Download as pdf or txt
Download as pdf or txt
You are on page 1of 48

Handbook of the Biology of Aging

Nicolas Musi
Visit to download the full and correct content document:
https://ebookmass.com/product/handbook-of-the-biology-of-aging-nicolas-musi/
HANDBOOK OF THE BIOLOGY OF AGING
NINTH EDITION
THE HANDBOOKS OF
AGING
Consisting of Three Volumes

Critical comprehensive reviews of research knowledge,


theories, concepts, and issues

Editors-in-Chief
Laura L. Carstensen and
Thomas A. Rando

Handbook of the Biology of Aging, 9th Edition


Edited by Nicolas Musi and Peter J. Hornsby

Handbook of the Psychology of Aging, 9th Edition


Edited by K. Warner Schaie and Sherry L. Willis

Handbook of Aging and the Social Sciences, 9th Edition


Edited by Kenneth F. Ferraro and Deborah Carr
HANDBOOK OF THE
BIOLOGY OF AGING
NINTH EDITION

Edited by

NICOLAS MUSI
Barshop Institute for Longevity and Aging Studies, South Texas Veterans Health Care System,
University of Texas Health Science Center, San Antonio, TX, United States

PETER J. HORNSBY
Barshop Institute for Longevity and Aging Studies and Department of Cellular and Integrative Physiology,
University of Texas Health Science Center, San Antonio, TX, United States
Academic Press is an imprint of Elsevier
125 London Wall, London EC2Y 5AS, United Kingdom
525 B Street, Suite 1650, San Diego, CA 92101, United States
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom
Copyright © 2021 Elsevier Inc. All rights reserved.
No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording,
or any information storage and retrieval system, without permission in writing from the publisher. Details on how to seek permission, further information
about the Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing
Agency, can be found at our website: www.elsevier.com/permissions.
This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be noted herein).

Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research meth-
ods, professional practices, or medical treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or
experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties
for whom they have a professional responsibility.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons
or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas
contained in the material herein.

British Library Cataloguing-in-Publication Data


A catalogue record for this book is available from the British Library
Library of Congress Cataloging-in-Publication Data
A catalog record for this book is available from the Library of Congress

ISBN: 978-0-12-815962-0

For Information on all Academic Press publications


visit our website at https://www.elsevier.com/books-and-journals

Publisher: Nikki Levy


Editorial Project Manager: Barbara Makinster
Production Project Manager: Swapna Srinivasan
Cover Designer: Matthew Limbert
Typeset by MPS Limited, Chennai, India
Dedication

This volume is dedicated to the memory of Dr. of aging. His early research in the biology of aging
Edward Masoro, editor of the fifth, sixth, and seventh focused on then-popular ideas of how calorie restriction
editions of this handbook; his name will always be in rodents extended lifespan. It was thought that calorie
associated with this important series of volumes. Dr. restriction affected the growth of young animals and that
Masoro passed away on July 11, 2020, at the age of 95. the smaller animals had an extended lifespan. However,
Dr. Masoro obtained his A.B. degree at the University he showed that calorie restriction worked in adult ani-
of California at Berkeley in 1947 and was then awarded mals. His subsequent work tested many physiological
the PhD degree in physiology in 1950. Following early hypotheses about the mechanisms of calorie restriction
faculty positions, he was appointed as the Founding (Masoro, 2009). Investigations into this topic continue to
Chair of the Department of Physiology at the newly this day, and work on the mechanisms of calorie restric-
established medical school of the University of Texas tion has stimulated new areas of research, such as phar-
system in San Antonio (see Chen, 2003, and Critser, macological interventions that act as calorie restriction
2010, for accounts of his early career). He persuaded mimetics.
other faculty members of the department to join him in a As members of the Barshop Institute, we will
new line of research that probed the mechanisms of always be grateful to Dr. Masoro for his work on the
aging in rodents, a line of research that began to put San basic research in the biology of aging at San Antonio,
Antonio on the map as a major center for studies in the and we recognize that the current thriving enterprise
basic biology of aging. He established the Aging in aging research here would never have been possible
Research and Education Center of the University of without his pioneering efforts.
Texas Health Science Center San Antonio to catalyze
research on the biology of aging across basic and clinical
science disciplines. The culmination of this process was
References
the founding of the Barshop Institute for Longevity and Chen, I. (2003). Hungry for science. Science of Aging Knowledge
Aging Studies, one of the first centers in the nation dedi- Environment, 2003(1), NF1, 8 January.
Critser, G. (2010). Eternity soup: Inside the quest to end aging. New York:
cated to basic research in the biology of aging.
Crown Publishing.
Dr. Masoro’s seminal work established the paradigm Masoro, E. J. (2009). Caloric restriction-induced life extension of rats
of extension of rodent lifespan by caloric restriction, and mice: A critique of proposed mechanisms. Biochimica et
which became a cornerstone of research into mechanisms Biophysica Acta, 10, 10401048.
This page intentionally left blank
Contents

List of contributors xi 4. The nature of aging and the geroscience


About the editors xiii hypothesis 69
Foreword xv PETER J. HORNSBY
Preface xvii
Aging and the geroscience hypothesis 69
What is aging and why does aging occur? 69
Immortal versus mortal systems and entities 70
I The disposable soma concept 71
Basic mechanisms, underlying What are the factors that determine longevity? 72
The force of natural selection over the lifespan 74
physiological changes, model organisms Genes that exhibit antagonistic pleiotropy 74
and interventions The molecular mechanisms of aging and the geroscience
hypothesis 74
Conclusions 75
1. Longevity as a complex genetic trait 3 References 75
GEORGE L. SUTPHIN AND RON KORSTANJE
5. Sirtuins, healthspan, and longevity in mammals 77
Introduction 3
SURINDER KUMAR, WILLIAM GIBLIN AND DAVID B. LOMBARD
Defining the aging gene-space 4
Nongenetic sources of complexity 17
Introduction 78
Emerging tools for studying complex genetic traits 22
Sirtuin-driven lifespan extension in invertebrates 78
Conclusions 31
Sirtuin enzymatic activity 79
References 31
Sirtuins and mammalian longevity 83
Genetic variants of human sirtuin genes 86
2. DNA damage and repair in aging 43 Sirtuins as modulators of responses to caloric restriction 91
AJINKYA S. KAWALE AND PATRICK SUNG
Roles for sirtuins in diverse disease states 94
Cancer 94
Introduction 43 Metabolic syndrome 104
Overview of the DNA damage response 43 Cardiovascular dysfunction 111
Linkage of DNA repair mechanisms to aging 44 Inflammatory signaling 116
DNA damage theory of aging 46 Neurodegenerative disease 118
Aging disorders associated with DNA damage 47 Pharmacological modulation of sirtuin activity 121
Conclusions and future perspectives 49 Conclusions 125
Acknowledgments 50 Acknowledgments 126
References 50 References 127

3. Mechanisms of cell senescence in aging 53 6. Integrative genomics of aging 151


THOMAS VON ZGLINICKI JOÃO PEDRO DE MAGALHÃES, CYRIL LAGGER AND ROBI TACUTU

What is senescence? Building blocks and feedback loops 53 Introduction 151


The building blocks of the senescent phenotype 55 Post-genome technologies and biogerontology 152
Terminology 59 Challenges in data analysis 157
Senescence during aging in vivo 59 Data integration 157
Senolytics and senostatics as antiaging interventions 61 Concluding remarks 165
Conclusions 63 Acknowledgments 165
References 63 References 165

vii
viii Contents

7. Thermogenesis and aging 173 Types of intervention proposals sought by the Interventions
JUSTIN DARCY, YIMIN FANG, SAMUEL MCFADDEN,
Testing Program 222
KEVIN HASCUP, ERIN HASCUP AND ANDRZEJ BARTKE Challenges encountered implementing testing
protocols 223
Introduction 173 Summary of Interventions Testing Program findings 224
Thermogenic adipose tissue 174 Studies nearing completion 230
Thermogenesis, energy metabolism, and aging 174 Pathology of drug-treated mice 230
Regulation of thermogenesis in brown adipose tissue 175 Collaborative Interactions Program 230
Impact of environmental temperature on laboratory mice 176 Tests of health outcomes 231
Concluding remarks 177 Interactions with the Mouse Phenome Database 231
References 178 The ITP at 15 years: synopsis and future goals 231
References 233
8. Yeast as a model organism for aging research 183 Further reading 235
ANITA KRISKO AND BRIAN K. KENNEDY

Introduction 183 11. Aging in nonhuman primates 237


Asymmetric cell division in yeast 184
SUZETTE D. TARDIF AND CORINNA N. ROSS
Emergence of yeast as a model organism in aging research 185
Types of aging in yeast 185 Introduction 237
Replicative lifespan 186 Why do primates live so long? 238
Alternative methods to measure replicative lifespan 186 Aging by domain 238
Mother enrichment program 186 Manipulation of aging 242
Microfluidics in the research into aging in yeast 187 References 244
Chronological lifespan 187
Replicative lifespan versus chronological lifespan 188
Hallmarks of aging and yeast 189
Epigenetic alterations in yeast aging 189
Calorie restriction in yeast 190 II
Adenosine monophosphate kinase 192
Loss of proteostasis 192
Organ systems in humans and
Systems biology and yeast aging 193 other animals, human health
Relevance of yeast in human aging and the future 193 and longevity
Acknowledgments 194
References 194
12. Senotherapeutics: Experimental therapy
9. Model organisms (invertebrates) 199
of cellular senescence 251
ERIN MUNKÁCSY AND ANDREW M. PICKERING
JAMIE N. JUSTICE, LAURA J. NIEDERNHOFER AND
MIRANDA E. ORR
Introduction 199
Caenorhabitis elegans 200 Premise for translational research and therapeutically
Drosophila melanogaster 203 targeting cellular senescence 252
Other invertebrate models in aging research 208 Cellular senescence and the senescence-associated secretory
Rotifers 208 phenotype 258
Flatworms 211 Cell senescence as a therapeutic target for age-related
Bivalves 211 diseases 259
Sea urchins 212 Biological markers of cellular senescence 260
Hydra 213 Experimental therapy for cellular senescence 261
Turritopsis dohrnii—“the immortal jellyfish” 214 Senolytics: Foundation and discovery 261
References 214 Fisetin 263
Senotherapeutics: Broader definitions and deeper
10. NIA Interventions Testing Program: understanding 264
A collaborative approach for investigating Other senotherapeutics 265
interventions to promote healthy aging 219 Senotherapeutics and the brain 265
FRANCESCA MACCHIARINI, RICHARD A. MILLER,
Senomorphics 267
RANDY STRONG, NADIA ROSENTHAL AND DAVID E. HARRISON Senomorphic and senotherapeutic actions of
geroscience-guided interventions 268
Introduction 219 Clinical translation of senotherapeutics 269
Features of the Interventions Testing Program experimental Conclusions 274
design 220 References 274
Contents ix
13. The role of neurosensory systems in the Impact of thymic involution and thymectomy
modulation of aging 285 on T cells 352
Immune cell function 355
GUANG YANG, YI SHENG AND RUI XIAO
T-cell function 355
Introduction 285 B-cell function 355
Sensations that regulate animal lifespan 286 Clinical consequences of immunosenescence 356
Signaling pathways involved in neurosensory modulation Effect of age on vaccination 357
of aging 289 Immune senescence and all-cause mortality 358
Opportunities and challenges in studying neurosensory Interventions to restore appropriate immunity 359
modulation of aging 290 Perspectives 360
Concluding remarks 291 References 361
Acknowledgments 292
References 292
17. Microbiome changes in aging 367
KELLY R. REVELES, ERIC H. YOUNG,
14. Aging of the sensory systems: hearing AMINA R.A.L. ZEIDAN AND QUNFENG DONG
and vision disorders 297
Introduction 367
SHINICHI SOMEYA AND AKIHIRO IKEDA
Overview of the structure and function of the microbiome 368
Relationship between the microbiome and aging 370
Introduction 297
Contribution of the microbiome to aging-related conditions 375
Aging of the auditory system 298
Microbiome-targeted therapies as potential antiaging
Aging of the visual systems 303
interventions 379
Conclusions 310
Conclusions 383
Abbreviations 312
References 383
References 312

15. Cardiac aging 323 18. Lipidomics of aging 391


YING ANN CHIAO, DAO-FU DAI, ROBERT J. WESSELLS AND JUAN PABLO PALAVICINI AND XIANLIN HAN
PETER S. RABINOVITCH
Introduction 391
Introduction 323 Lipids in biology of aging 392
Cardiac aging in humans 324 Acknowledgments 399
Cardiac aging in the mouse model 326 References 399
Other models of cardiac aging 326
Molecular mechanisms of cardiac aging 328
Interventions for cardiac aging 334 19. Trends in morbidity, healthy life expectancy,
Concluding remarks and future perspectives 337 and the compression of morbidity 405
References 337
EILEEN M. CRIMMINS, YUAN S. ZHANG, JUNG KI KIM AND
MORGAN E. LEVINE

16. The aging immune system: Dysregulation, Introduction 405


compensatory mechanisms, and prospects for Dimensions of morbidity 405
intervention 345 The length of life cycles and population health 406
LUDMILA MÜLLER AND GRAHAM PAWELEC Trends in population prevalence of physiological
dysregulation, diseases and conditions, functioning
Introduction 345 loss and disability, and life expectancy 406
Innate and adaptive immunity 346 Length of life and length of healthy life 409
Age and immunity 346 Conclusions 412
Effect of age on hematopoiesis 348 References 413
Effect of age on innate immunity 350 Further reading 414
Natural killer cells 351
Dendritic cells 351 Author Index 415
Effect of age on adaptive immunity 352 Subject Index 441
This page intentionally left blank
List of contributors

Andrzej Bartke Southern Illinois University School of and Geriatric Medicine, Wake Forest School of Medicine
Medicine, Department of Internal Medicine, Springfield, (WFSM), Winston-Salem, NC, United States
IL, United States Ajinkya S. Kawale Department of Biochemistry and
Ying Ann Chiao Aging and Metabolism Research Program, Structural Biology, University of Texas Health Science
Oklahoma Medical Research Foundation, Oklahoma City, Center at San Antonio, San Antonio, TX, United States
OK, United States Brian K. Kennedy Department of Biochemistry and
Eileen M. Crimmins Davis School of Gerontology, Physiology, Yong Loo School Lin School of Medicine,
University of Southern California, Los Angeles, CA, National University of Singapore, Singapore, Singapore;
United States Centre for Healthy Longevity, National University Health
Dao-Fu Dai Department of Pathology, Carver College of System, Singapore, Singapore; Singapore Institute for
Medicine, University of Iowa, Iowa City, IA, United States Clinical Sciences, A*STAR, Singapore, Singapore
Justin Darcy Section on Integrative Physiology and Jung Ki Kim Davis School of Gerontology, University of
Metabolism, Joslin Diabetes Center, Harvard Medical Southern California, Los Angeles, CA, United States
School, Boston, MA, United States Ron Korstanje The Jackson Laboratory, Bar Harbor, ME,
João Pedro de Magalhães Integrative Genomics of Ageing United States
Group, Institute of Ageing and Chronic Disease, Anita Krisko Department of Experimental Neurodegeneration,
University of Liverpool, Liverpool, United Kingdom University Medical Center Goettingen (UMG), Goettingen,
Qunfeng Dong Department of Public Health Sciences, Germany
Loyola University Chicago, Chicago, IL, United States Surinder Kumar Department of Pathology, University of
Yimin Fang Southern Illinois University School of Michigan, Ann Arbor, MI, United States
Medicine, Department of Neurology, Springfield, IL, Cyril Lagger Integrative Genomics of Ageing Group,
United States Institute of Ageing and Chronic Disease, University of
William Giblin Department of Pathology, University of Liverpool, Liverpool, United Kingdom
Michigan, Ann Arbor, MI, United States Morgan E. Levine Department of Pathology, Yale
Xianlin Han Barshop Institute for Longevity and Aging University School of Medicine, New Haven, CT, United
Studies, University of Texas Health Science Center at San States
Antonio, San Antonio, Texas, United States; Division of David B. Lombard Department of Pathology, University of
Diabetes, Department of Medicine, University of Texas Michigan, Ann Arbor, MI, United States; Institute of
Health Science Center at San Antonio, San Antonio, Gerontology, University of Michigan, Ann Arbor, MI,
Texas, United States United States
David E. Harrison The Jackson Laboratory, Bar Harbor, Francesca Macchiarini Division of Aging Biology, National
ME, United States Institute on Aging, Bethesda, MD, United States
Erin Hascup Southern Illinois University School of Samuel McFadden Southern Illinois University School of
Medicine, Department of Neurology, Springfield, IL, Medicine, Department of Neurology, Springfield, IL,
United States United States
Kevin Hascup Southern Illinois University School of Richard A. Miller Department of Pathology and Geriatrics
Medicine, Department of Neurology, Springfield, IL, Center, University of Michigan, Ann Arbor, MI, United
United States States
Peter J. Hornsby University of Texas Health Science Center Ludmila Müller Department Lifespan Psychology, Max
San Antonio, San Antonio, TX, United States Planck Institute for Human Development, Berlin,
Akihiro Ikeda Department of Medical Genetics and Germany
McPherson Eye Research Institute, University of Erin Munkácsy Barshop Institute for Longevity and Aging
Wisconsin, Madison, WI, United States Studies, UT Health, San Antonio, TX, United States;
Jamie N. Justice Sticht Center for Healthy Aging and Department of Molecular Medicine, UT Health, San
Alzheimer’s Prevention, Internal Medicine  Gerontology Antonio, TX, United States

