articulo-1-genÉtica (1).pdf

12
4 ILAR Journal Abstract Mice are an ideal mammalian model for studying the genet- ics of aging: considerable resources are available, the gen- eration time is short, and the environment can be easily controlled, an important consideration when performing mapping studies to identify genes that influence lifespan and age-related diseases. In this review we highlight some sa- lient contributions of the mouse in aging research: lifespan intervention studies in the Interventions Testing Program of the National Institute on Aging; identification of the genetic underpinnings of the effects of calorie restriction on lifespan; the Aging Phenome Project at the Jackson Laboratory, which has submitted multiple large, freely available phenotyping datasets to the Mouse Phenome Database; insights from spontaneous and engineered mouse mutants; and complex traits analyses identifying quantitative trait loci that affect lifespan. We also show that genomewide association peaks for lifespan in humans and lifespan quantitative loci for mice map to homologous locations in the genome. Thus, the vast bioinformatic and genetic resources of the mouse can be used to screen candidate genes identified in both mouse and human mapping studies, followed by functional testing, of- ten not possible in humans, to determine their influence on aging. Key Words: aging; calorie restriction (CR); gene mutation; genetics; lifespan; longevity; mouse genome; quantitative trait locus (QTL) Introduction M uch has been learned from the study of aging in worms and flies, but it is important to test the knowledge derived from these lower organisms in a mammalian species. For this, the mouse is ideal. Not only does it have a relatively short lifespan but, as a mamma- lian research model that shares 99% of its genes with hu- mans (Boguski 2002), outstanding genetic resources and Rong Yuan, Luanne L. Peters, and Beverly Paigen Rong Yuan, PhD, MD, is a research scientist and Animal Core Leader; Luanne L. Peters, PhD, is a professor and Director; and Beverly Paigen, PhD, is a professor and member of the Leadership Team, all at the Jackson Aging Center of the Jackson Laboratory in Bar Harbor, Maine. Address correspondence and reprint requests to Dr. Beverly Paigen, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 or email bev. [email protected]. Mice as a Mammalian Model for Research on the Genetics of Aging sophisticated genetic engineering technology are available for manipulating its genome (Paigen 1995). The many ge- netic resources of the mouse have been reviewed recently (Peters et al. 2007), and new resources, such as the Collab- orative Cross (Churchill et al. 2004; Threadgill et al. 2011), are being developed at a steady pace. Among the many aging studies that have used mouse models, we discuss testing of interventions (especially com- pounds that may extend lifespan) (Harrison et al. 2009; Miller et al. 2007; Strong et al. 2008), retardation of aging by calorie restriction, spontaneous or genetically engineered mutations that affect lifespan, the determination of lifespan in multiple inbred strains (Yuan et al. 2009), and quantitative trait locus (QTL 1 ) studies to find genomic regions associated with aging (de Haan et al. 1998; Gelman et al. 1988; Jackson et al. 2002; Klebanov et al. 2001; Lang et al. 2010; Miller et al. 1998, 2002a; Rikke et al. 2010; Yunis et al. 1984). Space limitations of this review prevent an in-depth discus- sion of the many aspects of aging; we refer the reader to re- cent outstanding reviews on calorie restriction (Fontana et al. 2010; Kemnitz 2011), the role of mitochondria (Larsson 2010) and telomeres in aging (Sahin and Depinho 2010), pathways known to affect aging (Kenyon 2010), and other mouse models of aging (Chen et al. 2010). As in any animal research, environmental and animal hus- bandry conditions may affect the outcome of aging studies. Lifespan may be affected by husbandry issues such as compo- sition of food, water, type of housing, density of mice/cage, enrichment, and animal room size and noise level, but very little is known about the impact of these factors on lifespan. Interventions Testing Program of the National Institute on Aging One practical use of the mouse is to test diets and compounds for their ability to slow aging and extend longevity in a mam- malian model. The Interventions Testing Program (ITP) of the National Institute on Aging is a three-site project with simultaneous identical lifespan studies at the Jackson Labo- ratory, University of Michigan, and University of Texas Health Science Center at San Antonio (Miller et al. 2007). 2 1 Abbreviations used in this article: Chr, chromosome; CR, calorie restriction; QTL, quantitative trait locus 2 Information is available at the ITP website (www.nia.nih.gov/research- information/scientificresources/interventionstestingprogram.htm); this and other websites cited in this article were accessed on December 22, 2010. at Universidad Autónoma de Guerrero on March 6, 2015 http://ilarjournal.oxfordjournals.org/ Downloaded from

Upload: blanca-leon

Post on 11-Nov-2015

241 views

Category:

Documents


0 download

TRANSCRIPT

  • 4 ILAR Journal

    Abstract

    Mice are an ideal mammalian model for studying the genet-ics of aging: considerable resources are available, the gen-eration time is short, and the environment can be easily controlled, an important consideration when performing mapping studies to identify genes that infl uence lifespan and age-related diseases. In this review we highlight some sa-lient contributions of the mouse in aging research: lifespan intervention studies in the Interventions Testing Program of the National Institute on Aging; identifi cation of the genetic underpinnings of the effects of calorie restriction on lifespan; the Aging Phenome Project at the Jackson Laboratory, which has submitted multiple large, freely available phenotyping datasets to the Mouse Phenome Database; insights from spontaneous and engineered mouse mutants; and complex traits analyses identifying quantitative trait loci that affect lifespan. We also show that genomewide association peaks for lifespan in humans and lifespan quantitative loci for mice map to homologous locations in the genome. Thus, the vast bioinformatic and genetic resources of the mouse can be used to screen candidate genes identifi ed in both mouse and human mapping studies, followed by functional testing, of-ten not possible in humans, to determine their infl uence on aging.

