ultimate result is only
to gratify various forms of vanity and greed. To exemplify by a
concrete instance what is unremunerative and what is remunerative,
let us take a hundred-guinea ball-gown and a pair of boots. It is not
possible to estimate the number of people employed in producing
the ball-gown. There is the silk, satin, or whatever the principal
material may be; there are the trimmings of chiffon, hand-
embroidery, lace, braid, beads, sequins, ribbons, etc., etc.; some
hundreds of pairs of hands, including factory-workers, dressmakers,
sempstresses, etc., will have touched some part of the gown before
it is delivered to the wearer. To what end are all these specialised
departments of labour concentrated? The gown is worn a few times
in the one season; the wearer has the satisfaction of feeling as well
dressed as A. to F., and far better dressed than F. to Z. In fact, the
net result of all this expenditure of energy is the generating of a
rather foolish pride, the encouragement of conceit on the one side
and envy on the other, and the hardening of a nature into ways of
worldliness and vanity.
As for the boots. Again, many more hands than can be
calculated have helped to produce them, but they are directly and
immediately serviceable to the purchaser, to whose activity the
wearing of boots is an essential, and in general they minister to the
efficiency of human machines.
But if balls are not wrong, ball-gowns must be worn. It is a
question of degree; and here again we get to the theory of the limit
which in this conjunction can be expressed thus: In relation to
human needs, in relation to human powers of enjoyment, in relation
to the beneficial effects of pleasure, even in relation to the dictates
of fashion, there is a distinct limit not to be expressed in figures up
to which expenditure (in this particular case on dress) is legitimate
and relatively productive, beyond which it becomes progressively
unremunerative and harmful. A hundred guineas, by any conceivable
method of calculation, greatly exceeds this limit.
68. To assert that the purchase of luxuries is good for trade is quite
as ridiculous as to say that a man can benefit the building and
furnishing trade by burning down his house once a year. We do not
want to create more artificial wants before we have satisfied the
crying human needs which already exist. There is no loophole
through which a reasonable defence of the senseless expenditure,
which goes on in an increasing measure, can be made. Luxurious
living has never been quite so blatant and unashamed as it is to-day,
and the effete epicureanism and decadent effeminacy it produces
stand out in rather sharp contrast to more hopeful signs of progress
and moral and intellectual refinement and vigour which, happily, are
visible around us.
A lady writing in a review in the early ’seventies describes life in
the country house, with its futile routine of heavy meals, sport, card-
playing, and vacuous inanities which take the place of conversation,
all very much as it is to-day. The writer speaks with dismay of gowns
costing sixty guineas and of £1000 a year spent on clothes. But
these figures are almost negligible compared with the sums spent
nowadays. It is only through occasional actions in the courts that the
outside public get an idea of what is actually spent, and it is
surprising that there are not more disclosures, considering the
mountainous debts that are piled up in West End shops. But the
shopkeepers are very reluctant to lose a really leading customer, and
they know how to meet the inconvenience of not being promptly
paid. A typical case may be given of an article of clothing, the cost
price of which was nine guineas, being sold for £28 7s. There may
be delay in payment, but there appears to be compensation in the
profit.
When one hears of the woman who spent last year £36 5s. on a
hat, or another who gave £1250 for a sable cape, it is not the
isolated action of criminal folly that chiefly strikes one, but it is that
the hat and the cape act as indicators of the sort of price such
women are in the habit of paying for their clothes, a large supply of
which are in the market ready to meet this artificial demand.
Moreover, the habit of extravagance, especially as regards female
69. clothing, is catching and runs through all classes once the example is
set. It is a common enough and very depressing sight to see
absurdly elaborate clothes, which are cheaper imitations of the latest
fashions, worn by women of the lower middle-class, whose
deplorable want of education is shown by their inability even to
pronounce their mother tongue. They watch the rich, and gather
from what they see that fine feathers make fine birds, and it is not
on them that the blame should rest.