xi
xii List of contributors

Laura J. Niedernhofer Institute on the Biology of Aging Shinichi Someya Department of Aging and Geriatric
and Metabolism, Department of Biochemistry, Molecular Research, University of Florida, Gainsville, FL, United States
Biology and Biophysics, University of Minnesota, Randy Strong Department of Pharmacology, The
Minneapolis, MN, United States University of Texas Health Science Center at San Antonio,
Miranda E. Orr Sticht Center for Healthy Aging and and the Geriatric Research, Education and Clinical Center
Alzheimer’s Prevention, Internal Medicine  Gerontology (GRECC) and Research Service of the South Texas
and Geriatric Medicine, Wake Forest School of Medicine Veterans Health Care System, Texas, TX, United States
(WFSM), Winston-Salem, NC, United States; W.G. Hefner Patrick Sung Department of Biochemistry and Structural
Veterans Affairs Medical Center, Salisbury, NC, United Biology, University of Texas Health Science Center at San
States Antonio, San Antonio, TX, United States
Juan Pablo Palavicini Barshop Institute for Longevity and George L. Sutphin University of Arizona, Tucson, AZ,
Aging Studies, University of Texas Health Science Center United States
at San Antonio, San Antonio, Texas, United States;
Robi Tacutu Systems Biology of Aging Group, Department
Division of Diabetes, Department of Medicine, University
of Bioinformatics and Structural Biochemistry, Institute of
of Texas Health Science Center at San Antonio, San
Biochemistry of the Romanian Academy, Bucharest,
Antonio, Texas, United States
Romania
Graham Pawelec Department of Immunology, University
Suzette D. Tardif Population Health Program, Southwest
of Tübingen, Tübingen, Germany; Health Sciences North
National Primate Research Center, Texas Biomedical
Research Institute, Sudbury, Ontario, Canada
Research Institute, San Antonio, TX, United States
Andrew M. Pickering Barshop Institute for Longevity and
Thomas von Zglinicki Newcastle University Biosciences
Aging Studies, UT Health, San Antonio, TX, United
Institute, Campus for Ageing and Vitality, Newcastle
States; Department of Molecular Medicine, UT Health,
University, Newcastle upon Tyne, United Kingdom
San Antonio, TX, United States; Center for
Neurodegeneration and Experimental Therapeutics, Robert J. Wessells Department of Physiology, Wayne State
Department of Neurology, The University of Alabama at University School of Medicine, Detroit, MI, United States
Birmingham, Birmingham, AL, United States; Department Rui Xiao Department of Aging and Geriatric Research,
of Neurobiology, The University of Alabama at Institute on Aging, University of Florida, Gainesville, FL,
Birmingham, Birmingham, AL, United States United States; Department of Pharmacology and
Peter S. Rabinovitch Department of Laboratory Medicine Therapeutics, College of Medicine, University of Florida,
and Pathology, University of Washington, Seattle, WA, Gainesville, FL, United States; Center for Smell and Taste,
United States University of Florida, Gainesville, FL, United States
Kelly R. Reveles College of Pharmacy, The University of Guang Yang Department of Aging and Geriatric Research,
Texas at Austin, Austin, TX, United States; Institute on Aging, University of Florida, Gainesville, FL,
Pharmacotherapy Education & Research Center, UT United States
Health San Antonio, San Antonio, TX, United States Eric H. Young College of Pharmacy, The University of
Nadia Rosenthal The Jackson Laboratory, Bar Harbor, ME, Texas at Austin, Austin, TX, United States;
United States Pharmacotherapy Education & Research Center, UT
Corinna N. Ross Population Health Program, Southwest Health San Antonio, San Antonio, TX, United States
National Primate Research Center, Texas Biomedical Amina R.A.L. Zeidan College of Pharmacy, The University
Research Institute, San Antonio, TX, United States; of Texas at Austin, Austin, TX, United States;
Department of Life Sciences, Texas A&M University San Pharmacotherapy Education & Research Center, UT
Antonio, San Antonio, TX, United States Health San Antonio, San Antonio, TX, United States
Yi Sheng Department of Aging and Geriatric Research, Yuan S. Zhang Carolina Population Center, The University
Institute on Aging, University of Florida, Gainesville, FL, of North Carolina at Chapel Hill, Chapel Hill, NC, United
United States States
About the editors

Dr. Nicolas Musi is a tenured Professor of as Director of a T32 Training Grant on the Biology
Medicine (Division of Geriatrics and Gerontology of Aging.
and Division of Diabetes) and Director of the
Barshop Institute for Longevity and Aging Studies Dr. Peter J. Hornsby obtained a PhD in Cell Biology
and the San Antonio Claude D. Pepper Older at the Institute of Cancer Research of the University of
Americans Independence Center. He is also London. He has held faculty positions at the University
Associate Director for Research of the San Antonio of California San Diego, the Medical College of Georgia,
Geriatric Research, Education and Clinical Center. and Baylor College of Medicine. Currently he is
He is an active educator and research mentor, and Professor in the Department of Physiology and Barshop
supervises clinical and research fellows, residents, Institute for Longevity and Aging Studies, University of
and graduate students. In this role, he also functions Texas Health Science Center, San Antonio.

xiii
This page intentionally left blank
Foreword

Since the inaugural publication of the Handbooks of experience of aging. The handbooks also provide
Aging in 1976, the series has played a key role in pro- cutting-edge updates to the understanding of genetics,
moting and guiding gerontological science. By preserv- built environments, and intergenerational commit-
ing foundational knowledge and illuminating ments. The 9th edition of the Handbook of the Biology of
emerging areas, the series has served as a core Aging introduces geroscience, a discipline that did not
resource for established researchers and an inspiration exist 10 years ago and is now among the most vibrant
for students of gerontology. From its inception, geron- in all of science. This edition also provides updates on
tological science has been cross-disciplinary. The three- the exciting advances in the genetics and integrative
volume series has played a key role in maintaining genomics of aging and longevity as well as the biology
cohesion in a science that spans dozens of disciplines. and therapeutic opportunities afforded by the studies
The need to understand aging only increases in of cellular senescence.
importance over time. The global population has now What has not changed over the editions is the
passed an important tipping point, moving from a superb synthesis of the field. The editors of the 9th edi-
world where children predominate to one in which tion extend a long tradition of giants in the field giving
there are more older people than youth. This reshap- generously of their time and knowledge to produce
ing of the age distribution in the population demands consistently excellent volumes. Their thoughtful selec-
grand investments in the science of aging. tion of topics and recruitment of deeply knowledge-
Thankfully, the science of aging is also growing fas- able authors is reflected throughout the series. We are
ter than ever across social and biological sciences. most grateful to Nicolas Musi and Peter J. Hornsby,
Along with phenomenal advances in the understand- editors of the Handbook of the Biology of Aging, Kenneth
ing of the biology of aging as well as genetic influences F. Ferraro and Deborah S. Carr, editors of the Handbook
on aging trajectories, and susceptibility to age-related of Aging and the Social Sciences, and K. Warner Schaie
diseases has come the awareness of the critical impor- and Sherry Lynn Willis, editors of the Handbook of the
tance of the physical and social environments in which Psychology of Aging.
people age and the psychological factors that modulate We also express our deep appreciation to our pub-
and sometimes alter genetic predispositions. lishers at Elsevier, whose profound interest and dedi-
The Handbooks of Aging series, comprised of the cation to the topic has facilitated the publication of the
Handbook of the Biology of Aging, the Handbook of Handbooks through many editions. We remain eternally
the Psychology of Aging, and the Handbook of Aging and grateful to James Birren, for establishing the series and
the Social Sciences, is now in its ninth edition. The shepherding it through the first six editions that
Handbook of Aging and the Social Sciences and the played a profound role in establishing the tradition of
Handbook of the Psychology of Aging have long provided multidisciplinary science in the field of aging.
conceptual anchors and frameworks to the social and
behavioral sciences while also addressing emerging
topics that did not exist decades ago, such as the fluid- Thomas A. Rando and Laura L. Carstensen
ity of race and gender, groundbreaking insights into
Stanford Center on Longevity, Stanford University,
the role of sleep in cognitive aging, and the ways that
Stanford, CA, United States
smartphones, robots, and social media can modify the

xv
This page intentionally left blank
Preface

As this series of volumes enters its 9th edition, it biology. As the field has matured, it has become
may well be said that the field of the biology of aging possible to apply biological findings to human
has reached its maturity. There are several themes that patients, either in clinical trials or clinical practice.
are reflected in the contents of this volume. Translational research has also assumed a much
greater prominence. Basic biology has benefited
1. In the past—now in reality the distant past—the
from these clinical and translational investigations.
biology of aging was a separate field of research,
As results have emerged from those studies, they
relatively unconnected to the mainstream of
have stimulated new avenues of investigation in
biological investigation. This has changed radically.
basic biology. For example, studies on interventions
Aging is now incorporated into a broad range of
that may increase rodent life span have already had
related fields, including cancer, diabetes and other
a major impact on clinical and translational
metabolic diseases, immunology, and more. This
investigation, and in turn those studies have
has changed the way research in aging is viewed in
impacted new research in basic biology. mTOR
many ways, and the benefits have been mutual on
inhibitors and senolytics are examples of
both sides. Research in aging has benefited from the
pharmacological interventions that increase rodent
input from these related areas, while those related
life span and have already been translated to the
areas have benefited from concepts developed in
clinic. Findings from clinical and translational
the biology of aging. Just one example is cellular
studies have in turn stimulated new avenues in
senescence. Once considered a topic of narrow
basic biology. We can expect this trend to continue
interest to a small group of investigators in technical
in the future.
aspects of cell culture, it has now been incorporated
into multiple other areas, including cancer and One of the more unfortunate consequences of these
metabolic diseases, as well as aging itself. exciting developments is that it becomes impossible
2. The second development reflected in the chapters in to produce a volume in the Handbook of the Biology of
this volume is the degree to which the biology of Aging series that comprehensively covers all aspects
aging has been incorporated into the basic biology of the topic. The selection of topics offered in the cur-
of each organ system. For example, the basic rent volume is an attempt to sample many of these
biology of bone and the musculoskeletal system is exciting areas with contributions from leading scien-
incomplete without a consideration of the changes tists in their fields. But we also offer in advance our
that take place in aging. Those include changes in apologies to readers whose particular areas of interest
the endocrine system, as well as stem cell biology. we had to omit. Omission does not indicate that we
No account of the complete biology of any organ felt the area of less importance, but we simply face
system is valid unless it incorporates the biology of the reality of an incredibly expanding and increas-
aging as an integral part. ingly exciting field.
3. The third significant development is the merging of
translational and clinical research with basic Nicolas Musi and Peter J. Hornsby

xvii
This page intentionally left blank
P A R T I

Basic mechanisms, underlying


physiological changes, model organisms
and interventions
This page intentionally left blank
C H A P T E R

1
Longevity as a complex genetic trait
George L. Sutphin1 and Ron Korstanje2
1
University of Arizona, Tucson, AZ, United States 2The Jackson Laboratory, Bar Harbor, ME, United States

O U T L I N E

Introduction 3 High-throughput life span assays in yeast and


worms 23
Defining the aging gene-space 4
Genome-scale mouse knockout collection 25
Direct screens for genetic longevity determinants 4
Collaborative cross and diversity outbred mice 26
Leveraging genetic diversity to identify aging loci 9
Expression QTLs 28
Nongenetic sources of complexity 17 Aging biomarkers 29
Tissue-specific aging 17
Conclusions 31
Geneenvironment interaction 18
References 31
Emerging tools for studying complex genetic traits 22

Introduction treatment of specific ailments. These include the mecha-


nistic target of rapamycin (mTOR)-inhibitor rapamycin,
Complex traits are phenotypic characteristics that the sirtuin activator resveratrol, and the glucose produc-
result from the integration of many genetic loci and envi- tion suppressor metformin, and are discussed in greater
ronmental factors. Longevity, along with the age- detail in other chapters. Extragenetic but organism-
dependent decline in cellular and physiological processes intrinsic factors, such as tissue-specific gene expression,
that define aging, is quintessentially a complex genetic parentally inherited molecules, and epigenetics can also
trait. A complete understanding of a complex trait contribute to aging phenotypes.
requires both defining the range of factors that contribute Many environmental factors have been identified
to the trait and developing models for how the various that impact longevity and age-associated disease.
factors interact. In the past several decades, hundreds of These include the abundance and composition of diet,
genes have been identified that are capable of influencing exposure to various forms of stress, environmental
longevity or other age-associated phenotypes across a temperature, social interaction, and even the presence
range of model systems. The majority of these genes can or absence of a magnetic field. Among these, dietary
be broadly assigned to one or more of the following restriction is by far the most widely studied. Reduction
genetic pathways: (1) protein homeostasis, (2) insulin/ in total dietary intake or a change in the composition
IGF-1-like signaling (IIS), (3) mitochondrial metabolism, in the diet can have a profound impact on longevity in
(4) sirtuins, (5) chemosensory function, or (6) dietary model systems. Short-term exposure to thermal, oxida-
restriction. Pharmacologic agents targeting several of tive, endoplasmic reticulum (ER), or other forms of
these pathways have been shown to increase lifespan and stress is sufficient to increase lifespan. In both worms
improve outcomes in age-associated disease in model sys- and fruit flies, adjusting the culture temperature can
tems and are either in use or in clinical trials for dramatically influence lifespan. In each case, genes

Handbook of the Biology of Aging.


DOI: https://doi.org/10.1016/B978-0-12-815962-0.00001-9 3 Copyright © 2021 Elsevier Inc. All rights reserved.
4 1. Longevity as a complex genetic trait

have been identified that mediate the organism’s be completed in a matter of months (Linford, Bilgir, Ro, &
response to the environmental stimuli. Pletcher, 2013).
This chapter examines aging as a complex trait. The
following sections review past and ongoing efforts to
define the scope of genetic, extragenetic, and environ- RNA interference screens in nematodes
mental factors that influence aging, outline strategies In C. elegans, targeted gene knockdown by RNA inter-
for building interaction models, and discuss emerging ference (RNAi) can be accomplished by feeding animals
tools that are furthering our ability to encompass the bacteria expressing double-stranded RNA containing the
complexities of aging. target sequence (Timmons & Fire, 1998). Two RNAi feed-
ing libraries targeting individual genes throughout the C.
elegans genome have been constructed and are commer-
cially available. The original Ahringer library contains
Defining the aging gene-space 16,256 unique clones constructed by cloning genomic
fragments targeting specific genes between two inverted
A primary task in understanding the genetic complex- T7 promoters (Fraser et al., 2000; Kamath et al., 2003).
ity underlying any highly integrative phenotype is to iden- This library has recently been supplemented with an
tify the range of genes capable of impacting that additional 3507 clones. The complete Ahringer library is
phenotype. Three approaches are commonly employed to commercially available through Source Bioscience (RNAi
uncover novel aging factors. In models where targeted Resources | Source BioScience, n.d.). The Vidal library
genome-scale genetic manipulation is possible and lifespan contains 11,511 clones produced using a full-length open
can be measured in a moderate- to high-throughput man- reading frames (ORFs) gateway cloned into a double T7
ner, screens have been carried out to identify single-gene vector (Rual et al., 2004) and is commercially available
manipulations capable of enhancing longevity. In longer- through either Source Bioscience (RNAi Resources |
lived models and those less amenable to high-throughput Source BioScience, n.d.) or Dharmacon Inc. (C. elegans |
targeted genetics, genetic mapping strategies are used to Dharmacon, 2019). Combined, these libraries provide
identify genetic loci at which natural variation is associ- single-gene clones targeting more than 20,000 unique
ated with differences in lifespan. A third approach is to sequences covering approximately 90% of known ORFs
leverage a secondary phenotype, such as stress resistance, in C. elegans.
that correlates with longevity but can be more rapidly In total, more than 300 C. elegans genes have been
screened to narrow the candidate gene list, and only identified for which reducing expression results in
screen genes that pass the primary threshold for longevity. prolonged lifespan (Braeckman & Vanfleteren, 2007;
Smith et al., 2008), the majority in longevity screens
using the RNAi feeding libraries (reviewed by Yanos,
Bennett, & Kaeberlein, 2012) or random mutagenesis
Direct screens for genetic longevity
screens (de Castro, Hegi de Castro, & Johnson, 2004;
determinants Muñoz & Riddle, 2003) (Table 1.1). These include three
Among models commonly used in aging research, the genome-wide screens using the Ahringer RNAi feed-
nematode Caenorhabditis elegans and the budding yeast ing library (Hamilton et al., 2005; Hansen, Hsu, Dillin,
Saccharomyces cerevisiae possess three characteristics allow- & Kenyon, 2005; Samuelson, Klimczak, Thompson,
ing for large-scale genetic screening for longevity: (1) Carr, & Ruvkun, 2007), two partial screens targeting
genetic tools allowing for targeted genome-scale manipu- genes on specific chromosomes (Dillin et al., 2002;
lation of individual genes, (2) relatively short lifespans, Lee et al., 2003), five screens of RNAi clones or mutant
and (3) techniques to rapidly and inexpensively culture sets selected in a preliminary screen for a secondary
large populations in the laboratory. Complete genome longevity-associated phenotype, such as arrested devel-
sequences are available for both organisms (C. elegans opment, upregulation of the mitochondrial unfolded pro-
Sequencing Consortium, 1998; Goffeau et al., 1996) and tein response, or resistance to thermal or oxidative stress
standardized lifespan assays can be completed in a matter (Bennett et al., 2017; Chen, Pan, Palter, & Kapahi, 2007;
of weeks (Murakami & Kaeberlein, 2009; Steffen, Curran & Ruvkun, 2007; de Castro et al., 2004; Kim &
Kennedy, & Kaeberlein, 2009; Sutphin & Kaeberlein, Sun, 2007; Muñoz & Riddle, 2003), and one recent screen
2009). Both models have been used in genome-scale of C. elegans orthologs of human genes differentially
screens for single-gene manipulations capable of increas- expressed at different ages in human whole blood
ing lifespan. In Drosophila melanogaster, while targeted (Sutphin et al., 2017). Combined, these studies have iden-
gene-modification is not available at the genome-scale, tified aging factors in a range of biological processes
random mutagenesis screens are used to identify novel including mitochondrial metabolism, mitochondrial
longevity determinants and lifespan assays can similarly unfolded protein response, cell structure, cell surface