    Key Words: aging; calorie restriction (CR); gene mutation; genetics; lifespan; longevity; mouse genome; quantitative trait locus (QTL)

    Introduction

    M uch has been learned from the study of aging in worms and fl ies, but it is important to test the knowledge derived from these lower organisms in a mammalian species. For this, the mouse is ideal. Not only does it have a relatively short lifespan but, as a mamma-lian research model that shares 99% of its genes with hu-mans (Boguski 2002), outstanding genetic resources and

    Rong Yuan, Luanne L. Peters, and Beverly Paigen

    Rong Yuan, PhD, MD, is a research scientist and Animal Core Leader; Luanne L. Peters, PhD, is a professor and Director; and Beverly Paigen, PhD, is a professor and member of the Leadership Team, all at the Jackson Aging Center of the Jackson Laboratory in Bar Harbor, Maine.

    Address correspondence and reprint requests to Dr. Beverly Paigen, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 or email [email protected].

    Mice as a Mammalian Model for Research on the Genetics of Aging

    sophisticated genetic engineering technology are available for manipulating its genome (Paigen 1995). The many ge-netic resources of the mouse have been reviewed recently (Peters et al. 2007), and new resources, such as the Collab-orative Cross (Churchill et al. 2004; Threadgill et al. 2011), are being developed at a steady pace.

    Among the many aging studies that have used mouse models, we discuss testing of interventions (especially com-pounds that may extend lifespan) (Harrison et al. 2009; Miller et al. 2007; Strong et al. 2008), retardation of aging by calorie restriction, spontaneous or genetically engineered mutations that affect lifespan, the determination of lifespan in multiple inbred strains (Yuan et al. 2009), and quantitative trait locus (QTL1) studies to fi nd genomic regions associated with aging (de Haan et al. 1998; Gelman et al. 1988; Jackson et al. 2002; Klebanov et al. 2001; Lang et al. 2010; Miller et al. 1998, 2002a; Rikke et al. 2010; Yunis et al. 1984). Space limitations of this review prevent an in-depth discus-sion of the many aspects of aging; we refer the reader to re-cent outstanding reviews on calorie restriction (Fontana et al. 2010; Kemnitz 2011), the role of mitochondria (Larsson 2010) and telomeres in aging (Sahin and Depinho 2010), pathways known to affect aging (Kenyon 2010), and other mouse models of aging (Chen et al. 2010).

    As in any animal research, environmental and animal hus-bandry conditions may affect the outcome of aging studies. Lifespan may be affected by husbandry issues such as compo-sition of food, water, type of housing, density of mice/cage, enrichment, and animal room size and noise level, but very little is known about the impact of these factors on lifespan.

    Interventions Testing Program of the National Institute on Aging

    One practical use of the mouse is to test diets and compounds for their ability to slow aging and extend longevity in a mam-malian model. The Interventions Testing Program (ITP) of the National Institute on Aging is a three-site project with simultaneous identical lifespan studies at the Jackson Labo-ratory, University of Michigan, and University of Texas Health Science Center at San Antonio (Miller et al. 2007).2

    1Abbreviations used in this article: Chr, chromosome; CR, calorie restriction; QTL, quantitative trait locus2Information is available at the ITP website (www.nia.nih.gov/research-information/scientifi cresources/interventionstestingprogram.htm); this and other websites cited in this article were accessed on December 22, 2010.

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • Volume 52, Number 1 2011 5

    Table 1 Lifespan characteristics of 32 inbred mouse strainsa

    Strain

    Female Male

    Age (in days) of20% longest-lived (mean SEM)

    Age (in days) of20% longest-lived (mean SEM)b

    25% death

    50% death

    75% death

    25% death

    50% death

    75% death

    AKR/J 224 254 308 395 24 244 288 336 415 18PL/J 373 471 596 736 17 365 469 558 674 19SJL/J 393 515 632 740 30 330 505 555 632 21MRL/MpJ 455 555 626 681 9 549 645 669 711 10NZO/H1LtJ 418 575 700 782 18 286 423 637 762 26CAST/EiJ 219 589 754 n.a. 239 591 754 n.a.KK/H1J 564 608 653 720 13 545 616 700 826 43BTBR T+tf/J 550 611 668 743 19 444 575 728 822 20BUB/BnJ 392 621 755 876 23 354 493 873 906 23SWR/J 499 630 814 n.a. 411 726 904 1020 29CBA/J 476 637 786 855 11 532 679 808 872 10A/J 505 639 739 806 19 541 623 708 785 18P/J 546 660 791 n.a. 439 607 673 n.a.NOD.B10-H2b 599 667 770 827 13 501 696 878 954 11C3H/HeJ 532 683 797 833 7 623 728 834 894 15DBA/2J 443 687 823 872 7 410 701 759 825 17MOLF/EiJ 590 705 n.a.c n.a. 503 686 730 n.a.C57L/J 700 721 749 800 5 658 736 768 806 9NZW/LacJ 600 732 866 950 16 607 792 1013 1126 14SM/J 650 733 817 902 15 730 783 833 873 6FVB/NJ 518 760 952 1023 13 553 591 708 879 56129S1/SvImJ 651 791 920 1012 25 798 882 992 1044 12BALB/cByJ 700 795 877 936 10 512 714 840 927 13NON/ShiLtJ 631 806 861 887 5 793 847 919 958 11RIIIS/J 691 813 883 938 5 779 886 940 970 12LP/J 715 833 966 1047 17 721 822 862 984 28PWD/PhJ 600 839 929 993 12 575 813 905 956 12C57BR/CDJ 757 861 917 973 7 737 849 943 993 21C57BLKS/J 672 867 926 989 12 770 826 932 983 21WSB/EiJ 629 886 1148 n.a. 470 1005 1110 1213 19C57BL/10J 692 889 1035 1135 9 677 792 852 893 13C57BL/6J 782 914 1006 1075 13 838 901 971 1061 17