Vanity exists and insists on being satisfied. It is no good blinking
the fact. Luxuries, in one form or another, will continue to be
produced. But there is no reason why we should not stem the
current lest it swell to danger point. There are many well-known
historical examples of the enervating and degenerating effect of
luxury on national life, and the modern tendency towards an
increased production of these indulgences should be combated not
only as a moral weakness, but as an integral factor in the general
economic problem. When one considers what real comfort of living,
with all the necessary intellectual and artistic equipage, opportunities
for amusement, and domestic convenience, can be secured to-day at
a comparatively moderate sum, it makes the wild and profligate
extravagance the more inexcusable and the more futile.
Anyhow, let us abandon once and for all the foolish and ignorant
attitude of regarding this display as a desirable form of industrial
stimulus which should be fostered and encouraged. Preaching and
writing against it has never been of the smallest avail, but it has
been necessary to deal with it here as a very important, if not
predominating, element in the analysis of the rich man’s conceptions
of his duties.
In addition to luxuries of establishment, clothes, and food there
is a complicated ritual of sport which in this country reaches an
almost incredible pitch. It has been estimated that forty-five millions
are permanently invested in the apparatus of sport, and an income
of over forty millions spent annually upon it. We need not discuss all
the intricacies of the numerous branches of sport, observing where
70. its effect is healthy and where harmful. No one will contend that the
most expensive forms of it are by any manner of means the best.
But the most obvious harm to be noted in this connection is the
amount of land which is taken away from agriculture for sporting
purposes. Landlords often keep up their shooting at a great loss,
amounting to something like five to ten pounds per bird shot, all for
the sake of having the shooting and asking friends down for a few
days in the year to enjoy it. It is gravely regarded as an essential
part of the education of a young man in this particular world to learn
how to shoot. No question, even with respect to his education or
possible professional career, is treated with more seriousness than
the moment he first handles a gun, and family advice is sought as to
how and when encouragement can be given to the development of
this essential qualification which, coupled with a knowledge of
bridge, will make him a desirable visitor in any country house.
At card-playing, which occupies a vast amount of time in the
lives of the rich, sums amounting to hundreds are often lost or
gained by one person in one evening. But of the various sinks which
help to drain away their money, horse-racing almost holds the first
place. There are no statistics to show how many people have been
ruined by it, or how many have been lured into a life of gambling by
their success in the betting ring. But its popularity is certainly on the
increase, as we can see by looking at the number of horses that
have run under the rules of racing in the last thirty years. In 1878
there were 2097; in 1908 this figure had risen to 3706. The number
of larger race meetings advertised in advance have more than
doubled since 1881 (78 in 1881, 164 in 1909). Some sort of estimate
of the money spent on it, apart from betting, can be gathered from
the amounts won. In 1908 the winning owners secured between
them nearly a quarter of a million pounds, the sums won by the first
thirty-six amounting to £246,001 15s., the largest total secured by
one owner being £26,246.
7
The populace are invited to join in this pursuit, though, of
course, they must be railed off to prevent too close contact with
71. those who come in coaches and motor-cars. The crowd is vaguely
supposed to be having a good time, and any attack on horse-racing
is met by hackneyed arguments about “keeping up the national
sport” or “improving the breed of horses,” and perhaps, again, the
objection of unemployment for jockeys and bookies might be
dragged in.
It does not appear, however, to be a good method of improving
the human breed. In observing the crowd on a race-course, whether
it be the well-dressed portion or the ill-dressed, the betters or the
bookies, neither a deep knowledge of humanity nor a very close
power of observation into physiognomy is required to note the
prevalence of a remarkably low type. But a still more vivid
impression of what the pleasures of racing mean can be gained by
going out on the road in the evening towards the scene of a large
race meeting when the people are returning. Brakes and carts in
endless procession will pass you loaded with men shouting in the
excitement of semi-drunkenness, or with heaps of humanity sodden
and silent in complete intoxication. Outside every public-house on
the roadside traps await those who are squandering their gains on
further refreshment or soothing the despair of losses in the
temporary oblivion of drink. The localities where there is an annual
race week suffer considerably, the inhabitants become infected by
the gambling and betting mania, and during the actual days of the
races the place is infested by the lowest dregs of the riff-raff who
journey about from one race meeting to another. This so-called sport
produces the lowest possible type; it degrades many who take part
in it with sinister rapidity, it encourages fraud and deception, it is a
canker of rottenness in public life, and it receives the highest
sanction and patronage.