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Defining the aging gene-space 5
TABLE 1.1 Invertebrate longevity screens
Study Primary gene selection No. No. genes Epistasis Functional groups identified
genes identified tested
tested

Worm lifespan
Dillin et al. Chr. I, Ahringer RNAi Library 2445 41a daf-2, daf-16 Metabolism, mitochondrial metabolism
(2002)
Lee et al. Chr. I & II, Ahringer RNAi Library 5690 52 1 b daf-16 Mitochondrial function, metabolism, gene
(2003) expression, protein homeostasis, signal
transduction, stress response

Hamilton Whole genome, Ahringer RNAi 16,475 89 daf-16, sir-2.1 Metabolism, signal transduction, protein
et al. (2005) Library homeostasis, gene expression
Hansen et al. Whole genome, Ahringer RNAi 13,417 29 daf-2, daf-12, Signal transduction, stress response, gene
(2005) Library daf-16, eat-2, expression, mitochondrial metabolism
glp-1
Samuelson Whole genome, Ahringer RNAi 16,757 115 daf-16 Metabolism, mitochondrial function, lysosomal
et al. (2007) Library functions, genomic stability, stress resistance
Chen et al. Developmental arrest (see Kamath 57 24 daf-2, daf-2 daf- Mitochondrial function, metabolism, protein
(2007) et al. (2003)) 16, eat-2 homeostasis, transcription
Curran and Developmental arrest, Ahringer 2700 64 daf-16 Protein homeostasis, signal transduction,
Ruvkun (2007) RNAi Library transcription, mitochondrial function, RNA
processing, chromatin binding factors

Muñoz and Thermal stress resistance, EMS 63 49 daf-16 Signal transduction, stress resistance
Riddle (2003) mutagenesis
de Castro Oxidative stress resistance 6 4 daf-16 Stress resistance
et al. (2004) (juglone), transposon-mediated
mutagenesis
Kim and Sun Oxidative stress resistance 608 84 daf-16 Stress resistance, cell structure, signal
(2007) (paraquat), Chr. III & IV, Ahringer transduction, metabolism, protein homeostasis,
RNAi Library transcription, chromatin binding, mitochondrial
function
Sutphin et al. Orthologs of human genes 82 50 daf-16, eat-2, Metabolism, calcium homeostasis, protein
(2017) differentially expressed with age in hif-1, rsks-1, homeostasis, signal transduction
whole blood sir-2.1
Yeast replicative lifespan
Kaeberlein Random, ORF Deletion Collection 564 13 Protein homeostasis, metabolism, DNA
et al. (2005) replication
Smith et al. Orthologs of worm pro-aging genes 264 25 Protein homeostasis, transcription, mitochondrial
(2008) function
Steffen et al. Ribosomal proteins 31 11 Protein homeostasis, metabolism
(2012)
McCormick Ribosomal proteins 4698 238 Protein homeostasis, mitochondrial function,
et al. (2015) metabolism, DNA replication
Yeast chronological lifespan
Powers et al Whole genome, ORF Deletion B4800 90 1 c Protein homeostasis, stress resistance
(2006) Collection
Fabrizio et al. Whole genome, ORF Deletion B4800 42 Protein homeostasis, metabolism, lipid
(2010) Collection production, stress tolerance, cell growth
Matecic et al. Whole genome, ORF Deletion B4800 12 Protein homeostasis, metabolism
(2010) Collection

(Continued)

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


6 1. Longevity as a complex genetic trait

TABLE 1.1 (Continued)


Study Primary gene selection No. No. genes Epistasis Functional groups identified
genes identified tested
tested

Burtner et al. Random selection, ORF Deletion 227 32 Mitochondrial function, stress resistance, protein
(2011) Collection homeostasis
Burtner et al. Orthologs of worm pro-aging genes 235 18 Mitochondrial function, cell division, metabolism,
(2011) protein homeostasis, exocytosis
Burtner et al. Increased replicative life span 47 10 Cell division, transcription, DNA replication,
(2011) metabolism
Burtner et al. Increased media pH 76 20 Endocytosis, protein homeostasis, signal
(2011) transduction, stress resistance

Garay et al. Whole genome, ORF Deletion 3878 262 Protein homeostasis, DNA repair, gene
(2014) Collection expression, Golgi function, metabolism

Campos et al. Whole genome, ORF Deletion 3718 254 Functional analysis not performed
(2018) Collection
Campos et al. Whole genome, ORF Deletion 3718 228 Functional analysis not performed
(2018) Collection, nitrogen-restricted
media
Fruit fly lifespan
Landis et al. Random dox-inducible P element 10,000 6 Vacuolar function, membrane transport, cell
(2003) insertion structure
Funakoshi Reduced wing and eye size; 716 2 Signal transduction, cell growth, protein
et al. (2011) random P{GS} element insertion homeostasis
Paik et al. Random EP element insertion 27,157 15d DNA replication, transcription, chromatin
(2012) binding or modification, protein homeostasis,
signal transduction, metabolism, immunity
Chen et al. MicroRNA Deletion Collection 130 12e MicroRNA
(2014)
Ostojic et al. Gustatory receptors 6 4 Food perception
(2014)
Shaposhnikov DNA repair genes 9 8f DNA damage detection and repair
et al. (2015)
a
The authors only pursue four genes, but do not report the total number found to significantly affect lifespan.
b
The authors only report the number of significant hits on chromosome I.
c
The authors pursue the 90 genes with the largest change in chronological lifespan, but do not report how many are statistically significant.
d
8736 of 27,157 lines were putatively classified as long-lived; the authors selected 45 and 15 remained long-lived after validation.
e
The authors tested 95 Drosophila strains with combined deletion of 130 miRNAs.
f
Genes that increased lifespan in at least one sex under at least one expression mode (constitutive vs adult only, whole body vs neuronal).

proteins, cell signaling, protein homeostasis, RNA pro- methodology. Another possibility is that subtle differ-
cessing, and chromatin binding. ences in experimental design may result in a different
Notably, while a large number of genes has been range of factors becoming prominent. These differences
identified through longevity screening in C. elegans, there include culture temperature, strain background, age at
is little overlap between screens (Yanos et al., 2012). RNAi induction, and the presence or absence of floxuri-
There are several possibilities that may account for this dine (FUdR) to prevent reproduction (Table 1.2). Recent
lack of overlap. RNAi is inherently noisy, which may evidence suggests that the strain of Escherichia coli used
result in a different degree of knockdown between in RNAi experiments (HT115) may have distinct interac-
experiments for a given clone. Many of these screens tions with the C. elegans genotype in the context of life-
were designed to assess maximum lifespan, scoring only span from the E. coli strain used in most non-RNAi
the number of worms alive after all control worms had experiments (OP50) (Xiao et al., 2015; Yen & Curran,
died. Between these two factors, the low overlap may 2016), further complicating comparisons between
reflect a high false-negative rate inherent in the RNAi screens and other categories of intervention. The

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Defining the aging gene-space 7
TABLE 1.2 Experimental conditions used in different C. elegans longevity screens
Study Strain background Worm stock temperature Experiment temperature RNAi start FUdR?
 
Dillin et al. (2002) Wild type 20 C 25 C Egg No
 
Lee et al. (2003) Wild type 20 C 25 C L1 Yes
 
Munoz et al. (2003) fer-15(b26ts) 20 C 25.5 C n/a No
 
de Castro et al. (2004) mut-7(pk242) 20 C 20 C n/a No

Hamilton et al. (2005) Wild type ? 20 C L1 Yes
  
Hansen et al. (2005) fer-15(b26); fem-1(hc17) 25 C 20 C or 25 C Egg No
 
Samuelson et al. (2007) lin-15b(n744); eri-1(mg366) ? 15 C to L4, 25 C thereafter L1 Yes
 
Chen et al. (2007) Wild type 20 C 20 C L4 Yes
 
Curran and Ruvkun (2007) eri-1(mg366) 20 C 20 C L4 Yes

Kim and Sun (2007) rrf-3(pk1426) ? 20 C L1 No
  
Sutphin et al. (2017) Wild type 20 C 15 C or 25 C egg Yes

Caenorhabditis Interventions Testing Program (CITP) was Knockout screens in budding yeast
established in 2013 as a platform to identify novel In the budding yeast S. cerevisiae, an analog to the C.
lifespan-extending pharmacological compounds through elegans RNAi feeding libraries exists in the form of a
rigorous testing across three independent laboratories, genome-wide single-gene deletion strain collection. This
modeled after the successful Interventions Testing collection contains B4800 strains, each containing a
Program in mice (Nadon, Strong, Miller, & Harrison, complete open reading frame (ORF) deletion for a sin-
2017). Initial testing revealed significant problems in gle nonessential gene in a common genetic background
reproducibility across test sites, resulting in a multiyear (Winzeler et al., 1999). Versions of this collection are
project to standardize reagents, materials, and methods available in both haploid mating types and in the
(Lithgow, Driscoll, & Phillips, 2017; Lucanic et al., 2017) homozygous diploid life stage. When conceptualizing
culminating in the recent release of a set of standardized longevity in a single-celled organism like S. cerevisiae,
protocols for measuring worm lifespan (Lucanic et al., the first question to consider is the definition of “life-
2017). Wide adoption of these protocols may go some span.” Two aging paradigms are commonly studied in
way toward limiting reproducibility and variation across the budding yeast (Steinkraus, Kaeberlein, & Kennedy,
labs. Petrascheck and Miller (2017) recently employed 2008). Replicative lifespan refers to the number of times
statistical models of wild-type C. elegans lifespan data to a cell can divide prior to undergoing senescence
simulate aspects of methodological variation (e.g., scor- (Kaeberlein, 2006; Mortimer & Johnston, 1959). In con-
ing frequency, sample size) and population structure trast, chronological lifespan refers to the length of time
(e.g., hazard distribution) in longevity studies and deter- a cell can remain in a quiescent state while retaining the
mine their influence on reproducibility under “ideal” ability to re-enter the cell cycle (Fabrizio & Longo, 2003;
conditions (i.e., in the absence of methodological error Kaeberlein, 2006; Fabrizio, Pozza, Pletcher, Gendron, &
and environmental variation). They conclude that statisti- Longo, 2001).
cal detection of lifespan differences is surprisingly resil- Until recently, high-throughput techniques had only
ient to scoring frequency and shape of the lifespan been developed for measuring chronological lifespan in
distribution within a population. Perhaps unsurprisingly, yeast. Chronological lifespan is typically measured by
they determined that sampling size was the primary growing yeast cells in liquid culture until they enter sta-
driver of poor reproducibility and that most studies are tionary phase, maintaining the cells in the expired media,
underpowered to detect small (,10%) changes in life- and periodically sampling the aging culture to assess via-
span (Petrascheck & Miller, 2017). Regardless of the bility (Kaeberlein, 2006). Viability has traditionally been
cause, the small degree of overlap and the fact that these measured by plating a defined culture volume onto rich
screens only identified pro-aging genes—genes for which solid media and counting the number of colonies to cal-
reduced expression increases lifespan—suggests that the culate colony-forming units (CFUs). Powers, Kaeberlein,
range of genetic factors involved in C. elegans aging has Caldwell, Kennedy, and Fields (2006) dramatically
yet to be exhaustively bounded. increased throughput by replacing the labor-intensive

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


8 1. Longevity as a complex genetic trait

(though quantitative) process of counting CFUs with the chronological lifespan, the three studies used different
more qualitative approach of diluting a sample from the subsets of the yeast deletion collection (diploid BY4743
aging culture back into rich liquid media and measuring vs haploid BY4741) and different media types (rich vs
optical density at 600 nm (OD600) after a fixed outgrowth defined media). Smith et al. (2016) recently revisited
time. They used this approach to screen the homozygous these screens, examining the methodological differ-
diploid deletion collection, identifying 90 chronologically ences in detail, concluding that differences in media
long-lived mutants (Powers et al., 2006; Table 1.1). This components were likely a major contributor to lack of
technique was later improved to quantitatively assess consensus in identified genes, but that other methodo-
outgrowth using a combined instrument that provides logical factors were also involved (aeration, method of
continuous culture agitation, temperature control, and measuring chronological lifespan, competitive vs non-
OD600 measurement (Burtner, Murakami, & Kaeberlein, competitive environments).
2009; Murakami & Kaeberlein, 2009; Olsen, Murakami, & In contrast to chronological lifespan, the typical
Kaeberlein, 2010) and has been used to screen selected method for measuring replicative lifespan in yeast
sets of mutants from the yeast ORF deletion collection for involves the manual and labor-intensive removal of
increased chronological lifespan (Burtner, Murakami, daughter cells from a dividing mother. To bypass this
Olsen, Kennedy, & Kaeberlein, 2011). Campos, Avelar- problem, a moderate-throughput iterative strategy was
Rivas, Garay, Juárez-Reyes, and DeLuna (2018) used a devised to identify long-lived mutants in the yeast
similar method adapted for a robotic platform to measure deletion collection by determining replicative lifespan
the chronological lifespan of 3718 strains from the yeast initially for only five cells per strain and using statisti-
deletion collection in media with either glutamine (a pre- cal methods to select strains for further testing
ferred nitrogen source; considered nonrestricted) or (Kaeberlein et al., 2005). A preliminary report identi-
γ-aminobutyric acid (GABA; a model of nitrogen restric- fied 13 genes for which deletion extends replicative
tion) as the sole nitrogen source. They identified 573 lifespan out of the first 564 strains initially tested in
(15%) chronologically short-lived and 254 (7%) chronolog- the ORF deletion collection (Kaeberlein et al., 2005). Of
ically long-lived single-gene deletion strains in the nonre- the 13 genes, five map to the mTOR signaling pathway
stricted media, and 510 (6%) chronologically short-lived (ROM2, RPL6B, RPL31A, TOR1, and URE2). This
and 228 (14%) chronologically long-lived single-gene screen was recently completed, identifying 238 long-
deletion strains in the nitrogen-restricted media. In a lived deletion strains among 4698 tested, including 189
modified approach, Garay et al. (2014) monitored the that were not previously reported to influence lifespan
chronological lifespan of 3878 fluorescently labeled dele- (McCormick et al., 2015). Pathway enrichment among
tion collection strains, each pooled into a common culture the final pro-aging gene set confirmed the importance
during stationary phase arrest with the wild-type strain of mTOR signaling and cytosolic translation, and iden-
expressing a distinct fluorescent label. Viability was tified a number of other pathways and cellular pro-
assessed for each mutant strain relative to the pooled cesses that play an important role in replicative aging:
wild-type based on the relative fluorescence during out- mitochondrial translation, the tricarboxylic acid (TCA)
growth. This method identified 516 (13%) chronologically cycle, mannosyltransferases, and the SAGA complex.
short-lived and 262 (7%) chronologically long-lived Two additional replicative lifespan screens have been
strains. Fabrizio et al. (2010) and Matecic et al. (2010) both reported examining gene sets selected for either orthol-
employed an alternative competitive strategy, chronologi- ogy to known worm aging genes (Smith et al., 2008) or
cally aging a pooled culture containing cells from each of ribosomal components (Steffen et al., 2008). Combined,
the single-gene deletion strains in the ORF deletion collec- longevity screens in yeast have identified approxi-
tion and using microarrays to genotype the longest sur- mately 250 pro-aging genes related to a range of cellu-
viving cells. A recent improvement to further increase lar processes including protein homeostasis,
throughput shifts the chronologically aging yeast cultures metabolism, stress resistance, and mitochondrial func-
from culture tubes to 96-deep-well plates with outgrowth tion (Table 1.1).
analysis performed in 384-well plates (Jung, Christian,
Kay, Skupin, & Linster, 2015). Overexpression and knockout screens in fruit flies
Similar to the RNAi screens for increased lifespan in Tools for genome-scale targeted genetic modifica-
C. elegans, the screens by Powers et al. (2006), Matecic tion have yet to be used in the context of aging in D.
et al. (2010), and Fabrizio et al. (2010) found a remark- melanogaster. Drosophila does provide a unique tool
able lack of overlap in genes identified to affect yeast among invertebrate aging models in the form of trans-
chronological lifespan (Smith, Maharrey, Carey, White, posable enhancer and promoter elements that can be
& Hartman, 2016). These yeast screens varied method- randomly inserted into the genome allowing for unbi-
ologically to a greater extent than the C. elegans ased identification of genes that increase lifespan when
screens. Beyond using distinct methods for measuring overexpressed. In an early study using this method,