    n.a., not available; SEM, standard error of the meanaLifespan traits reported by Yuan and colleagues (2009) and updated in August 2009. Age of 25%, 50%, and 75% at death and mean lifespan of the 20% longest-lived mice were calculated using JMP 6.0.4 software. bMean lifespan of the 20% longest-lived mice is not available for strains for which mice are still alive.cAge at death of 75% MOLF/EiJ was not available because there were too few mice to evaluate.

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • 6 ILAR Journal

    Tabl

    e 2

    Mut

    atio

    ns in

    mou

    se g

    enes

    that

    incr

    ease

    long

    evity

    Gen

    e in

    form

    atio

    nTy

    pe o

    f m

    uta

    tion

    Targ

    et g

    ene

    expr

    essi

    onEf

    fect

    on

    lifes

    pan

    (sex)

    Ref

    eren

    ceSy

    mbo

    laFu

    ll na

    me

    Chr

    Mb

    Adcy

    5Ad

    enyla

    te c

    ycla

    se 5

    1635

    Knoc

    kout

    Red

    uces

    Incr

    ease

    s (po

    oled)

    Yan

    et a

    l. 20

    07Ce

    bpb

    CCAA

    T/en

    hanc

    er b

    indi

    ng

    pr

    otei

    n (C

    /EBP

    ), beta

    2

    168

    Knoc

    k-in

    Incr

    ease

    sIn

    crea

    ses

    (fem

    ale

    , m

    ale

    )Ch

    iu e

    t al. 2

    004

    Coq7

    Dem

    ethy

    l-Q 7

    512

    6Kn

    ocko

    ut

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le, m

    ale

    )Li

    u et

    al.

    2005

    Ghr

    Gro

    wth

    hor

    mone r

    ece

    ptor

    153

    Knoc

    kout

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le, m

    ale

    )Co

    schi

    gano

    et a

    l. 200

    3G

    hrhr

    Gro

    wth

    hor

    monere

    leas

    ing

    ho

    rmone r

    ece

    ptor

    655

    Spon

    tane

    ous

    Red

    uces

    Incr

    ease

    s (po

    oled)

    Flur

    key

    et a

    l. 200

    1

    Igf1

    bIn

    sulin

    -like

    gro

    wth

    fact

    or 1

    1088

    Transg

    ene

    Incr

    ease

    sIn

    crea

    ses

    (male

    )Li

    and

    Ren

    200

    7Ig

    f1r

    Insu

    lin-li

    ke g

    row

    th fa

    ctor

    I re

    cept

    or7

    75Kn

    ocko

    ut

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le)

    Hol

    zen

    berg

    er e

    t al. 2

    003

    Insr

    Insu

    lin re

    cept

    or8

    3Kn

    ocko

    ut

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le, m

    ale

    )Bl

    uher

    et a

    l. 20

    03Irs

    1In

    sulin

    rece

    ptor

    sub

    stra

    te 1

    182

    Knoc

    kout

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le)

    Selm

    an e

    t al. 2

    008a

    Irs2c

    Insu

    lin re

    cept

    or s

    ubst

    rate

    28

    11Kn

    ocko

    ut

    Red

    uces

    Incr

    ease

    s (po

    oled)

    Tagu

    chi e

    t al. 2

    007

    Kld

    Klot

    ho5

    152

    Transg

    ene

    Incr

    ease

    sIn

    crea

    ses

    (fem

    ale

    , m

    ale

    )Ku

    rosu

    et a

    l. 20

    05M

    catb

    Mal

    onyl

    CoA:

    ACP

    acylt

    ransf

    era

    se

    (m

    itoch

    ondri

    al)

    1583

    Transg

    ene

    Incr

    ease

    sIn

    crea

    ses

    (fem

    ale

    , m

    ale

    ) Sc

    hrin

    er e

    t al.