Many people are present at a race meeting without being
conscious that it is attended by any evil consequences. They go to
meet their friends, perhaps putting an occasional sovereign on a
horse to give them some interest in the racing. To them the crowd is
a natural part of the proceedings, the heavy bets of the ring an
amusement. To have been there is something to boast of, and
72. conveys the idea that they have associated with smart people.
Thoughtless, as in so many of their other pursuits, they accept the
whole proceeding as a recognised sport and they inquire no further.
The philosophy of these people is the prevailing philosophy: “Do not
examine below the surface, or you are bound to find something
disagreeable. Take things as they come; skim the cream off the top;
avoid that which is unpleasant or difficult to explain; and above all
things, do what others do.”
Yachting, which also runs away with a great deal of money,
comes under a very different category. It is a health-giving and often
strenuous occupation, and the seamen employed are, anyhow,
deriving incidentally some positive benefit from the life they lead.
Nevertheless, out of the 4655 private yachts registered in the
current year (an increase of over 3500 in the last forty years),
8
only
a very small proportion are actually navigated by owners who have
any knowledge or love of seamanship. The great majority are
floating houses of luxury (viz. a 700-ton steam yacht, for which £25
a day is paid for coal when in use), or racing yachts, mere toys used
to minister to the fanciful pleasures of the rich.
But in expressing the strongest disapproval of these excessive
luxuries, it is not for a moment suggested that people should rush
into the opposite extreme—live in discomfort and adopt the craze for
“the simple life,” which is only an inverted form of vanity and
ostentation. There are many of the lesser luxuries which give great
pleasure and sufficient honest gratification to justify their existence.
There may even be some reluctance in condemning extravagance,
because the nature of the extravagant man is far preferable to the
economical and cautious disposition which sometimes sinks into
niggardly meanness. Moreover, any attempt at excessive restrictions
and unnecessarily harsh discipline in the upbringing of children
invariably leads to a violent reaction in the direction of profligacy and
extravagance.
Let human nature be allowed free play in all directions; but it is
not taking up the attitude of an ascetic or of a prig to condemn
73. unhesitatingly unnatural excesses, reckless licence, the extremes of
self-indulgence and greed, the exercise of which by some few
involves the neglect, misery, and ruin of so many others.
Thieves when they steal use violence and are pronounced
enemies of society. These few people, by a silent conspiracy in which
we all seem to acquiesce, are also stealing and are equally enemies
of society.
74. W
Chapter IV
The rich man’s charities—His generosity—His hospitality—His
land—The Feudal System—His responsibilities—The
agricultural problem.
E must now turn from what the rich man spends on himself
and consider what good and what harm he does by his
subscriptions and donations to philanthropic and charitable objects.
In so far as he himself is concerned these gifts do not involve
any element of personal sacrifice; the moral benefit which is by way
of falling on a giver is therefore nil. The exertion of writing a cheque
or banker’s order and the satisfaction of imposing a tax on himself
complete the transaction on his side. Occasionally the sight of his
name published at the head of a list with a large figure next it gives
him a further agreeable sensation, and he can become famous as a
household word of generous philanthropy without the very smallest
personal inconvenience. But as an instance of pure charity—that is,
loving sacrifice—the poor woman who gives a penny from her
meagre store is on an entirely different plane. The picture presents
itself to the present writer of a woman at the doorway of a wretched
tenement, with her child in her arms, giving to a passing vagrant
who was suffering from hunger and fatigue a penny from the few
coins she had in her purse. The expression of her face as she
handed him the money was the most sublime illumination of pure
charity—no subscription list in the newspapers, no public
recognition, and the sacrifice, not of luxuries, but of something that
she and her baby needed. That something went with her penny, and
75. in return she received something else for which there is no price, no
name, and no description. From such an experience as this the rich
are for ever cut off. “Probably the most generous people in the
world,” says J. D. Rockefeller, perhaps realising that charity is
something he can never reach, “are the very poor, who assume each
other’s burdens in the crises which come so often to the hard
pressed.”