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Defining the aging gene-space 9
Landis, Bhole, and Tower (2003) screened 10,000 lines the determination of lifespan. Hundreds of genes have
and identified six genes for which overexpression been identified across a range of central biological pro-
increased longevity, including factors involved in vac- cesses, the most prominent being mitochondrial metab-
uolar function, membrane transport, and cell structure olism, protein homeostasis, and stress resistance
(Table 1.1). Paik et al. (2012) initiated a longevity (Table 1.1). There is still work to be done in this area,
screen examining 27,157 lines and reported the first 15 particularly with respect to understanding how the
long-lived transgenic strains overexpressing genes range of factors important for lifespan is affected by
involved in transcription, translation, cell signaling, different environmental conditions, such as changes in
metabolism, and immunity (Table 1.1). Detailed mech- temperature or in response to dietary restriction.
anistic studies have now been published for two of
these genes: (1) malic enzyme (Men), overexpression of
which during larval development is sufficient to Leveraging genetic diversity to identify
extend lifespan, drives a range of metabolic changes, aging loci
and improves resistance to oxidative stress (Kim et al.,
The previous sections describe reverse genetic
2015); and (2) NAD-dependent methylenetetrahydrofo-
late dehydrogenase-methenyltetrahydrofolate cyclohy- approaches to identifying aging genes, in which large
drolase (Nmdmc), which increases lifespan and numbers of genes are knocked out or overexpressed
improves mitochondrial homeostasis when overex- individually and the effect on lifespan measured.
pressed either whole body or in fat (Yu, Jang, Paik, Because of the scale, this approach has only been car-
Lee, & Park, 2015). ried out in short-lived invertebrate models that are sim-
ple and inexpensive to maintain in the laboratory.
A third study used growth impairment in the form
Current genome-scale knockout efforts like the
of reduced wing and eye size as a surrogate marker
International Mouse Knockout Consortium (IMKC) and
for longevity in a screen of 716 transgenic Drosophila
lines (Funakoshi et al., 2011). Two genes were identi- International Mouse Phenotyping Consortium (IMPC;
fied with previous links to insulin/IGF-1-like and see discussion of emerging tools later in this chapter)
mTOR signaling (Table 1.1). Finally, Shaposhnikov, may lend themselves to a similar strategy in mice on a
Proshkina, Shilova, Zhavoronkov, and Moskalev (2015) smaller scale, though the cost of maintaining statistically
overexpressed nine genes involved in DNA damage meaningful numbers of mice throughout life will still
likely prevent full-genome mouse lifespan screens.
detection and repair in both sexes under different tem-
An alternative approach is to use forward genetics
poral (constitutive vs adult-only) and tissue (whole-
to leverage the natural phenotypic variation in geneti-
body vs neuronal) expression patterns and in different
environmental contexts (well fed, starvation, heat cally diverse populations to map candidate aging loci.
stress, oxidative stress). The impact of each gene on This approach has the advantage of directly identify-
lifespan was highly dependent on expression pattern ing genes even in long-lived mammalian systems.
and environmental context. Of the nine genes exam- Genes identified in mammalian systems are more
ined, overexpression of eight increased lifespan of likely to be relevant to human aging than those identi-
fied in evolutionarily distant invertebrate screens. This
well-fed, unstressed flies under at least one of the
approach also provides a complement to the screens
tested patterns of expression.
carried out in invertebrate systems. Invertebrate
While genome-scale tools analogous to the C. elegans
RNAi feeding library or the yeast deletion collection are screens tend to increase or decrease gene expression to
not currently available in Drosophila, more targeted col- levels outside of what is typically experienced from
lections of genes have been examined in the context of allelic variants in natural populations. Natural variants
aging. Chen et al. (2014) screened a collection of 95 can result in large changes in gene activity, including
Drosophila strains with deletions in 130 different complete inactivation of a gene, but more typically
cause subtler changes in gene action or specificity.
microRNAs (miRNAs) (representing 99% of known
Gene mapping will therefore both identify longevity
Drosophila miRNAs) for lifespan and a variety of other
effects from less dramatic gene interventions, and
phenotype. They report increased lifespan in 12 strains
and decreased lifespan in 23 lines, demonstrating that point to genes that make the largest contributions to
miRNAs are a potentially rich source for genetic targets the variation in aging within the population examined.
to modify longevity. Ostojic et al. (2014) found that
knocking out four of six examined gustatory receptors Mapping longevity genes in human populations
resulted in increased lifespan in one or both sexes. The first gene-mapping studies to examine longev-
Genetic screening for lifespan variants using inver- ity employed linkage analysis in human families.
tebrate models has been invaluable for defining the Three studies of this type mapped loci associated with
range of factors and biological processes involved in extreme longevity in 137 sibling pairs with one

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


10 1. Longevity as a complex genetic trait

member being at least 98 years old and other members (rather than subject lifespan) to increase sample size
being at least 90 (males) or 95 (females) years old (Joshi et al., 2016, 2017; McDaid et al., 2017; Pilling
(Puca et al., 2001), 95 pairs of male fraternal twins with et al., 2016; Timmers et al., 2019) and/or employ meth-
healthy aging (Reed, Dick, Uniacke, Foroud, & ods to amplify variants previously associated with
Nichols, 2004), and 279 families with multiple long- phenotypes or diseases that increase age-associated
lived siblings (Boyden & Kunkel, 2010). All three stud- risk of mortality (Fortney et al., 2015; McDaid et al.,
ies identified one or more loci associated with varia- 2017; Timmers et al., 2019). These studies have identi-
tion in longevity, most notably a common locus on fied numerous novel longevity-associated regions and
chromosome 4 (Table 1.3). confirmed several previously identified loci (Table 1.3).
With the availability of relatively inexpensive high- Notably, while APOE remains the major longevity-
density single nucleotide polymorphism (SNP) arrays associated gene identified through GWAS, there are
and exome sequencing, most mapping efforts are now now several loci that have reached genome-wide sig-
concentrating on genome-wide association studies nificance in at least two GWAS studies: the major
(GWAS) with increasing population sizes. An example histocompatibility complex (HLA) and lipoprotein(a)
of this is type of study is work by Newman et al. (LPA) loci on chromosome 6, the cyclin-dependent
(2010), in which more than 2 million polymorphisms kinase inhibitor 2B (CDKN2B) and α-1-3-(N)-acetylga-
were examined in a meta-analysis of four prospective lactosaminyltransferase (ABO) loci on chromosome 9,
cohort studies combining 1836 individuals that sur- and the hydroxylysine kinase (HYKK) locus on chro-
vived beyond 90 and a control group of 1955 indivi- mosome 15 (Table 1.3). While these loci have yet to be
duals. Despite the large sample size, no loci reached validated, convergence suggests that the recent
genome-wide significance for association with longev- improvements in GWAS power from increased sample
ity, though MINPP1, an inositol phosphatase involved size and new methodologies have begun to break the
in cell proliferation, approached significance. This long-standing barrier to directly uncovering novel
example illustrates a common challenge in longevity aging variants in human populations.
mapping studies. Many loci throughout the genome
have been found to be associated with variation in life- Mapping longevity genes in mouse populations
span; however, few loci reach genome-wide statistical Like humans, mice have been employed as a model
significance when corrected for multiple testing and in studies to map genetic loci associated with variation
little overlap is found between studies (Table 1.3). The in lifespan. While most mouse populations are main-
notable exception is APOE, which has been identified tained as inbred strains, a survey of 32 inbred strains
by multiple independent genome-wide longevity map- found substantial variation in median lifespan among
ping studies (Beekman et al., 2013; Broer et al., 2015; different inbred strains ranging from 251 days for
Deelen et al., 2014, 2011; Fortney et al., 2015; Joshi AKR/J to 964 days for WSB/EiJ (Yuan et al., 2009).
et al., 2016, 2017; McDaid et al., 2017; Nebel et al., This inherent variation can be exploited by crossing
2011; Sebastiani et al., 2012, 2013, 2017; Timmers et al., strains with different lifespans and perform linkage
2019). APOE is located on chromosome 19 and encodes analysis. To date, 11 such studies have been completed
an apolipoprotein that is a major component of very using crosses between seven classical inbred strains:
low-density lipoproteins (VLDLs), which are responsi- BALB/c (BALB), C3H/HeJ (C3H), C57BL/6J (B or B6),
ble for removing excess blood cholesterol. Allelic var- DBA/2J (D or D2), KK/HIJ (KK), LP/J (LP), NZW/
iants in APOE are associated with Alzheimer’s disease, LacJ (NZW), PL/J (PL), and ST/bJ (ST); three wild-
atherosclerosis, and other age-associated pathologies. derived inbred strains: CAST/Ei (CAST), MOLD/Rk
While not yet identified in genome-wide mapping (MOLD), and POHN/DehJ (POHN); and two strains
studies, numerous targeted studies have found signifi- developed by crossing classical inbred strains and
cant association between FOXO3 and human longevity selecting for sleep response to ethanol: inbred long
(Anselmi et al., 2009; Flachsbart et al., 2009; Li et al., sleep (ILS/IbgTejJ or ILS) and inbred short sleep (ISS/
2009; Pawlikowska et al., 2009; Soerensen et al., 2010; IbgTejJ or ISS). CAST, MOLD, and POHN are particu-
Willcox et al., 2008; Zeng et al., 2010). FOXO3 encodes larly important, as they were inbred from wild-caught
a forkhead family transcription factor that has been mice without domestication and provide a large frac-
linked to oxidative stress resistance and tumorigenesis. tion of the genetic diversity found among strains used
The FOXO3 homologs in C. elegans (daf-16) and D. mel- to generate crosses for mapping studies. CAST and
anogaster (dFOXO) mediate many of the beneficial MOLD originate from the subspecies Mus m. castaneus
effects of reduced insulin/IGF-1-like signaling with and Mus musculus molossinus, respectively, which
respect to longevity and age-associated pathology. diverged from the subspecies of most classical inbred
Two recent approaches to improve the power of strains, Mus m. musculus, approximately half a million
GWAS use parental lifespan of genotyped subjects years ago.

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Defining the aging gene-space 11
TABLE 1.3 Significant and suggestive loci identified in genome-wide human mapping studies.
Where available, marker names were used to standardize all genomic locations to human genome assembly
GRCh37.p13. Genes listed are located within 10 kb of a marker location. Loci within 10 Mb of each other are
grouped. Bold 5 significant genome-wide association; not bold 5 suggestive genome-wide association.
Chr Region or marker Gene(s) References
location (Mb)

1 3.6 TP73 Samuelson et al. (2007)


41.2, 44.2 KCNQ4, NFYC, ST3GAL3 Kim and Sun (2007)
55.5, 56.9, 63.4 PPAP2B Kim and Sun (2007), Yanos et al. (2012)
109.8, 114.2 CELSR2, PSRC1, MAGI3 Curran and Ruvkun (2007), Yanos et al. (2012)

195.9-197.3 (196.4) KCNT2 Fraser et al. (2000)


237.1, 240.9 RPL35P1 Murakami and Kaeberlein (2009), Sutphin and
Kaeberlein (2009)
2 0.6, 8.5, 10.1 AC010969.1, RP11.254F7.1 Bennett et al. (2017), Chen et al. (2007), Curran and
Ruvkun (2007)

26.9, 28.7, 35.4 AC013442.1, KCNK3 Curran and Ruvkun (2007), de Castro et al. (2004),
RNAi Resources | Source BioScience (n.d.)

50.5 NRXN1 Bennett et al. (2017)


70.8 TGFA Bennett et al. (2017)
139.4 NXPH2 Braeckman and Vanfleteren (2007)
162.9, 166.4 CSRNP3, DPP4 Sutphin and Kaeberlein (2009), Yanos et al. (2012)
237.1, 238.3 AC079135.1, COL6A3, GBX2 Curran and Ruvkun (2007), Kim and Sun (2007)
3 0.2, 2.0 Linford et al. (2013), Rual et al. (2004), Samuelson
et al. (2007)
28.633.6 (30.2), 29.6, 38.0 CTDSPL, RBMS3 C. elegans and Dharmacon (2019), Goffeau et al.
(1996), Rual et al. (2004)
48.5, 49.9, 50.1 ATRIP, CAMKV, CCDC51, RBM6, TMA7, TRAIP Bennett et al. (2017), Linford et al. (2013), Yanos et al.
(2012)
71.8 EIF4E3 Dillin et al. (2002), Lee et al. (2003)
85.5 CADM2 Murakami and Kaeberlein (2009)
96.2, 101.0 AC108697.1, IMPG2 Smith et al. (2008), Sutphin et al. (2017)

115.1 Samuelson et al. (2007)


162.7, 168.7 Kim and Sun (2007)
192.5192.6 MB21D2 C. elegans and Dharmacon (2019)
4 1.4, 2.3, 3.1, 3.2 HTT, MSANTD1, RP11-478C1.7, UVSSA, ZFYVE28 Bennett et al. (2017), Curran and Ruvkun (2007),
Sutphin et al. (2017), Yanos et al. (2012)
41.6, 42.3 LIMCH1 Murakami and Kaeberlein (2009), Sutphin et al.
(2017)
60.2 Kamath et al. (2003)
76.9 SDAD1 Bennett et al. (2017)
89.8, 94.5 GRID2, FAM13A Sutphin et al. (2017), Sutphin and Kaeberlein (2009)
108.4, 110.6, 111.2, 120.7 CCDC109B, RP11-236P13.1 Chen et al. (2007), Goffeau et al. (1996), Hansen et al.
(2005), Samuelson et al. (2007), Sutphin and
Kaeberlein (2009)

(Continued)

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


12 1. Longevity as a complex genetic trait

TABLE 1.3 (Continued)


Chr Region or marker Gene(s) References
location (Mb)

137.7 Bennett et al. (2017)

160.9162.5 C. elegans and Dharmacon (2019)


5 55.9 AC022431.2 Yanos et al. (2012)
74.878.9 (76.2) S100Z Rual et al. (2004)
96.3 LOC101929747 Linford et al. (2013)
110.8 CAMK4 Smith et al., 2008
140.2, 149.4, 149.6, 157.8 CSF1R, SLC6A7, PCDHA1, PCDHA2, PCDHA3, Dillin et al. (2002), Kamath et al. (2003), Lee et al.
PCDHA4, PCDHA5, PCDHA6, PCDHA7 (2003), Sutphin and Kaeberlein (2009)
6 8.2 MYLK4 Linford et al. (2013)
20.0, 25.733.0 (32.4), 26.2, AIF1, BTNL2, C2, HCP5, HIST1H2BD, Chen et al. (2007), Curran and Ruvkun (2007), de
29.629.7 (29.7), 31.9, 32.6 HIST1H2BE, HLA-DQA1, HLA-DQA2, HLA- Castro et al. (2004), Dillin et al. (2002), Fraser et al.
DQB1, HLA-DRB5, HLADRB9, HLA-F, HLA-F- (2000), Lee et al. (2003), Murakami and Kaeberlein
AS1, LY6G6F, MICA, MOG, MSH5, SKIV2L, (2009), RNAi Resources | Source BioScience (n.d.)
ZFP57