    2005

    Mtb

    Met

    allo

    thio

    nein

    897

    Transg

    ene

    Incr

    ease

    sIn

    crea

    ses

    (male

    )Ya

    ng

    et a

    l. 200

    6Pa

    ppa

    Preg

    nanc

    y-as

    socia

    ted

    plas

    ma

    pr

    otei

    n A

    465

    Knoc

    kout

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le, m

    ale

    )Co

    nove

    r a

    nd

    Bale

    200

    7

    Pck1

    Phos

    phoe

    nolp

    yruva

    te

    ca

    rbox

    ykin

    ase

    1, c

    ytos

    olic

    210

    3Tr

    ansg

    ene

    Incr

    ease

    sIn

    crea

    ses

    (fem

    ale

    , m

    ale

    )H

    akim

    i et a

    l. 20

    07

    Pou1

    f1PO

    U do

    mai

    n, c

    lass

    1, t

    ransc

    riptio

    n

    fact

    or 1

    1666

    Spon

    tane

    ous

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le, m

    ale

    )Fl

    urke

    y et

    al. 2

    002

    Ppar

    gPe

    roxi

    som

    e pr

    olife

    rato

    rac

    tivate

    d

    rece

    ptor

    gam

    ma

    611

    5Kn

    ock-

    inIn

    crea

    ses

    Incr

    ease

    s (m

    ale)

    Hei

    kkin

    en e

    t al.

    2009

    Prop

    1Pa

    ired-

    like h

    omeo

    dom

    ain

    tra

    nsc

    riptio

    n fa

    ctor

    111

    51Sp

    onta

    neou

    sR

    educ

    esIn

    crea

    ses

    (fem

    ale

    , m

    ale

    )Br

    own

    -Bor

    g et

    al. 1

    996

    Rps

    6kb1

    Rib

    osom

    al p

    rote

    in S

    6 kin

    ase,

    po

    lypep

    tide

    111

    86Kn

    ocko

    ut

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le)

    Selm

    an e

    t al. 2

    009

    Shc1

    Src

    hom

    olog

    y 2

    dom

    ain

    cont

    aini

    ng

    tra

    nsf

    orm

    ing

    prot

    ein

    C13

    89Kn

    ocko

    ut

    Red

    uces

    Incr

    ease

    s (no

    t spe

    cifi e

    d)M

    iglia

    ccio

    et a

    l. 199

    9

    Surf1

    Surfe

    it ge

    ne 1

    227

    Knoc

    kout

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le, m

    ale

    )D

    ella

    gnel

    lo e

    t al. 2

    007

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • Volume 52, Number 1 2011 7

    The diets and compounds tested are selected from proposals by the extramural research community (Nadon et al. 2008). The ITP mice are generated by breeding two hybrids, (BALB/cByJ C57BL/6J) F1 (C3H/HeJ DBA/2J) F1, so that all mice are genetically heterogeneous but the genetic variation of the population is reproducible. The use of these mice avoids genotype-specifi c effects on disease susceptibil-ity while ensuring the replicability of the study.

    Although all three sites follow the same standardized pro-tocols, both control and drug-exposed mice at the University of Michigan site were signifi cantly smaller throughout adult life than those at the other two sites, and researchers observed signifi cant differences in survival of male (but not female) mice in the control groups (Harrison et al. 2009; Strong et al. 2008). The researchers hypothesized that such differences could be due to the sources and formulations of food. At the start of the program, the diets used for breeders and wean-lings (before drug exposure) differed in fat content (4.5-6.5%), supplemental levels of thiamine and other heat-sensitive vitamins, and protein source and content (18-24%). Starting with Cohort 4 (born in 2007), however, the three ITP sites adopted a uniform protocol for diet composition at all stages of the test process, including diets for breeder mice and for test mice before drug administration. It is also possible that other site-specifi c factors, such as minor differences in water quality, noise level, ventilation, extraneous odors, or cage-changing frequency contribute to site-specifi c differences.

    The ITP website provides the list of compounds in test-ing. So far, one of the major fi ndings of the study is that ra-pamycin, an inhibitor of mTOR (mammalian target of rapamycin) signaling, signifi cantly increased lifespan in both males and females even though treatment did not start until mice were 600 days old (Harrison et al. 2009). How-ever, the rapamycin-treated mice did not differ from control mice in the pattern of diseases as shown by pathology. Two other compoundsNDGA (p = 0.0006) and aspirin (p = 0.01), as assessed using the log rank test, which evalu-ates survivorship of the entire cohortextended the median lifespan in male mice but not maximum lifespan as shown by comparisons of the proportion of mice alive at the age of 90% mortality (Strong et al. 2008). This suggests that the drugs may delay the onset or reduce the severity of specifi c diseases but that they do not affect the rate of aging.

    Calorie Restriction

    One of the interventions most reliably associated with an extension of lifespan and a reduced rate of aging is calorie restriction (CR1), the reduction of food intake without malnutrition. CR has been shown to extend the lifespan of yeast, fl ies, worms, fi sh, rodents, and rhesus monkeys (Fontana et al. 2010) and, in mammals, decrease the risk of age-related diseases such as diabetes, cardiovascular diseases, and can-cers (Fontana and Klein 2007; Morley et al. 2010).