The rich man’s so-called charity therefore must be to a large
extent mechanical and conventional. He gives because others with
the same means give, and the charity touts know how a list headed
by Lord A. with a substantial sum will produce equally or perhaps
even more substantial sums from Lord B., Sir. C. D., Alderman E.,
and Mr. F. The extraction of money from the rich is a business in
itself, requiring considerable skill, and the rich are fleeced far more
than they realise. In practical America they take the trouble to teach
people professionally how to write what they call “letters of appeal.”
When we hear of subscriptions to charities being stopped it may
serve to remind us that it is most inexpedient that institutions such
as hospitals should be at the mercy of the casual caprice of rich
men. Nothing could eventually be more desirable than that every
one of them should cut off their charitable contributions. It might
entail a severe temporary shock to the funds of charitable
institutions, as over seven millions a year is being spent in London
alone on charities, but at the same time many ill-managed and
misdirected endeavours would disappear, and the State would come
to realise all the sooner its responsibilities in respect to the
maintenance of really necessary institutions for the relief of suffering
and the nursing of the sick, in the same way as it is beginning to
recognise its duties towards poverty, old age, and unemployment.
There are other enterprises which the State should undertake that
are often delayed in their institution owing to the plea that the
private munificence of rich men can be depended upon. It is
certainly better that the funds should be expended thus than in
sheer self-indulgence, but it is evident that the money would be far
better spent and the object on which it is spent better served if the
76. source were not controlled by the whims and fancies of a single
individual.
In regard to the more private and personal aspects of the
generosity attributed to riches: “Surely,” a critic will say, “if the rich
man is benevolent and kindly disposed he can in a hundred
thoughtful ways help his poorer friends by presents, by attention and
timely help, by opening the doors of his houses, lending his
conveyances, and showing many other attentions which his money
allows him to do, thereby becoming justly popular and a source of
great good.”
The admiration, and just admiration, for open-handed generosity
and the justifiable dislike of anything approaching miserliness in
others cause an entirely erroneous impression that large gifts of
money must unquestionably be praiseworthy and commendable. But
this is not the question at issue. These are two moral qualities, the
one admirable, the other objectionable. The generous disposition
can show itself in many other ways besides money gifts, and the real
man behind the rich man, though he may be one and the same
individual, often comes forward with simple acts of thoughtful
kindness because the finer qualities of human nature cannot be
stifled even by money. But in so far as the rich man indulges his
generosity in thoughtlessly giving away money broadcast, it amounts
to a form of self-indulgence, and he is distinctly to blame for not
estimating more precisely the effect of his actions. No doubt the
harm of unwise and foolish actions is palliated by the purity and
excellence of the motive. In so far as these people intend to show
kindness they are amply justified in what they do. But let us consider
for a moment what the effect of their benevolence is. In the first
place they are made to occupy an entirely false position as
dispensers of charity. Often, too, the desire to patronise and gain the
power that patronage gives blights the spirit of genuine and
unadulterated kindness, and further, the recipients are placed in the
extremely uncomfortable and embarrassing situation of receiving
benefits, presents, and comforts which they know they are not and
probably never will be in a position to return. To force anyone to be
77. Welcome to Our Bookstore - The Ultimate Destination for Book Lovers
Are you passionate about books and eager to explore new worlds of
knowledge? At our website, we offer a vast collection of books that
cater to every interest and age group. From classic literature to
specialized publications, self-help books, and children’s stories, we
have it all! Each book is a gateway to new adventures, helping you
expand your knowledge and nourish your soul
Experience Convenient and Enjoyable Book Shopping Our website is more
than just an online bookstore—it’s a bridge connecting readers to the
timeless values of culture and wisdom. With a sleek and user-friendly
interface and a smart search system, you can find your favorite books
quickly and easily. Enjoy special promotions, fast home delivery, and
a seamless shopping experience that saves you time and enhances your
love for reading.
Let us accompany you on the journey of exploring knowledge and
personal growth!
ebookgate.com