48.262.2 (50.2), 50.9 RCBTB1 Linford et al. (2013), Yanos et al. (2012)
102.8, 102.9, 106.8, 109.0 AIM1, FOXO3 Bennett et al. (2017), Murakami and Kaeberlein
(2009)

131.4 RNAi Resources | Source BioScience (n.d.)


160.3161.7 (161.0), 160.5, PRR18, IGF2R, LPA Bennett et al. (2017), Curran and Ruvkun (2007),
161.0, 164.3, 164.4169.6 Fraser et al. (2000), Hamilton et al., (2005), Kamath
(164.4, 166.2), 166.3, 166.7 et al. (2003), Linford et al., (2013), RNAi Resources |
Source BioScience (n.d.), Timmons and Fire (1998),
Yanos et al. (2012)
7 1.0, 1.9, 6.2 C7orf50, MAD1L1, USP42 Bennett et al. (2017), Chen et al. (2007)
22.8 AC073072.5, IL6 Sutphin et al. (2017)
36.8 AC007349.4 Sutphin and Kaeberlein (2009)
48.3, 49.575.5 (64.7), 75.1, AP5Z1, MIR4656, POM121C, RADIL Braeckman and Vanfleteren (2007), Curran and
75.2, 82.3 Ruvkun (2007), Hamilton et al. (2005), Linford et al.
(2013)
92.4, 103.9, 109.8 CDK6 Dillin et al. (2002), Kim and Sun (2007), Lee et al.
(2003)
122.8, 129.7, 134.3 AKR1B15, SLC13A1, ZC3HC1 Bennett et al. (2017), Curran and Ruvkun (2007),
Linford et al. (2013)
152.6 Smith et al. (2008)
8 6.7 DEFB1 Dillin et al. (2002), Lee et al. (2003)
19.9 Yanos et al. (2012)
31.0 WRN Dillin et al. (2002), Lee et al. (2003)
41.667.0, 53.9, 72.3 Dillin et al. (2002), C. elegans Sequencing Consortium
(1998), Lee et al. (2003), Sutphin and Kaeberlein
(2009)
134.5 ST3GAL1 Linford et al. (2013)
9 5.35.4 RLN1, RLN2 C. elegans and Dharmacon (2019)

(Continued)

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Defining the aging gene-space 13
TABLE 1.3 (Continued)
Chr Region or marker Gene(s) References
location (Mb)

21.922.1 (22.1), 22.1, 27.2, CDKN2B, CDKN2B-AS1, LOC729983, Curran and Ruvkun (2007), Dillin et al. (2002), Fraser
28.1 LOC100130239, TEK et al. (2000), Yanos et al. (2012)
73.8 TRPM3 Bennett et al. (2017)
97.5, 97.6 C9orf3 Dillin et al. (2002), Lee et al. (2003)

113.1 SVEP1 Bennett et al. (2017)


128.3, 136.0136.3 (136.1), ABO, MAPKAP1, COL5A1 Bennett et al. (2017), Curran and Ruvkun (2007),
136.1, 137.7, 138.3 Fraser et al. (2000), Goffeau et al. (1996), Hamilton
et al. (2005)
10 3.35.6 (4.3), 4.1, 5.1 AKR1C3, LOC101927946, R11-49907.4, U8.23 Bennett et al. (2017), Sutphin et al. (2017), Timmons
and Fire (1998)
23.4 MSRB2 Smith et al. (2008)
51.952.3 (52.1) SGMS1 Rual et al. (2004)

89.3, 96.6 CYP2C58P Muñoz and Riddle (2003), Sutphin and Kaeberlein
(2009)

108.6 SORCS1 Dillin et al. (2002), Lee et al. (2003)


11 0.31.1, 3.0 CARS Bennett et al. (2017), C. elegans and Dharmacon
(2019)

15.9 Murakami and Kaeberlein (2009)


50.051.4 C. elegans and Dharmacon (2019)
90.9 Braeckman and Vanfleteren (2007)
122.9, 124.0, 126.2 DCPS, LOC341056, VWA5A Dillin et al. (2002), Kim and Sun (2007), Lee et al.
(2003)
12 14.1 GRIN2B Kim and Sun (2007)
48.5, 51.7, 61.661.7 (61.6), BIN2, FEN1, FADS1, PFKM Fraser et al. (2000), Kim and Sun (2007), Murakami
68.9 and Kaeberlein (2009), Sutphin and Kaeberlein (2009)
83.4 TMTC2 Chen et al. (2007)
110.1113.0 (111.9), 112.1, ATXN2, GPR133, RP11-575F12.1, SH2B3 Bennett et al. (2017), Curran and Ruvkun (2007),
121.4, 127.4, 131.5, 132.1 Fraser et al. (2000), Goffeau et al., (1996)
13 19.4 ANKRD20A9P, RNU6-55P, RP11-38M15.11 Sutphin et al. (2017)
48.4, 53.8, 61.7 RN7SL618P Bennett et al. (2017), Kim and Sun (2007), Sutphin
et al. (2017)
71.9, 73.1 Bennett et al. (2017), Smith et al. (2008)
90.6, 93.9, 95.7, 99.1 ABCC4, GPC6, STK24 Bennett et al. (2017), Dillin et al. (2002), Lee et al.
(2003)
14 20.423.7, 29.0 Bennett et al. (2017), C. elegans and Dharmacon
(2019), C. elegans Sequencing Consortium (1998)
40.7, 43.554.5 (49.5), 62.4 CTD-2277K2.1, SYT16 C. elegans and Dharmacon (2019), Linford et al.
(2013), Sutphin and Kaeberlein (2009)
90.8 NRDE2 Bennett et al. (2017)
104.2106.1 C. elegans and Dharmacon (2019)
15 27.0, 27.935.0, 33.7 GABRB3, RYR3 Bennett et al. (2017), Dillin et al. (2002), C. elegans
Sequencing Consortium (1998), Lee et al. (2003)

(Continued)

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


14 1. Longevity as a complex genetic trait

TABLE 1.3 (Continued)


Chr Region or marker Gene(s) References
location (Mb)

53.8 WDR72 Bennett et al. (2017), Dillin et al. (2002)

78.878.9 (78.8), 78.8, 78.9 AC027228.1, CHRNA3, CHRNA5, CHRNB4, Curran and Ruvkun (2007), Hamilton et al. (2005),
HYKK, IREB2, PSMA4, RP11-335K5.2 Kamath et al. (2003), RNAi Resources | Source
BioScience (n.d.), Yanos et al. (2012)
91.4, 94.8 FES, FURIN, MCTP2 Curran and Ruvkun (2007), Hamilton et al. (2005),
Linford et al. (2013)
16 2.1 NTHL1, TSC2 Bennett et al. (2017)
12.5, 19.026.6 (24.0), 19.9, ATXN2L, IQCK, PRKCB, RP11-1348G14.4, SNX29 Linford et al. (2013), Timmons and Fire (1998), Yanos
28.8 et al. (2012)
48.5, 50.7, 53.8, 57.9 AC023818.1, FTO, KIFC3, RN7SL54P, SIAH1, Chen et al. (2007), Samuelson et al. (2007), Smith
SNX20 et al., (2008), Yanos et al. (2012)
72.1, 78.5 TXNL4B, WWOX Curran and Ruvkun (2007), Dillin et al. (2002), Lee
et al. (2003)
17 8.9, 9.811.5 (10.1) GAS7, NTN1 Sutphin and Kaeberlein (2009), Timmons and Fire
(1998)
31.4, 36.755.5, 47.9 ASIC2, FLJ45513, RP11-40A13.1, TAC4 C. elegans Sequencing Consortium (1998), Rual et al.
(2004), Sutphin et al. (2017)
60.9, 61.4, 64.5, 70.0, 79.3 PRKCA, SLC38A10, TANC2 Dillin et al. (2002), Smith et al. (2008), Sutphin and
Kaeberlein (2009)

18 70.274.1 (72.2), 71.0, 74.9 CNDP1, ZNF516 Bennett et al. (2017), Kim and Sun (2007), Rual et al.
(2004)

19 6.717.5, 11.2 LDLR Curran and Ruvkun (2007), C. elegans Sequencing


Consortium (1998)
35.346.6 (45.4), 38.3, AC016582.2, CTB-129P6.4, APOC1, APOE, BCAM, Bennett et al. (2017), Chen et al. (2007), Curran and
45.145.5 (45.4), 45.4, BCL3, CEACAM16, PVRL2, TOMM40 Ruvkun (2007), de Castro et al. (2004), Dillin et al.,
49.153.5 (2002), C. elegans Sequencing Consortium (1998),
Fraser et al., (2000), Kamath et al., (2003), Lee et al.,
(2003), RNAi Resources | Source BioScience (n.d.),
Steffen et al. (2009), Sutphin et al. (2017), Sutphin
and Kaeberlein (2009)
20 0.64.1 (0.9) ANGPT4 Timmons and Fire (1998)
52.2, 59.9, 60.2 CDH4, Y RNA Bennett et al. (2017), Sutphin and Kaeberlein (2009)
21 14.8, 17.1, 22.9 AL050302.1, ANKRD30BP1, NCAM2, UPS24 Bennett et al. (2017), Kamath et al. (2003), Sutphin
et al. (2017)
47.6 LSS Linford et al. (2013)
22 37.6, 44.5, 47.5, 51.1 PARVB, SHANK3, SSTR3, TBC1D22A Bennett et al. (2017), Hamilton et al. (2005)

X 141.1, 142.2 Linford et al. (2013), Samuelson et al. (2007)

One approach to gene mapping in mice is to generate 2010) or the ILSXISS RI strain panel (Rikke, Liao,
recombinant inbred (RI) strain panels by crossing two or McQueen, Nelson, & Johnson, 2010). A second
more strains and inbreeding subsequent generations to approach is to genotype and measure lifespan of non-
produce novel but related sets of inbred strains. Four inbred offspring generated by crossing two or more
studies have measured lifespan and mapped associ- inbred strains. Lifespan has twice been measured in
ated loci using either the BXD RI strain panel (de the UM-HET3 four-way cross, (BALBxB6)x(C3HxD2)
Haan, Gelman, Watson, Yunis, & Van Zant, 1998; (Jackson, Galecki, Burke, & Miller, 2002; Miller,
Gelman, Watson, Bronson, & Yunis, 1988; Lang et al., Chrisp, Jackson, & Burke, 1998), and once each in two

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Defining the aging gene-space 15
four-way crosses that include a wild-derived strain, Age-associated gene expression studies
(STxB6)x(CASTxD2) and (LPxMOLD)x(NZWxBALB) Instead of measuring the response of age-related pro-
(Klebanov et al., 2001). An early study by Yunis et al. cesses to changes in gene activity, the impact of age on
(1984) and a more recent study by Yuan, Flurkey, gene expression can be used to identify potential lon-
Meng, Astle, and Harrison (2013) identified gevity factors. Comparing expression studies has the
longevity-associated loci in the (B6xD2)xB6 and potential to provide insight into general mechanisms of
(POHNxB6)xPOHN backcross populations, respec- aging that cross tissue and species boundaries.
tively. Another study mapped loci associated with Microarray studies comparing young and old in flies
lifespan in a backcross between short-lived SAMP1 (Landis et al., 2004; Pletcher et al., 2002; Zou, Meadows,
mice and long-lived B10.BRH2k/SgSnSlc (B10.BR), a Sharp, Jan, & Jan, 2000), mice (Weindruch, Kayo, Lee, &
congenic strain related to B57BL/10 that shares the Prolla, 2001), and monkeys (Kayo, Allison, Weindruch,
histocompatibility 2 (H2) locus with SAMP1, remov- & Prolla, 2001) all found an increase in oxidative stress
ing the influence of H2 on lifespan (Guo et al., 2000). genes with age, which is in agreement with an observed
A final study mapped loci associated with longevity, increase in expression of oxidative stress genes in
circulating IGF1, and age at vaginal patency in a cross young individuals from long-lived C. elegans strains
between KK/HlJ and PL/J mice (Yuan et al., 2015). (McElwee, Bubb, & Thomas, 2003; Murphy et al., 2003).
These strains were selected based on a robust differ- Age-associated gene expression changes between C. ele-
ence in circulating IGF1 at 6 months of age (a differ- gans and D. melanogaster were directly compared in a
ence that is shown to be not dependent on differences single study that identified a conserved expression pro-
in locus Igf1q8, which is known to influence IGF1 gram involving mitochondrial metabolism and DNA
levels) and a lack of previous mapping studies using repair, among others (McCarroll et al., 2004). In 2009, a
either strain background. Combined, these studies meta-analysis of 27 microarray datasets examining dif-
have identified more than 60 suggestive or signifi- ferent tissues in humans, mice, and rats revealed a com-
cantly longevity-associated loci located throughout mon age-associated gene expression signature (de
the mouse genome (Table 1.4). As with humans, few Magalhães, Curado, & Church, 2009). This signature
loci are identified by more than one study. Six geno- included an age-associated increase in expression of
mic regions located on chromosomes 1, 2, 7, 8, 11, and genes to immunity and inflammation, as well as
16 were identified by at least two studies. In nine lysosome-associated genes, and a decrease in expression
cases—on chromosomes 1, 2, 5, 7, 10, 16, 17, and 19— of genes involved in mitochondrial function, cell cycle,
markers with suggestive association with lifespan senescence, and apoptosis. Further studies of this type
were identified by at least two studies within the will be of interest, particularly involving comparison of
same 10 Mb region. These regions represent the stron- gene expression patterns between invertebrates and
gest candidates for further investigation. Two regions mammals. For a more detailed discussion of specific
on chromosome 1 (120.0136.3 and 147.0185.5 Mb) microarray aging studies and associated technical chal-
and one region on chromosome 11 (5.228.9 Mb) are lenges, readers can refer to numerous reviews (Becker,
of particular interest, as they contain markers that 2002; Golden, Hubbard, & Melov, 2006; Han et al., 2004;
reached genome-wide statistical significance in two Melov & Hubbard, 2004; Nair, Golden, & Melov, 2003;
independent studies (Table 1.4). Werner, 2007).
Examining age-associated changes in gene expression
is also a useful and relatively noninvasive approach to
Mousehuman concordance
study aging in humans. The first study to examine age-
Based on the assumption that the longevity loci dependent changes in human gene expression on a
identified in corresponding human and mouse geno- genome scale used microarrays to examine peripheral
mic regions result from orthologous genes, Yuan, blood leukocyte transcripts and identified 295 genes
Peters, and Paigen (2011) integrated mouse and human with robust differential expression with age (Harries
data by projecting the identified human loci onto the et al., 2011). Similar studies have been performed in the
mouse genetic map. Out of 10 human GWAS regions, Framingham Heart Study and other cohorts that are
eight mapped within 10 Mb of a mouse QTL peak. The part of the Cohorts for Heart and Aging Research in
probability of this occurring by chance is very low Genomic Epidemiology (CHARGE) consortium (Peters
(P 5 .0025) (Yuan et al., 2011), strongly suggesting that et al., 2015). Human gene expression studies can be
the mechanisms responsible for variation in longevity used for two purposes downstream. The first is to iden-
within mouse and human populations is evolutionarily tify specific diagnostic transcripts that are useful bio-
conserved and validating the mouse as a model for markers of the biological age of the individual
human aging. (discussed further in the “Aging biomarkers” section of

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


16 1. Longevity as a complex genetic trait

TABLE 1.4 Significant and suggestive loci identified in genome-wide mouse mapping studies.Where available,
marker names were used to standardize all genomic locations to mouse genome assembly GRCm38. Genes listed are located within 10 kb
of a marker location. Bold 5 significant genome-wide association; not bold 5 suggestive genome-wide association.
Chr Region or marker location (Mb) Gene(s) References

1 13.3 Ncoa2 Murakami and Kaeberlein (2009)


31.534.5 (33.8) Zfp451 Linford et al. (2013)
120.0, 121.9133.7 (126.5) Gm101, Nckap5, Sctr Goffeau et al. (1996), Linford et al. (2013)

147.0185.5 (171.9), 148.8, 161.2, 167.8, Cd48, Fmn2, Gm10521, Lmx1a, Prdx6 Goffeau et al. (1996), Kamath et al. (2003),
174.6 Murakami and Kaeberlein (2009)
2 61.469.9 (65.4) Linford et al. (2013)

102.8, 108.3 Cd44 Goffeau et al. (1996), Steffen et al. (2009)


122.1, 124.6 B2m, Sema6d Goffeau et al. (1996), RNAi Resources | Source
BioScience (n.d.)
4 17.4151.1 (63.8), 22.5, 59.799.2 (80.5) Pappa, Pou3f2 Kamath et al. (2003), Rual et al. (2004), Steffen et al.
(2009)

5 25.0 Prkag2 Murakami and Kaeberlein (2009)