    Mouse models have been used extensively to investigate the underlying mechanisms of the antiaging effects of CR. G

    ene

    info

    rmat

    ion

    Type

    of

    mu

    tatio

    nTa

    rget

    gen

    eex

    pres

    sion

    Effe

    ct o

    n lif

    espa

    n (se

    x)

    Ref

    eren

    ceSy

    mbo

    laFu

    ll na

    me

    Chr

    Mb

    Ucp2

    eUn

    coup

    ling

    prot

    ein

    27

    108

    Transg

    ene

    Incr

    ease

    sIn

    crea

    ses

    (fem

    ale

    , m

    ale

    )Co

    nti e

    t al. 2

    006

    Mif

    Mac

    roph

    age

    mig

    ratio

    n in

    hibi

    tory

    fact

    or

    1075

    Knoc

    kout

    Red

    uces

    Incr

    ease

    s (fe

    ma

    le)

    Har

    per e

    t al. 2

    010

    Chr,

    chro

    mos

    ome;

    M

    b, m

    ega

    base

    (millio

    ns of

    base

    pairs

    )aG

    ene

    nam

    es a

    nd s

    ymbo

    ls ar

    e ac

    cord

    ing

    to th

    e M

    ouse

    Gen

    ome

    Info

    rmatic

    s da

    taba

    se (w

    ww.in

    form

    atic

    s.jax

    .org).

    b Mod

    els

    were

    gen

    erate

    d by

    transf

    err

    ing

    the

    hum

    an g

    ene.

    c Irs2

    kn

    ocko

    ut h

    eter

    ozyg

    otes

    sho

    wed

    an e

    xten

    ded

    lifesp

    an in

    Tagu

    chis

    stu

    dy (T

    agu

    chi a

    nd W

    hite

    200

    8) bu

    t faile

    d to

    ext

    end

    lifesp

    an in

    a s

    tudy

    by

    Selm

    an a

    nd c

    olle

    ague

    s (20

    08a).

    Th

    e au

    thor

    s of

    th

    e tw

    o s

    tudi

    es d

    iscus

    s po

    ssib

    le re

    ason

    s fo

    r th

    e di

    ffere

    nt r

    esul

    ts: d

    iffere

    nce

    s in

    the

    lifesp

    an o

    f con

    trols,

    num

    ber o

    f tim

    es th

    e k

    nock

    out w

    as

    back

    cros

    sed

    to C

    56BL

    /6, d

    iet,

    and

    hous

    ing

    cond

    itions

    .

    d Kuro

    sus

    mode

    l is a

    transg

    enic

    mod

    el th

    at o

    vere

    xpre

    sses

    Klo

    tho.

    eTh

    e tra

    nsg

    enic

    mod

    el o

    vere

    xpre

    sses

    Ucp

    2 in

    hyp

    ocer

    tin n

    euro

    ns, w

    hich

    cau

    ses

    elev

    ate

    d te

    mpe

    ratu

    re in

    the

    ther

    most

    at c

    ente

    r and

    resu

    lts in

    a lo

    werin

    g of

    cor

    e bo

    dy te

    mpe

    ratu

    re.

    Tabl

    e 2

    (cont

    inued

    )

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • 8 ILAR Journal

    One of the most interesting recent studies was an investiga-tion of the effects of CR in different genetic backgrounds. A set of 42 recombinant inbred strains of mice, generated by crossing strains ILS and ISS, was examined for lifespan under ad libitum (AL) or CR conditions (Liao et al. 2010; Rikke et al. 2010). Although CR signifi cantly extended fe-male lifespan in nine strains, it signifi cantly reduced lifespan in four other strains and had no signifi cant effect in 29 strains, suggesting that genetic background affects the ability of CR to alter aging. This gene-environment interaction is not surprising, nor does the fi nding that CR acts only in certain genetic backgrounds contradict the widespread observation that CR usually extends lifespan in species with mixed ge-netic background. The mean lifespan under CR showed no signifi cant correlation to lifespan under AL, suggesting that different genes modulate lifespan under each experimental condition. The study by Rikke and colleagues (2010) also found that increased effi ciency of food utilization correlated with longer lifespan (R = 0.34, p = 0.026) as measured by the ability to maintain body weight, hair growth, and tail growth during CR.

    The Aging Phenome Project

    The Aging Center at the Jackson Laboratory characterized the lifespan and aging-related phenotypes of 32 inbred mouse strains, providing a baseline for further use of mouse models to improve understanding of the genetic regulation of aging. The project included both longitudinal and cross-sectional studies. The former not only assessed lifespan (using 96 mice per strain) but also carried out noninvasive clinical assessments of neuromuscular function at 6, 12, 18, and 24 months (Wooley et al. 2009), kidney and heart func-tion (Tsaih et al. 2009; Xing et al. 2009), hematology, hor-mone levels, and immune system parameters (Petkova et al. 2008). The cross-sectional study euthanized 30 mice of each strain at 6, 12, and 20 months for body composition, bone density, necropsy, and pathology (Sundberg et al. 2008) and for the collection of tissues to evaluate apoptosis, DNA re-pair, and chromosome fragility. A reproductive study evalu-ated the age of sexual maturity in females of the same 32 strains (Yuan et al. manuscript in preparation). In addition to individual reports, all of these data are available in the Mouse Phenome Database (MPD; http://phenome.jax.org), where the Aging Center submits all data after quality control, even before publication. The MPD also provides statistical tools to enable the assessment of correlations of lifespan with other parameters in this and other studies (Grubb et al. 2009).