44.4 Murakami and Kaeberlein (2009)
64.6, 65.486.6 (79.5) Linford et al. (2013), Murakami and Kaeberlein
(2009)
6 55.2 Fam188b Murakami and Kaeberlein (2009)
73.8-83.5 (79.6), 84.4101.2 (93.5), 6030492E11Rik, Ctnna2, Foxp1 Linford et al. (2013)
91.5101.2 (99.0)
113.3121.9 (113.3) Camk1 Linford et al. (2013)
7 3.56.9 (3.5) Gm7789, Vstm1, Zscan4c Linford et al. (2013)
25.9, 26.8 Cyp2a5, Cyp2b10 Goffeau et al. (1996)
49.187.9 (84.6), 58.5, 65.7, 72.1, Gm10624, Gm19831, Hbb-y, Hbb-bh1, Fraser et al. (2000), Linford et al. (2013), RNAi
89.3107.3 (96.9), 103.9 Hbb-bh2, Mctp2, Tenm4, Zfand6 Resources | Source BioScience (n.d.), Timmons and
Fire (1998)
8 14.725.7 (14.7), 23.3 4930518F22Rik, Dlgap2, Golga7 Linford et al. (2013), Sutphin and Kaeberlein (2009)
36.0 Murakami and Kaeberlein (2009)
102.4115.8 (108.8) Zfhx3 Linford et al. (2013)

9 91.4 Zic1, Zic4 Steffen et al. (2009)


10 48.7 Timmons and Fire (1998)
67.0 Reep3 Steffen et al. (2009)
102.9116.1 (110.5), 119.7 Grip1 Linford et al. (2013), Sutphin and Kaeberlein (2009)
11 6.220.7 (8.3, 19.7), 16.824.4 (17.9), Etaa1, Slit3 C. elegans Sequencing Consortium (1998), Linford
17.126.8 (19.6), 35.739.1 (35.7) et al. (2013)
45.959.1 (55.8) Linford et al. (2013)
12 45.2 Gm24731 RNAi Resources | Source BioScience (n.d.)
105.1, 113.3 Igh-7, Igh-8 Goffeau et al. (1996), Steffen et al. (2009)

14 46.4 Bmp4, Gm15217 Murakami and Kaeberlein (2009)


16 5.7 Steffen et al. (2009), Timmons and Fire (1998)
20.2, 29.137.5 (32.3) 5830438M01Rik, Rnf168, Yeats2 Linford et al. (2013), Murakami and Kaeberlein
(2009)

(Continued)

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Nongenetic sources of complexity 17
TABLE 1.4 (Continued)
Chr Region or marker location (Mb) Gene(s) References

59.870.1 (64.6) Linford et al. (2013)


17 13.739.5 (34.5), 42.7, 50.1 Btnl6, Gpr111, Rftn1 Murakami and Kaeberlein (2009), Rual et al. (2004)
18 53.2 Timmons and Fire (1998)
19 32.0, 39.7 Asah2, Cyp2c69 Fraser et al. (2000), Timmons and Fire (1998)
47.2, 53.9 Calhm1, Pdcd4, Bbip1 Murakami and Kaeberlein (2009), Timmons and
Fire (1998)
X 45.458.6 (51.7) 2900011F02Rik, Hs6st2 Linford et al. (2013)
122.3155.6 (130.4) 9530062E16Rik, Diap2 Linford et al. (2013)

this chapter). Biomarkers are not necessarily causal 50 genes (61%) significantly affected lifespan (Sutphin
players in the aging process, but simply consistent cor- et al., 2017). This approach has the advantage of nar-
relates with the health state of the individual. The sec- rowing candidate genes to those that likely have an
ond purpose is to identify candidate genes that play a evolutionarily conserved role in aging at the cost of
causal role in the aging process. A disadvantage in discarding genes with a human- or mammal-specific
human gene expression studies is the limited availabil- role in aging. In the same vein, this method is not
ity of different tissue types. The question arises as to suitable for all mammalian candidates, as roughly half
whether gene expression in leukocytes accurately repre- of human and mouse genes do not have clear worm
sents what happens in the whole organism. Mice and orthologs.
other animal models can be used to measure gene
expression in different tissues in order to identify both
genes associated with tissue-specific aging and genes
with common age-associated expression patterns across
Nongenetic sources of complexity
tissues.
After GWA studies and human expression studies
Tissue-specific aging
identify candidate genes, the next step is to validate One important aspect of aging is the differences
the capability of each gene to impact aging, character- between tissues. In human populations, mortality can
ize their role in the aging process, and determine why result from pathology in virtually every organ system,
the specific causal allele leads to variation in lifespan. though genetic and environmental factors can increase
For genes that are consistently identified in multiple risk for tissue-specific diseases. This suggests that differ-
studies, such as APOE and FOXO3, direct follow-up in ent tissues generally age at similar rates on average
a mammalian system may be appropriate. Indeed, within the human population, but that different tissues
APOE is under intense scrutiny, particularly in the can experience different rates of aging between indivi-
areas of cardiovascular research, cholesterol metabo- duals. In C. elegans, studies of tissue-specific aging have
lism, and neurodegenerative disease. The creation of a concluded that muscle cell function tends to gradually
genome-wide knockout mouse strain collection and decline beginning around the transition to the post-
novel gene-editing technologies—particularly the reproductive life stage, while neurons largely retain
recent and widespread adoption of the CRISPR/Cas9 function in old animals (Herndon et al., 2002). The
system—for rapid introduction of novel variants into decline in muscle function is accompanied by decreased
inbred test populations allows for rapid progression pharyngeal pumping, resulting in reduced food con-
from gene identification to characterization. In the next sumption (Huang, Xiong, & Kornfeld, 2004; Kenyon,
tier, where the number of candidate genes makes Chang, Gensch, Rudner, & Tabtiang, 1993; Smith et al.,
direct characterization in mammalian models cost- 2008) and autofluorescent age pigment accumulation
prohibitive, the use of RNAi feeding libraries in C. ele- throughout the body (Gerstbrein, Stamatas, Kollias, &
gans allows for rapid screening of large numbers of Driscoll, 2005; Klass, 1977). Characterizing these differ-
candidate genes for direct effects on lifespan. We ences, understanding the underlying molecular mecha-
recently demonstrated this approach by screening 82 nism, and determining how these mechanisms result in
genes from the CHARGE gene expression study age-related disease will be important in developing
(Peters et al., 2015) in C. elegans. RNAi knockdown of effective treatments.

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


18 1. Longevity as a complex genetic trait

Tissue-specific age-related DNA methylation kinds of differences are also found for ER stress and
One source of differential aging among tissues autophagy where variation leads to different outcomes
appears to be epigenetics. Studies in rats (Thompson depending on the tissue. Fat-specific insulin receptor
et al., 2010), mice (Maegawa et al., 2010), and humans knockout (FIRKO) mice live B18% longer than wild-
(Day et al., 2013; Levine et al., 2018) have identified type mice and are protected against obesity and
tissue-specific patterns of age-related DNA methyla- related glucose intolerance (Blüher et al., 2002). In con-
tion. In mammals, DNA methylation occurs mostly trast, pancreatic β-cell knockout (βIRKO) mice have an
within the context of CpG dinucleotides. Clusters of insulin secretion defect resembling type II diabetes
CpG dinucleotides—CpG islands—are often located (Kulkarni et al., 1999) while muscle-specific insulin
near the 5’ end of genes. Methylation of CpG islands is receptor knockout (MIRKO) recapitulates some fea-
associated with a closed chromatin structure and tran- tures of diabetes (increased fat mass, serum triglycer-
scriptional silencing of the gene. Aging leads to epige- ides, and free fatty acids) but is normal with respect to
netic dysregulation, with different sites showing either others (blood glucose, insulin, and glucose tolerance)
hypermethylation or hypomethylation. This process is (Brüning et al., 1998).
common across tissues for certain loci and tissue spe- Many of the processes that play a primary role in
cific for others. Centenarians display delayed age- aging are core regulators of cellular growth and metab-
related methylation changes and can even pass the olism in response to environmental queues. These
preservation of methylation states on to their offspring examples highlight several important challenges both
in a manner independent of genotype (Gentilini et al., in developing a comprehensive understanding of the
2013). How the observed dysregulation of methylation role that known aging genes play in the aging process
patterns impacts gene expression, activity in aging- and in translating basic aging discoveries into clinical
relevant pathways, or progression of specific age- applications. An intervention that increases lifespan
related diseases has yet to be fully elucidated. Which may provide an overall benefit while driving disease
loci are controlling differences in age-related methyla- processes in specific tissues. Conversely, a beneficial
tion and how variation in these loci can lead to differ- impact on aging in one tissue may be masked by a det-
ences in age-related phenotypes is also unknown. rimental impact on another when an intervention is
The epigenetics of aging and the use of epigenetic applied to the whole organism, resulting in a net
signatures as “clocks” (see the section on “Aging increase in mortality. Factors with this type of outcome
Biomarkers” later in this chapter) that measure biologi- would not expect to be identified in the type of genetic
cal age are an area of substantial research effort and screens or gene mapping studies described above.
the topic of recent detailed reviews (Ecker & Beck, Clinical treatment may require altering the interven-
2019; Gabbianelli & Malavolta, 2018; Guevara & tion agent or delivery method to target a subset of tis-
Lawler, 2018; Horvath & Raj, 2018; Wagner, 2019). sues or developing a combined therapy with a second
intervention to counteract the negative impacts of the
first. Interventions that target the fundamental aspects
Tissue-specific responses of aging pathways of aging—for instance, using senolytic drugs to clear
Distinct tissues experience different changes in func- accumulated senescent cells (see other chapters)—are
tion with age, respond differently to different forms efficacious in only a subset of tissue types and will
and combinations of stress, and have differential likely need to be used in combination to achieve bene-
access to endogenous and exogenous factors in circula- fit in all tissues.
tion. Understanding how tissue-specific function and
the interaction between tissues changes during aging
will be critical to developing efficacious interventions
Geneenvironment interaction
to slow aging at the level of the organism. Two
changes in activity through two of the central aging This chapter so far has discussed organism-intrinsic
pathways—IIS and mTOR signaling—have dramati- factors contributing to aging. Extrinsic factors—and the
cally different effects in different tissues. Knocking interaction between extrinsic and intrinsic factors—
out RAPTOR, a component of mTOR complex 1 must also be considered when developing a complete
(mTORC1), in adipose tissue shows increased leanness model of organismal aging. Extrinsic factors that impact
and resistance to diet-induced obesity accompanied by aging include any aspect of the environment that an
improved glucose tolerance and insulin sensitivity organism interacts with: diet, weather, temperature,
(Polak et al., 2008). In contrast, knocking out RAPTOR microflora, other members of the same species, chemical
in skeletal muscle leads to muscular dystrophy associ- stressors such as external reactive oxygen and nitrogen
ated with reduced mitochondrial biogenesis and mus- species, etc. Aging research has led to the discovery of
cle oxidative capacity (Bentzinger et al., 2008). These numerous genetic, pharmacological, and environmental

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Nongenetic sources of complexity 19
interventions that increase lifespan in one or more identifying the genes responsible for the beneficial
model systems in the laboratory; however, interventions response to dietary restriction is the development of
capable of extending lifespan in the laboratory setting dietary restriction mimetics, pharmacological agents
may turn out to be sensitive to context. Understanding capable of reproducing the beneficial outcomes without
how the interaction between an organism’s genetic— altering diet. Rapamycin and other pharmacological
and epigenetic—identity interacts with its environment inhibitors of mTOR signaling (termed “rapalogs”) are
will be important to understanding how longevity inter- being pursued in basic aging research as well as clinical
vention studies in the lab will translate into clinical trials for multiple age-related diseases. Understanding
application in human populations. the molecular impacts of mTOR signaling and the
development of rapalogs are highly active areas of cur-
Genetic response to dietary restriction rent investigation in aging science (Johnson et al., 2013;
Several methods of environmental manipulation are Kaeberlein, 2013; Lamming et al., 2013).
known to impact lifespan. Dietary restriction, typically The second pathway that has been studied exten-
defined as a reduction in dietary intake without mal- sively with respect to the interaction with dietary
nutrition, is the most studied environmental interven- restriction is IIS. In the majority of studies in C. elegans
tion capable of increasing lifespan. Dietary restriction and Drosophila, epistasis analysis appears to place IIS
is commonly put forward as the most consistent means into a pathway at least partially distinct from dietary
of increasing longevity; indeed reducing dietary intake restriction (Giannakou et al., 2008; Houthoofd et al.,
has been shown to extend lifespan in most organisms 2003; Kaeberlein, 2006; Lakowski & Hekimi, 1998; Lee
where it has been attempted, including yeast, worms, et al., 2006); however, other studies do report interac-
fruit flies, spiders, mice, rats, hamsters, dogs, and rhe- tion under specific forms of dietary restriction or
sus monkeys (Colman et al., 2014; Kennedy, Steffen, & genetic interventions (Clancy et al., 2002; Greer et al.,
Kaeberlein, 2007; Masoro, 2005; Weindruch & Walford, 2007; Iser & Wolkow, 2007). Lifespan of the long-lived
1988). The complete picture is more complicated. The growth hormone receptor knockout (GHRKO) mice,
outcome of dietary restriction depends on a range of which have reduced levels of insulin and IGF-1, is not
factors including degree of restriction, the specific further increased by dietary restriction (Al-Regaiey
composition of both baseline and restricted diets, feed- et al., 2007; Bonkowski et al., 2006). In contrast, dietary
ing schedule, age at onset, and the genetic identity of restriction is capable of increasing lifespan of mice with
the subject population. pituitary mutations that cause a defect secretion of sev-
A large body of research is directed at understanding eral hormones including growth factor (Bartke et al.,
the biological processes underlying the beneficial effects 2001). The combined evidence indicates that dietary
of dietary restriction. A number of genetic pathways restriction is not dependent on IIS to impact lifespan,
have been proposed as mediators of dietary restriction. but that the two pathways interact, perhaps by influenc-
The evidence is most consistent for mTOR signaling, a ing overlapping sets of downstream factors or through
highly conserved nutrient-responsive signaling pathway compensatory feedback mechanisms. The genetic tar-
that regulates many cell growth and proliferation pro- gets of dietary restriction are the topic of many review
cesses. Lifespan extension by dietary restriction and articles (Anderson & Weindruch, 2010; Fontana et al.,
reduced mTOR signaling are nonadditive in yeast, 2010; Kennedy et al., 2007; Lee & Longo, 2016; Schleit
worms, and flies. Dietary restriction both reduces activ- et al., 2012; Swindell, 2012; Xiang & He, 2011).
ity through the mTOR signaling pathway and induces A central question in dietary restriction research is
mTOR-mediated changes to metabolism and protein whether the positive effects observed in genetically
homeostasis. Together, the accumulating invertebrate homogeneous laboratory populations will be widely
evidence places dietary restriction and mTOR signaling realized if dietary restriction is applied to a genetically
into at least partially overlapping pathways. In mice, diverse population, such as humans. Several studies
reducing mTOR signaling through genetic manipulation suggest that the genetic makeup of an individual can
of multiple factors increases lifespan (Lamming et al., profoundly change the impact of dietary restriction on
2012; Selman et al., 2009; Wu et al., 2013). In multiple lifespan and healthspan. Dozens of dietary restriction
studies using inbred and outbred populations rapamy- experiments have been carried out in mice using a
cin increases lifespan, reduces cancer incidence, and variety of strain backgrounds (reviewed in depth by
improves tissue-specific decline with age (Anisimov Swindell, 2012). While lifespan extension is observed
et al., 2011; Harrison et al., 2009; Miller et al., 2011; Neff in most cases, there are numerous examples—most
et al., 2013; Wilkinson et al., 2012; Zhang et al., 2014). studies using DBA/2 male mice for instance—where
Despite similar outcomes in mice, a direct link between dietary restriction appears to have no effect or even
dietary restriction and mTOR signaling has yet to be shortens lifespan. Two studies have examined the
established in a mammalian system. One goal of impact of dietary restriction on lifespan in ILSXISS