    Among the 32 strains, four were recently derived from the wild and represent the major subspecies of laboratory mice: WSB/EiJ for Mus domesticus, PWD/PhJ for M. mus-culus, CAST/EiJ for M. castaneus, and MOLF/RkJ for M. molossinus. The remaining 28 strains were chosen for ge-netic diversity and common use. Median lifespan varied dra-matically among the inbred strains (Table 1); the shortest

    was that of AKR/J (251 and 288 days for female and male, respectively), and the longest, female WSB/EiJ (964 days) and male C57BL/6J (901 days). These results confi rmed that genetics plays an important role in determining longevity. Median lifespans for females and males were signifi cantly correlated with each other (R = 0.88; p < 0.001). Propor-tional hazard analysis showed that sex did not signifi cantly affect lifespan for most strains (Yuan et al. 2009).

    Among the 32 inbred strains, circulating insulinlike growth factor (IGF)-1 levels signifi cantly (p < 0.05) corre-lated with body weight at 6, 12, and 18 months in both fe-males and males (data available in the MPD): lower levels were associated with lighter body weight, which in turn was associated with extended longevity in a heterogeneous mouse population (Miller et al. 2002c). Our analysis found that IGF-1 levels at 6 months negatively correlated with me-dian lifespan (R = 0.33, p = 0.01) (Yuan et al. 2009). After excluding the six short-lived strains (median lifespan less than 600 days), which presumably died of a particular strain-specifi c disease (e.g., leukemia in strain AKR), the negative correlation of IGF-1 and lifespan among long-lived strains became stronger and more signifi cant (R = 0.53, p < 0.01).

    These results underscore the importance of genetic regu-lation of IGF-1 signaling in regulating body weight and lon-gevity, as has been suggested by studies in other models. For example, in domesticated dogs a single nucleotide poly-morphism (SNP) in Igf1 signifi cantly correlated with body weight (Sutter et al. 2007); in human populations, genetic polymorphisms of IGF-1 receptor (IGF-1R) (Suh et al. 2008) and phosphatidylinositol 3-kinase catalytic beta polypeptide (PIK3CB) (Bonafe et al. 2003) associated with human lon-gevity. The variation in circulating IGF-1 levels among in-bred strains of mice and the correlation of these levels with longevity suggest that they may be a useful focus in research on the genetic regulation of longevity.

    Genes Implicated in Aging

    Single-gene mutations that affect lifespan provide valuable tools for exploring the molecular basis for aging mecha-nisms. A number of mutations, either spontaneous or ge-netically engineered, that affect lifespan in the mouse are known; these are summarized in Table 2 and their location on the genome shown in Figure 1. The fi rst of these mutants were spontaneous dwarf mice (e.g., the Ames dwarf, the Snell dwarf) and the little mouse, which have defects in the growth hormone (GH)/IGF/insulin signaling pathway (Brown-Borg et al. 1996; Flurkey et al. 2001, 2002). The little mouse has a defect in the gene Ghrhr (growth hor-monereleasing hormone receptor), and the Ames and Snell dwarf mice in the genes Prop1 (paired-like homeodomain transcription factor 1) and Pou1f1 (POU domain, class 1, transcription factor 1), respectively. These three mutations result in abnormal development of the anterior pituitary gland and corresponding defi ciency of pituitary hormones such as growth hormone, thyroid-stimulating hormone, and

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • Volume 52, Number 1 2011 9

    Figu

    re 1

    Qua

    ntitat

    ive tr

    ait l

    oci (Q

    TLs)

    for m

    ouse

    longe

    vity

    and

    geno

    mew

    ide a

    ssoc

    iatio

    n (G

    WA) p

    eaks

    for h

    uman

    long

    evity

    , b

    oth

    depi

    cted

    on

    the m

    ouse

    gen

    ome (

    mapp

    ed in

    Mb).

    The l

    engt

    h of t

    he c

    olor

    ed b

    ars r

    epre

    sent

    s the

    95%

    con

    fi den

    ce in

    terv

    al if

    repo

    rted

    or a

    n es

    timat

    ed 4

    0 M

    b if

    not r

    epor

    ted;

    the

    blac

    k ba

    rs a

    cros

    s the

    col

    ored

    bar

    s rep

    rese

    nt Q

    TL pe

    aks.

    We

    dete

    rmin

    ed th

    e M

    b po

    sitio

    n us

    ing

    a rec

    ently

    revise

    d m

    ouse

    map

    (Cox

    et al

    . 200

    9) an

    d the

    Mou

    se M

    ap C

    onver

    ter f

    rom

    the C

    ente

    r for

    Gen

    ome D

    ynam

    ics

    (http:

    //cgd

    .jax.o

    rg/m

    ouse

    map

    conv

    erte

    r/). A

    rrow

    s on

    the

    left

    of c

    hrom

    osom

    es re

    pres

    ent h

    uman

    GW

    A p

    eaks

    at t

    he h

    omol

    ogou

    s mou

    se g

    enom

    e lo

    catio

    ns. C

    hr, ch

    rom

    osom

    e, M

    b, m

    egab

    ase

    (milli

    ons o

    f bas

    e pa

    irs).