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


20 1. Longevity as a complex genetic trait

mice, a set of recombinant inbred strains derived from dietary changes from quantity to composition to tempo-
eight inbred founders designed for variation in sensi- ral consumption patterns, and the range of biological
tivity to ethanol sedation (Williams et al., 2004). In processes underlying the response appears to vary
both studies dietary-restricted mice were fed a diet according to the specific form of dietary restriction
with 40% fewer calories than strain-matched ad employed.
libitum-fed mice (Liao et al., 2010; Rikke et al., 2010). In C. elegans, multiple forms of dietary restriction are
The results are striking: roughly half of the strains commonly used (Greer & Brunet, 2009; Mair et al.,
showed no significant change in lifespan, 5%25% dis- 2009). Most methods involve reducing the amount of
played increased lifespan, and 20%25% displayed bacterial food provided to the worms, but differ in the
decreased lifespan in response to dietary restriction. In amount of bacteria provided. Other variables include
yeast, a screen of 166 single-gene deletion strains whether the growth environment is liquid or solid agar-
found a similar variability in response to dietary based, whether the food is alive or killed, and whether
restriction ranging from a 79% decrease to a 103% the amount of food is constant or varied (akin to feed-
increase in mean replicative lifespan (Schleit et al., ing/fasting cycles in mammals) (Greer & Brunet, 2009).
2013). A meta-analysis of reported mouse dietary Under some conditions an extreme form of dietary
restriction longevity studies found that dietary restric- restriction called bacterial deprivation, referring to the
tion may be more beneficial in outbred populations complete removal of the food during adulthood, results
than inbred strains (Swindell, 2012), suggesting that in optimal lifespan extension (Kaeberlein, 2006; Lee
inbreeding depression may limit the effectiveness of et al., 2006). Shifting a population to dietary restriction
dietary restriction. Confounding this possibility, die- can result in early mortality in a fraction of individuals
tary restriction failed to increase lifespan in male wild- depending on whether the shift is gradual and the age
derived mice (grand-offspring of wild-caught mice) at onset, and these factors can also vary from study to
which were never inbred (Harper et al., 2006). Dietary study. In the case of bacterial deprivation, similar
restriction appeared to cause an increase in early life median lifespan extension can be achieved for dietary
mortality and a decrease in late-life mortality in this restriction initiated between days 4 and 14 and similar
population however, resulting in a significant increase maximal lifespan extension can be achieved for dietary
in maximum lifespan despite the lack of mean lifespan restriction initiated as late as day 24 (Smith et al., 2008),
extension. Taken together, these studies point to a while starting dietary restriction earlier than day 4 often
strong genotype dependence in the individual results in early mortality in a subset of worms. A com-
response to dietary restriction and promote further mon genetic model of dietary restriction in worms is
investigation of dietary restriction in genetically het- loss of function mutations in the eat-2 gene, which
erogeneous populations. results in defective pharyngeal pumping and thus
reduced bacterial food consumption. An additional
Dietary restriction: quantity, composition, layer complicating the impact of dietary restriction on
and timing lifespan is food sensing. Smith et al. (2008) found that
Each dietary restriction study uses a defined change simply the ability to detect food in the local environ-
in diet, usually a specific percent reduction in food, but ment was sufficient to partially suppress the increased
what range of interventions does dietary restriction lifespan from bacterial deprivation.
encompass? An earlier term—caloric restriction— In yeast, dietary restriction typically refers to reducing
reflected the idea that net caloric intake was the pri- the media glucose concentration from 2% to 0.5% or lower
mary factor causing increased longevity and other (Lin et al., 2000), with optimal lifespan extension for the
health benefits. From this perspective, dietary restriction yeast ORF deletion collection strain background achieved
can be seen as a shift on a dietary doseresponse curve at 0.05% glucose (Kaeberlein et al., 2004; Lin et al., 2000).
and the genotype-dependent dietary response can be Less common forms of dietary restriction include restrict-
interpreted as some individuals being closer than others ing amino acids (Jiang et al., 2000) or replacing media glu-
to the optimal dietary intake with respect to longevity cose with a nonfermentable carbon source such as
when fed ad libitum due to genetically defined traits glycerol, ethanol, or raffinose (Delaney et al., 2011;
such as metabolic rate or appetite. In individuals for Kirchman & Botta, 2007). Deletion of HXK2, which
which ad libitum is near optimum, a typical dietary encodes a hexokinase responsible for converting glucose
restriction regimen may result in malnutrition and thus into glucose-6-phosphate prior to entry into the glycolytic
be detrimental to longevity. Even this view represents a pathway, extends replicative lifespan (Lin et al., 2000) and
simplified model of dietary restriction that does not has been used as a genetic model of dietary restriction
take into account the myriad of components that make (Walsh et al., 1983), though it remains unclear whether
up a diet even in the most basic organism. The current this is attributable to reduced cellular hexokinase activity
conception of dietary restriction includes a range of (Rodrı́guez et al., 2001; Walsh et al., 1991).

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Nongenetic sources of complexity 21
Drosophila has been used extensively to explore non- in the source and quantity of fat, protein, carbohy-
genetic manipulations that extend lifespan and the drate, and supplemental vitamins. A number of mouse
Drosophila aging community has perhaps done the studies highlight the interaction between genotype and
most to characterize the impact of dietary composition dietary composition based on these differences. In 22
on lifespan. Flies are typically fed ad libitum on a glu- studies examining the impact of dietary restriction on
coseyeast or cornmealyeastsugar mixture in agar C57BL/6 mice, the observed change in median lifespan
and dietary restriction accomplished by diluting one ranges from a 32.8% decrease to a 26.8% increase
or more of the component ingredients (Bass et al., (Swindell, 2012). C57BL/6 mice are susceptible to ath-
2007; Chapman & Partridge, 1996; Good & Tatar, erosclerosis and obesity and, at least in some instances,
2001). A standard dietary restriction experiment will the failure of dietary restriction to extend lifespan may
examine multiple dilutions of the selected dietary com- result from a lower fat content in the chow (Cheney
ponent, rather than only comparing two nutrition et al., 1980; Harrison & Archer, 1987; Swindell, 2012;
points, thus examining the impact of the intervention Turturro et al., 1999). Wild-derived mice are smaller
under scrutiny across an entire dietary dose response. but eat more as a percentage of body weight than
One study mapped the impact on longevity and other inbred laboratory strains, suggesting a faster metabo-
metabolic parameters of diets with a range of different lism that may account for the less favorable response
yeast and glucose concentrations, finding that a diet to dietary restriction (Austad & Kristan, 2003). In a
with a balance of yeast and sugar results in optimal study analogous to that examining Drosophila dietary
lifespan while a high-yeast diet improves fecundity composition by Skorupa et al. (2008), Solon-Biet et al.
(Skorupa et al., 2008). Like worms, food sensing can (2014) fed mice a range of diets that varied not only in
modulate fly longevity independent of food consump- total caloric content, but also in the balance of protein,
tion (Libert & Pletcher, 2007). fat, and carbohydrate, and measured lifespan and
The most common method for dietary restriction in other metabolic parameters. They found that the die-
mice is to restrict access to food based on the con- tary protein and carbohydrate content, and not total
sumption observed in ad libitum fed control, with typ- calories or fat content, were the primary determinants
ical restriction levels ranging from 60% to 70% of ad of lifespan. In particular, high-carbohydrate low-pro-
libitum. An alternative method is to not limit quantity, tein diets maximized lifespan. A detailed review of
but instead restrict the time that animals have access aging studies examining dietary composition was
to food (intermittent feeding). Providing access to food recently published by Le Couteur et al. (2016).
only every other day has been shown to increase life- Different forms of dietary restriction interact with
span in both rats (Goodrick et al., 1982; Goodrick et al., different genetic pathways. As mentioned in the dis-
1983a, b) and mice (Goodrick et al., 1990). In mice, the cussion of IIS, bacterial deprivation is capable of
extension only occurred when started in young adult increasing lifespan independently from the insulin
mice and the degree of extension was greater in receptor daf-2 or the FOXO family transcription factor
C57BL/6J and C57BL/6JxA/J F1 hybrid mice than in daf-16, placing it in a pathway parallel to IIS. sDR, a
A/J mice (Goodrick et al., 1990). Intermittent feeding form of dietary restriction where worms are main-
also reduced serum glucose and insulin and increase tained on solid agar-plates in the presence of diluted
neuronal stress resistance in mice, despite a similar net bacteria, requires daf-16 to increase lifespan, indicating
caloric intake to continuously ad libitum fed mice that it acts, at least in part, through IIS. The precise
(Anson et al., 2003). Simply, restricting the dietary molecular mechanisms through which dietary restric-
methionine is sufficient to increase lifespan in both tion acts to extend lifespan in yeast are not yet known;
mice and rats (Miller et al., 2005). A similar outcome however, it is commonly thought that dietary restric-
results from restricting tryptophan in rats (Ooka et al., tion manipulates these mechanisms, at least in part, by
1988; Segall and Timiras, 1976; Timiras et al., 1984), influencing several partially redundant nutrient-
although these results may simply point to an over- responsive signaling kinases, including mTOR, cyclic
abundance of methionine and tryptophan in the labo- AMP-dependent protein kinase (PKA), and Sch9.
ratory diet. Indeed, Masoro et al. (1989) found that Mutants with reduced activity for any of these kinases
methionine restriction did not contribute to the life- have long replicative lifespans that cannot be further
span extension resulting from a 40% decrease in over- extended by dietary restriction (Fabrizio et al., 2004;
all food intake in rats, suggesting that specific dietary Kaeberlein et al., 2005; Lin et al., 2000). Yeast PKA is
components may impact phenotypes via distinct bio- an essential complex consisting of three catalytic subu-
logical processes. While the baseline diet is typically nits and regulated by two upstream sensing pathways,
standardized within an institution, and even between one involving RAS and the other a G protein-coupled
institutions when a common supplier and product are receptor system. Two genes, GPA2 and GPR1, encode
used, published studies use a range of diets that vary subunits of the G protein-coupled receptor. Mutants

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


22 1. Longevity as a complex genetic trait

lacking either GPA2 or GPR1 are replicatively long- throughout adult life have substantially decreased life-
lived relative to wild-type and are commonly used as span, with the caveat that these experiments were not
models of reduced PKA activity (Lin et al., 2000). The carried out under specific pathogen-free (SPF) condi-
third kinase, Sch9, shows sequence homology to Akt tions. Immersing rats in 23 C water to reduce body
kinase, a component of IIS (Burgering & Coffer, 1995; temperature for 4 hours/day, 5 days/week under SPF
Paradis & Ruvkun, 1998), but also functions as a ribo- conditions causes a nonsignificant trend toward
somal S6 kinase, a substrate of mTOR and regulator of increased lifespan, and also results in a 44% increase
translation in multicellular eukaryotes (Powers, 2007; in food consumption and a reduced incidence of can-
Urban et al., 2007). While yeast does not possess a for- cer offset by a higher incidence of myocardial fibrosis
mal IIS pathway, Sch9 may fulfill an equivalent role in (Holloszy & Smith, 1986). Maintaining mice at 10 C
both Akt and S6 kinases in multicellular eukaryotes. throughout life has no effect on lifespan relative to
control mice maintained at 22 C (Vaanholt et al., 2009).
Other environmental factors that influence aging Additional research will be needed to determine
A primary determinant of invertebrate lifespan is whether animals that actively maintain body tempera-
temperature. Culture temperature can dramatically ture will respond similarly to environmental changes
impact lifespan in worms (Klass, 1977; Lakowski & in temperature as invertebrate models. The answer
Hekimi, 1996; Leiser et al., 2011, 2016; Mendenhall will likely involve a mix of responses that are both
et al., 2017; Miller et al., 2017; Sutphin et al., 2012; Van shared (e.g., the activation of heat-shock proteins and
Voorhies & Ward, 1999), flies (Helfand & Rogina, 2003; other temperature-stress response machinery) and
Loeb & Northrop, 1917; Miquel et al., 1976), and chro- unshared (e.g., temperature-dependent changes in the
nologically aging yeast (MacLean et al., 2001). In rate of catalytic process in poikilotherms).
worms there are clear differential interactions between The molecular response of organisms to a variety of
genotype and environmental temperature (Miller et al., environmental stressors can be leveraged to extend
2017; Sutphin et al., 2017). Knockdown of the gene lifespan. Heat-shock—the short-term exposure to high
encoding the hypoxia inducible factor, hif-1, increases temperature—increases lifespan in worms (Cypser &
lifespan at 25 C but not at 15 C or 20 C, while stabili- Johnson, 2002; Lithgow et al., 1995; Olsen et al., 2006),
zation of HIF-1 protein by knocking down the E3 flies (Hercus et al., 2003; Le Bourg et al., 2004), and
ligase VHL-1 increases lifespan at all three tempera- chronologically aging yeast (Harris et al., 2001), and
tures, but to a much greater extent at 15 C (Leiser replicatively aging yeast (Shama et al., 1998) through
et al., 2011). Conversely, maintaining adult worms on hermetic activation of heat-shock factors. A similar
plates containing up to 20 mM caffeine increases life- outcome can be achieved by short-term exposure to
span at both 15 C and 20 C, but not 25 C (Sutphin cold, oxidative stress, hypergravity, or radiation
et al., 2012). Knockdown of rsks-1, encoding the ribo- (Cypser & Johnson, 2002; Le Bourg, 2007; Le Bourg &
somal protein S6 kinase, has opposite effects on life- Minois, 1997; Vaiserman et al., 2003).
span at 15 C and 25 C, while knockdown of daf-2, Flies generally have a strong inverse correlation
encoding the insulin/IGF-1-like receptor, extends life- between reproduction and longevity. Strains bred for
span to a similar degree across the temperature spec- longevity by selecting offspring from late life repro-
trum (Miller et al., 2017). Different genetic programs duction show reduced egg laying early in life relative
therefore have variable impact on longevity depending to ancestral strains (Luckinbill et al., 1984; Rose, 1984).
on environmental temperature. Preventing mating can also double female lifespan
Whether the temperature-responsive processes that (Smith, 1958), though in D. melanogaster seminal factors
strongly influence aging in poikilothermic invertebrate have been implicated in shortening female lifespan as
species will have clear analogs in homeothermic verte- opposed to some intrinsic cost associated with repro-
brate species remains to be determined and the few duction (Ueyama & Fuyama, 2003).
mammalian studies that have directly examined the
impact of temperature on longevity do not provide a
clear answer (see the review by Keil et al., 2015). Rats Emerging tools for studying complex genetic
maintained at 34 C lived significantly longer than rats traits
maintained at 28 C; however, the high temperature
also caused a reduction in food consumption, and sim- The maturing field of genomics and its derivatives—
ply restricting food to the same degree at 28 C resulted epigenomics, transcriptomics, proteomics, metabolomics—
in an even greater increase in longevity (Kibler & in combination with genome-scale lifespan screens and
Johnson, 1966). Conversely, rats maintained at 6 C gene-mapping techniques allow us to begin to glimpse in
(Heroux & Campbell, 1960) or 9 C (Johnson et al., detail the changes associated with complex phenotypes
1963; Kibler & Johnson, 1961; Kibler et al., 1963) like longevity at the level of a whole organism. As these

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Emerging tools for studying complex genetic traits 23
fields continue to advance, new tools are being developed on solid NGM-agar plates, though the worms cultured in
to help assemble the vast amounts of information gener- WormFarm tended to be physically smaller.
ated into a form amenable to identification of novel genes The second system is an automated solid NGM
and key regulatory junctions in the network of factors that plate-based platform called the C. elegans Lifespan
integrate to determine longevity and the processes under- Machine (Stroustrup et al., 2013). In the Lifespan
lying age-associated disease. Leveraging these tools will Machine worms are cultured on solid NGM-agar
inform future studies and pinpoint targets for clinical plates that are maintained on a series of modified flat-
intervention. The following sections outline emerging bed scanners. Scanners capture periodic images of
tools that are allowing us to encompass the range of fac- worms on each plate which are processed in a custom
tors that contribute to age-associated phenotypes and software package to identify worms and assess viabil-
define their interaction. ity based on movement over time. Stroustrup et al.
(2013) demonstrate the capability of the Lifespan
Machine to generate reproducible survival data that
are in agreement with published studies when applied
High-throughput life span assays in yeast
to mutant strains, worms treated with RNAi-
and worms expressing bacteria, and stress response. In addition to
S. cerevisiae and C. elegans are the primary models survival, the Lifespan Machine is capable of character-
used in genome-scale reverse genetic screens to izing body size, body shape, and motility. The
directly identify genes capable of influencing lifespan, throughput of this system is B560 worms per scanner.
due mainly to the combination of short lifespans, ease The Lifespan Machine has been adopted as the life-
of manipulation in the laboratory setting, and the abil- span platform for the CITP (Lucanic et al., 2017).
ity to systematically target individual genes in a high- In 2017, Churgin et al. (2017) presented a third sys-
throughput manner. With the exception of yeast chro- tem called the WorMotel. The WorMotel features a
nological lifespan, experimental techniques for deter- custom-molded polydimethylsiloxane (PDMS) plate
mining lifespan are labor-intensive and studies that consists of a set of 240 wells laid out on a grid
published in the past few years have established the with the same spacing as a standard 384-well plate
first proof-of-principle demonstrations for methods that allows culturing of individual C. elegans on solid
and technology capable of true high-throughput analy- media. Each well effectively acts as a single-worm
sis of worm lifespan and yeast replicative lifespan. Petri plate surrounded by a moat filled with an aver-
Two studies were published in 2013 for automated, sive agent (e.g., copper sulfate) to keep the animals in
high-throughput life analysis systems in C. elegans. the well. The plate is paired with a camera system that
WormFarm employs an integrated microfluidic platform takes images and video of the worm at set intervals,
to maintain worms in liquid culture in 3050 worm allowing for automated monitoring of activity (to
groups using a customized transparent eight-chamber assess healthspan) and longevity determination. While
chip (Xian et al., 2013). The microfluidics chip is designed the initial report did not report high-throughput data,
to contain adult animals while refreshing the bacterial it did include a system design that paired a plate-
food supply and automatically removing larva, which can handling robot with the WorMotel camera system to
be collected for further analysis. Photographs and video allow the potential for high-throughput monitoring of
taken periodically throughout the experiment allow for a large number of WorMotel plates.
automated collection of phenotypic data. In addition to The WormFarm, Lifespan Machine, and WorMotel
lifespan, the transparent WormFarm chips allow auto- demonstrate the potential for high-throughput, auto-
mated age-dependent characterization of body size and mated analysis of worm lifespan, and both provide a
shape, motility, development rate, and brood size, as well platform for measuring lifespan, body dimensions, and
as quantification of fluorescent markers when combined motility with little manual labor after initial setup. The
with fluorescence microscopy. After the initial setup, the primary advantage to the Lifespan Machine and
prototype WormFarm was capable of determining life- WorMotel over WormFarm is the ability to culture
span for B320 worms per chip without manual interven- worms on NGM-agar, allowing direct comparison to the
tion. Xian et al. (2013) demonstrated the experiment-to- majority of published worm studies. WormFarm also
experiment consistency and agreement with published necessarily excludes strains that are intolerant of liquid
data when WormFarm was applied to a variety of com- culture. Despite this limitation, WormFarm offers a
mon survival assays, including comparison of mutant greater range of automated phenotyping, including
strains, treatment with RNAi-expressing bacteria, stress quantification of brood size and development, monitor-
response, and changes in media composition. In the ing of fluorescent markers, and collection of larvae for
strains and under the conditions examined, WormFarm further analysis. WormFarm also provides precise con-
produced similar survival data to manual measurement trol over the entire environment experienced by the