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • 10 ILAR Journal

    Table 3 Mutations in mouse genes that reduce longevity

    Gene informationType of mutation

    Target geneexpression

    Effect on lifespan (sex) ReferenceSymbola Full name Chr Mb

    Bub1b Budding uninhibited by benzimidazoles 1 homolog, beta

    2 118 Knockout Reduces Reduces (female, male)

    Baker et al. 2004

    Kl Klotho 5 152 Transgeneb Reduces Reduces (female, male)

    Kuro-o et al. 1997

    Lmna Lamin A 3 88 Knock-in n.a.c Reduces (not specifi ed)

    Mounkes et al. 2003

    Msra Methionine sulfoxide reductase A

    14 65 Knockout Reduces Reduces (female, male)

    Moskovitz et al. 2001

    PolgA Polymerase (DNA directed), gamma

    7 87 Knock-in Reduces Reduces (pooled) Trifunovic et al. 2004

    Prdx1 Peroxiredoxin 1 4 116 Knockout Reduces Reduces (not specifi ed)

    Neumann et al. 2003

    Top3b Topoisomerase (DNA) III beta

    16 17 Knockout Reduces Reduces (not specifi ed)

    Kwan and Wang 2001

    Chr, chromosome; Mb, megabase (millions of base pairs); n.a., not availableaGene names and symbols are according to Mouse Genome Informatics database (www.informatics.jax.org). bThe transgene causes an insertional mutation in the Klotho gene that suppresses its expression.cThis knock-in model introduces a nucleotide polymorphism that results in the substitution of proline for leucine at amino acid 530 in the Lmna gene.

    prolactin. These dwarf mutants all have extended lifespan compared to controls.

    Mutations in several other genes (Ghr, Igf1r, Insr, Irs1, Irs2 and Pappa) reduce GH/IGF/insulin signaling and ex-tend lifespan. Cardiac-specifi c overexpression of IGF-1 sig-nifi cantly prolongs lifespan, probably due to the protective effects of IGF-1 on cardiac failure. Mutations such as knock-outs of Shc1, Surf1, Adcy5, and Coq7, as well as transgenes of Mcat and Mt, which increase resistance to stress, also suc-cessfully extend longevity. Knock-in/transgenic models that increase the expression of Pparg, Cebpb, Pck1, and Ucp2 have shown increased lifespan by regulating metabolism and energy expenditure (Table 2).

    Mutations that extend lifespan are likely to affect the rate of aging, while those that reduce lifespan either alter aging or increase the risk or severity of a particular disease. Ac-cording to Mouse Genome Informatics (www.informatics.jax.org), 301 mutations decrease survival by causing or pro-moting susceptibility to disease and 46 promote features of premature aging. In Table 3 we list genes whose mutations decrease longevity and appear to alter aging. The roles of these genes, similar to the mutations that extend longevity, suggest that maintaining DNA stability and antioxidative stress are important molecular mechanisms that regulate ag-ing and longevity. For example, a knockout of Bub1b induces chromosome (Chr1) instability, reduced expression of PolgA increases mutations in mitochondrial DNA, and knockouts of Msra and Prdx1 increase oxidative stress.

    Aging studies in mutant gene models also provide clues for understanding the molecular mechanisms that extend lifespan by CR. For example, mice heterozygous for a Foxo1 (forkhead box O1) knockout did not differ signifi cantly in lifespan compared to wild-type controls under AL or CR conditions. However, Foxo1 may play a role in CRs antineo-plastic effect, which, as indicated by reduced incidence of tumors at death in the diet-restricted wild-type mice, was mostly abrogated in the heterozygous knockout mice (Yamaza et al. 2010). The noticeable increase of MIF (mac-rophage migration inhibitory factor) in CR mice suggests that it may be important for CR-related lifespan exten-sion, but the signifi cantly extended longevity in Mif knock-out mice challenges this hypothesis (Harper et al. 2010). Interestingly, deletion of S6k1 not only extended longevity but also induced gene expression patterns similar to those seen in CR or with pharmacological activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK), a conserved regulator of the metabolic response to CR (Selman et al. 2009). This suggests that therapeutic manipu-lation of S6K1 and AMPK might mimic CR and could pro-vide broad protection against diseases of aging.

    One problem with a lifespan extension study is that altering the risk of a disease may change the mean or median lifespan but not reduce the rate of aging. One method to distinguish be-tween these outcomes is to calculate the age-specifi c mortality rate (de Magalhaes et al. 2005). For example, CR changes age-specifi c mortality and delays aging, as do mutations of Cebpb,

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • Volume 52, Number 1 2011 11

    Msra, Shc1, Ghr, Pou1f1, and Polg, but studies in other mutants were either insuffi ciently powered for such calculations or changed disease risk without changing the rate of aging.

    Lifespan Studies

    QTLs in Mice

    Examining spontaneous or genetically engineered mutants to determine a genes effect on lifespan is one way to unravel the genetic basis of aging. Another approach, which is unbi-ased and does not start with a defi ned hypothesis, is to con-duct a quantitative trait locus study to determine the genomic locations of genes that affect lifespan. Although all the QTL studies performed so far on aging in the mouse were under-poweredin the number of animals or markers genotyped or bothwe think these QTLs are worthy of further investi-gation, especially if they have been replicated in another mouse cross or if a human genomewide association study has identifi ed a peak at a homologous location. Thus, we list all the suggestive and signifi cant QTLs in Table 4 and depict them on the mouse genome in Figure 1.