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


24 1. Longevity as a complex genetic trait

worm, including temporal control over media composi- for biochemical and gene-expression studies than earlier
tion, bacterial food concentration, and temperature. The size-separation techniques. In addition to applications
ability to wash away larvae also alleviates the need to in young versus old comparisons, the MEP has been
use chemical or genetic means to prevent reproduction, proposed as a high-throughput method to measure rep-
thus removing a possible confounding factor. The pri- licative lifespan. The viability of MEP cultures is deter-
mary advantage to the WorMotel system is the capabil- mined specifically by the replicative lifespan of the
ity to monitor individual worms over the course of their mother cells. The use of a plate reader system, such as
lifespan and correlate longevity to individual pheno- that used to measure chronological lifespan (Murakami
types collected earlier in life. Each system is, in princi- et al., 2008) can allow rapid, high-throughput measure-
ple, scalable to any desired size. The current conception ment of replicative lifespan for strains carrying the MEP
of the Lifespan Machine is more limited in this regard, biological machinery in liquid media. Since the MEP
as it requires the plates to be left in place throughout requires multiple genetic modifications, this technique
analysis, thus necessitating a new scanner for each set of will not likely be useful for large-scale genetic screens.
B560 worms to be run in parallel; however, this should There is promise for the application of MEP to replica-
be remediable in future versions by using smaller and/ tive lifespan drug screening, where current labor-
or mobile optics or plate handling. A common challenge intensive methods for measuring replicative lifespan
among these systems is the storage and analysis of large limit the number of drugs that can be easily tested, and
quantities of imaging data. Aside from the development where further genetic manipulation of the strains is not
of more efficient analysis or advances in storage and necessary.
processing technology, there is not a clear direction for In an approach that resembles the chemical cell
escaping this problem. Comparing the automated sur- labeling approached used by Belak et al. (2018) to mea-
vival analysis data, the Lifespan Machine and WorMotel sure chronological lifespan, Sarnoski et al. (2017)
appear to produce much smoother survival curves than developed the high-throughput replicative lifespan
WormFarm. This may result partially from more closely measurement (High-Life) system, which uses fluores-
spaced time points, but is likely also a result of the for- cently labeled yeast in combination with flow cytome-
mer systems providing a more precise determination of try to rapidly measure replicative age. High-Life yeasts
individual time of death. are labeled with three fluorescent markers: (1) fluores-
In yeast, new approaches to automated quantifica- cein conjugated N-hydroxysuccinimide-ester (NHS-
tion of both chronological and replicative lifespan are ester), which is asymmetrically segregated to daughter
being developed. For chronological lifespan, Belak cells and allows selection of progenitor cells; (2) the
et al. (2018) used the reduction of 3-(4,5-dimethylthia- viability dye propidium iodide to identify viable cells;
zol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; yel- and (3) the protein lectin wheat germ agglutinin conju-
low in color) to formazan (purple in color), which only gated to the blue fluorophore CF405M, which selec-
occurs in viable S. cerevisiae (Hodgson et al., 1994), to tively labels bud-scars and provides a quantitative
quantify the viable fraction in chronologically aging measure of the replicative age of viable cells. Sarnoski
yeast, allowing rapid and high-throughput quantifica- et al. (2017) used High-Life to screen 2640 compounds
tion of chronological lifespan. This method success- for increased replicative lifespan and identified terreic
fully recapitulated lifespan changes resulting from acid and mycophenolic acid as novel pro-longevity
glucose restriction, several short- and long-lived dele- compounds. This initial application of High-Life was
tion mutants, and several pro-longevity drugs (Belak carried out in 384-well plates and used the MEP strain
et al., 2018). background to prevent the yeast from exhausting the
In the replicative paradigm, Lindstrom and limited volume of growth media (MEP yeast exhibit
Gottschling (2009) constructed a genetic platform called linear, rather than exponential population expansion).
the Mother Enrichment Program (MEP) to selectively Because of the complicated genetic background of
enrich yeast colonies for replicatively aged mother cells. MEP yeast, this version of High-Life will be useful for
In this system, Cre-lox recombination is used to specifi- drug screening but will be of limited use for genetic
cally disrupt two essential genes, UBC9 and CDC20, in screening. In principle, the High-Life methodology can
newly formed daughter cells, thus preventing cell divi- be employed with any strain background using alter-
sion in the daughter cells without altering the replica- native strategies to replenish exhausted media.
tive capacity of the mother. In MEP cultures, daughter The most prominent trend in yeast aging is the
cells are produced but do not continue dividing, result- emergence of microfluidic systems to replace the time-
ing in a decaying population growth and an enrichment and labor-intensive practice of manual microdissection
for replicatively aged mother cells over time relative to of daughter cells to measure yeast replicative lifespan
non-MEP cultures. This enrichment allows for single- (Crane et al., 2014; Fehrmann et al., 2013; Huberts
step affinity purification of much older cell populations et al., 2013a, b, 2014; Jo et al., 2015; Lee et al., 2012; Liu

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Emerging tools for studying complex genetic traits 25
et al., 2015; Meitinger et al., 2014; Rafelski et al., 2012; computationally intensive and still requires some
Spivey et al., 2014; Xie et al., 2012, 2015; Zhang et al., human input. Despite current limitations, the primary
2012; recently reviewed by Chen et al., 2017). In gen- challenge of automated lifespan determination has
eral, these systems use microfluidic devices to chemi- been met and the remaining technical problems
cally and/or mechanically trap mother cells. The should be solvable in subsequent design iterations.
geometry of the device is designed to selectively trap The application of high-throughput lifespan analysis
mother cells based on size or budding pattern. has the potential to bring invertebrate aging studies
Flowing media through the chamber replenishes nutri- to a new level of detail. On a first order, the expan-
ents while flushing away newly divided daughter sion of genome-scale longevity screens with relatively
cells. A dissection scope with an automated stage com- large population sizes and full survival curves will
bined with time-lapse microscopy is used to monitor allow a more complete picture of the range of genetic
cells and count divisions. Several studies demonstrate factors involved in aging and a more detailed under-
the capability of these systems to automatically mea- standing of their impact on survival. Expanding these
sure replicative lifespan and reproduce published screens to compare different genetic backgrounds and
results while simultaneously monitoring numerous environmental conditions combined with network
other phenotypes over the course of the replicative life analyses will progress our understanding of how
of each cell, including cell morphology, number of bud pathways involved in aging interact. The simulta-
scars, individual cell division times, and replicative neous measurement of healthspan markers (e.g.,
age-dependent changes in fluorescent marker expres- motility in worms) and fluorescent expression mar-
sion (Chen et al., 2017). As with the worm concepts, kers will provide additional detail about how age-
these microfluidics-based systems are scalable, but dependent changes in gene expression are influencing
require a dedicated dissection microscope and some lifespan and age-associated decline.
level of competency with microfluidics systems, which
currently limits widespread adoption. Microfluidic
solutions to high-throughput yeast replicative aging is
still in its infancy and technical challenges remain. One
Genome-scale mouse knockout collection
example is the dependence of these systems on size Inspired by the complete sequencing of the mouse
differentiation between mother and daughter cells, genome in 2002 and motivated by the potential power of
which is achieved by the precise geometry of the readily available knockout mouse strains for any desired
microstructures used to capture cells. Early in replica- gene as a genetic tool for studying virtually any aspect of
tive lifespan, optimizing the size of the structures is mammalian biology, the International Knockout Mouse
challenging because mother and daughter cells are Consortium (IKMC) was formed in 2007 and set out to
similar in size. Later in replicative lifespan, the generate single-gene knockout mouse lines in the C57BL/
increased size of the mother cell causes the cell surface 6 background for each of the B21,000 genes in the mouse
to press against the microstructures, which may have genome. The IKMC consisted of an international collabo-
unexpected effects on cellular aging. All systems to ration between four programs: The Knockout Mouse
date have focused on the use of haploid cells. Distinct Project (KOMP; www.komp.org), European Conditional
microstructures may be needed for yeast cells with dif- Mouse Mutagenesis Program (EUCOMM; www.eucomm.
ferent baseline size resulting from ploidy (e.g., diploid org), North American Conditional Mouse Mutagenesis
cells are larger than haploid cells), genetic manipula- Project (NorCOMM; www.norcomm.org), and Texas
tion, pharmacology, or the use of different yeast A&M Institute for Genomic Medicine (TIGM; www.tigm.
strains. Finally, the microfluidic parameters and pre- org). The first phase of the project, generating single-gene
treatment conditions used in these experiments—flow embryonic stem (ES) cell knockout lines for every gene in
speed, culture growth phase, sonication—have yet to the mouse genome, is now complete and both ES cell
be optimized. At present, these technical challenges lines and germ cells or live mice for recovered strains
appear surmountable and the coming years are likely mice are available to the broader research community for
to see many or all resolved. purchase. The major focus of this effort is now on the sec-
These recent platforms demonstrate that automated ond phase, a collaborative effort under the auspices of the
survival analysis is possible in both yeast and worms. IMPC (www.mousephenotype.org), which aims to use the
The systems are largely touted as moderate through- ES cell lines to generate and broadly phenotype knockout
put in their current configurations, primarily because mice for each gene. The IMPC consists of 17 research insti-
the platforms require a large amount of physical tutions and five national funding programs. The goal is to
space and the experimental setup still takes a fair extensively characterize each knockout mouse strain using
amount of manual labor to prepare plates and culture a standardized set of physiological, behavioral, molecular,
animals. The final survival analysis is also and biochemical assays and make the resulting dataset

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


26 1. Longevity as a complex genetic trait

publicly available, thus providing a genome-scale resource produce a series of inbred lines with a uniformly dis-
for comparative genetics in the mouse. tributed genetic contribution from the parental gen-
The prospect of determining lifespan for every omes (Collaborative Cross Consortium, 2012). Each CC
knockout mouse strain is both financially and logisti- line is developed independently, meaning that recom-
cally impractical and is not part of the IMPC pheno- bination events are not shared among strains, a prob-
typing pipeline (the standard pipeline includes a lem that has confounded association mapping studies
battery of measurements out to approximately 16 using other RI strain panels (Manenti et al., 2009). At
weeks of age). Regardless, the IKMC and IMPC efforts present, 69 CC lines are available with heterozygosity
will provide an invaluable resource to the aging ranging from 0.5%15% (average 8.04%) (Srivastava
research community. A broadly accessible, genome- et al., 2017; UNC Systems Genetics, 2019). When com-
wide knockout collection removes the first time- plete, the CC panel will include an estimated 100200
consuming hurdle to any knockout mouse aging study: inbred lines.
generating a knockout line. The availability of a large The DO population was seeded using 144 partially
array of standardized phenotype data will allow inbred CC lines (Chesler et al., 2008) and is maintained by
researchers to make preliminary decisions about which a random breeding strategy designed to minimize genetic
genes to pursue based on nonlongevity age-associated drift and allele frequency selection (Churchill et al., 2012;
traits, either starting from a preliminary set of candi- Rockman and Kruglyak, 2008). The DO mice are currently
date genes or in de novo analyses generated directly in the 35th generation (G35). Even at G16, each individual
from IMPC phenotype data. was estimated to have approximately 500 informative
recombination events per animal (Svenson et al., 2012)
providing a mapping resolution well below 1 Mb, an
order-of-magnitude improvement over classical genetic
Collaborative cross and diversity outbred mice crosses which typically produce resolution in the
The advents of relatively inexpensive genome 1050 Mb range. Mapping in the DO mice also has
sequencing and high-density marker arrays now allow advantages over using inbred strain panels or classical
human GWAS to map genetic loci at a hitherto unprec- crosses in both degree and distribution of genetic variation
edented precision. A critical characteristic of human (Rockman and Kruglyak, 2008). The DO random breeding
populations that allows for this high-resolution map- strategy also avoids problems with confounding linkage
ping is the high degree of genetic diversity among disequilibrium resulting from historical points of common
individuals. This diversity is balanced by a lack of con- ancestry common among strains in other RI panels
trol in other potential confounding areas such as envi- (Collaborative Cross Consortium, 2012; Petkov et al.,
ronment and population structure. Traditional model 2005). Each locus in a DO mouse has 36 possible diplo-
systems provide this control at the cost of genetic types resulting from the eight founder haplotypes. The
diversity. The collaborative cross (CC) and diversity Mouse Universal Genotyping Array (MUGA), a high-
outbred (DO) mouse populations are a recently devel- density SNP array containing 7854 pairs of allele-specific
oped pair of complementary tools designed to capture probes with an average spacing of 325 kb was used in
the advantages of both approaches—high genetic genotyping the initial generations of the DO mice in order
diversity to allow high-precision mapping while to capture the small haplotype blocks produced by
retaining control over environmental and population repeated outbreeding (Collaborative Cross Consortium,
factors—within a single model system. 2012; Wang et al., 2012). By G8, even MUGA was missing
The CC and DO mice were bred from the same eight an estimated 20% of recombination events. To address
inbred founder strains, including five classical strains: this problem MegaMUGA (containing 77,800 SNPs) and
A/J (AJ), C57BL/6J (B6), 129S1/SvImJ (129), NOD/ GigaMUGA (containing 143,000 SNPs) are now being
ShiLtJ (NOD), and NZO/HlLtJ (NZO); and three wild- employed to capture the increasingly fragmented diplo-
derived strains representing different Mus musculus sub- type structure in the latest DO generations (Morgan et al.,
species: CAST/EiJ (CAST), PWK/PhJ (PWK), and 2015). Another heterozygous stock derived from CC
WSB/EiJ (WSB) (Churchill et al., 2012; Svenson et al., strains (HS-CC) is available that uses circular mating to
2012). More than 36,000,000 SNPs are present in the control genetic drift at the cost of map expansion (Iancu
founder strains, with B75% of the genetic variation et al., 2010).
resulting from the inclusion of the three wild-derived The large number of recombination events per indi-
strains (Keane et al., 2011; Yalcin et al., 2011). vidual, the ability to identify the originating parental
The CC lines are generated using a two-phase fun- strain for each haplotype, and the wide phenotypic vari-
nel breeding strategy, starting with three rounds of ation make the DO mice a powerful platform for genetic
outbreed to combine the eight found genomes fol- mapping. The large number of possible diplotypes does
lowed by repeated sibling mating to eventually make calculating LOD scores for QTL analysis

I. Basic mechanisms, underlying physiological changes, model organisms and interventions


Another random document with
no related content on Scribd:
distributed Project Gutenberg™ eBooks with only a loose
network of volunteer support.

Project Gutenberg™ eBooks are often created from several


printed editions, all of which are confirmed as not protected by
copyright in the U.S. unless a copyright notice is included. Thus,
we do not necessarily keep eBooks in compliance with any
particular paper edition.

Most people start at our website which has the main PG search
facility: www.gutenberg.org.

This website includes information about Project Gutenberg™,


including how to make donations to the Project Gutenberg
Literary Archive Foundation, how to help produce our new
eBooks, and how to subscribe to our email newsletter to hear
about new eBooks.

You might also like