    The earliest study was a (C57BL/6J DBA/2) C57BL/6 backcross using only four markers: two coat color genes on Chrs 4 and 9, the H2 antigen on Chr 17, and sex (Yunis et al. 1984). Subsequent studies tested 20 of the BXD (C57BL/6J DBA/2J) recombinant inbred (RI) lines for lifespan (de Haan et al. 1998; Gelman et al. 1988), using, as markers, 101 genes that are distinguishable between B6 and D2, but these mark-ers were not evenly distributed and only 14 chromosomes were covered. The QTL on Chr 17 identifi ed in these two studies contains the major histocompatibility complex re-gion, and thus may be related to the infection that occurred in the colony before the end of the study. A recent study of longevity using BXD RI strains, a more sophisticated lifespan analysis, and 671 markers failed to replicate the Chr 17 QTL (Lang et al. 2010). No infection occurred in the colony during this second study, which is, to date, the QTL lifespan study with the greatest statistical power (Lang et al. 2010) and will prove to be very useful, as considerable infrastructure resources (e.g., genotyping, sequence, and expression data) are available for these RI lines at GeneNetwork (www.genenetwork.org) and will enable the application of bioinformatics and system ge-netics approaches to the study of aging.

    Both the backcross and RI QTL designs carry homozy-gous alleles that may cause deleterious effects on lifespan without affecting aging. To minimize such effects, researchers conducted three different QTL studies using a four-way cross population. The fi rst, using a (BALB/cJ C57BL/6) (C3H/HeJ DBA/2J) cross, showed that different loci were involved in regulating the lifespans of female and male mice (Jackson et al. 2002). In a post hoc study of the same population, Miller and colleagues (1998, 2002a) found that the genotype associ-ated with increased survival in mice dying of cancer also correlated with a similar degree of lifespan extension in mice dying of other causes, suggesting that many forms of late-life

    disease may be infl uenced by shared pathophysiologic mech-anisms that are under coordinated genetic control.

    Miller and colleagues (2002b) suggested that wild mice or inbred strains recently derived from the wild may carry alleles that delay sexual maturation and aging and that are missing in domesticated inbred strains. Thus, two additional four-way cross QTL studies each included one wild-derived inbred strain, MOLD or CAST (LP/J MOLD/Rk) (NZW/LacJ BALB/cJ) and (ST/bJ C57BL/6J) (CAST/EiJ DBA/2J) (Klebanov et al. 2001). These crosses revealed the alleles of wild-derived inbred strains that confer extended longevity on Chr 8 and Chr 10 (Klebanov et al. 2001).

    Although we have included all the suggestive and sig-nifi cant QTLs for lifespan in Figure 1 and Table 4, we have more confi dence that replicated QTLs are true positives. Eight of these QTLsChr 1, Chr 2, Chr 7 (proximal and mid-), Chr 8, Chr 10 distal, Chr 11 proximal, and Chr 19have been replicated in another mouse cross (Table 4). We have counted as replicated those whose QTL peaks are within 10 Mb of each other, but further investigation may reveal that some of these are independent QTLs.

    Concordance of Human and Mouse Lifespan Peaks

    A recent genomewide association study of longevity, a meta-analysis of four separate studies by the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) consortium, compared 1900 human subjects that lived to age 90 with an equal number of controls that died earlier (Newman et al. 2010). Although none of the peaks reached statistical signifi cance, we have included the 10 highest peaks on the mouse map (arrows in Figure 1). Remarkably, eight of the 10 are located in a mouse QTL; the probability that this is due to chance is very low (p = 0.0025 using Fishers exact test, based on lifespan QTLs covering 860 Mb of the 2700 Mb genome and each human peak being 1 Mb in size). Five of these human peaks (Chrs 1, 9, 10, 11, 16) are located within 10 Mb of a mouse QTL peak. Concordance of human and mouse QTLs has been reported previously (Garrett et al. 2010; Sugiyama et al. 2001; Wang and Paigen 2005), but for traits such as plasma lipids, hypertension, and kidney dis-ease. Lifespan as a trait would be highly infl uenced by chance and by environmental factors, so one might think that concordance would be reduced or perhaps even nonexistent. Yet Figure 1 clearly shows concordance between humans and mice for lifespan, suggesting that the data for both spe-cies can be integrated and that the bioinformatic and genetic resources of the mouse can be used to narrow the QTL and test candidate genes.

    Future Directions

    Mouse models are valuable for studies of the genetics of hu-man aging not only because of the availability of extensive mouse resources but also because of the similarity of the

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • 12 ILAR Journal

    Table 4 Signifi cant and suggestive lifespan quantitative trait loci (QTLs) detected in the mouse

    ChraPeak (Mb) Cross

    High allele strain (sex) Reference

    Replicated in mice

    Replicated in humans

    1 34 B6 D2 RI strains D2 (male) Lang et al. 2010120 B6 D2 RI strains D2 (female) Gelman et al. 1988 X128 B6 D2 RI strains D2 (female) Lang et al. 2010 X163 B6 D2 RI strains B6 (female) Gelman et al. 1988 X

    2 65 B6 D2 RI strains B6 (female) Lang et al. 2010103 B6 D2 RI strains B6 (female) Gelman et al. 1988 X108 (BALB/cJ B6)

    (C3H D2)C3H (female) Jackson et al. 2002;

    Miller et al. 2002aX

    121 B6 D2 RI strains D2 (female) Gelman et al. 19884 80 (B6 D2) D2 B6 (female) Yunis et al. 19845 80 B6 D2 RI strains D2 (female) Lang et al. 20106b 77 B6 D2 RI strains D2 (male) Lang et al. 2010

    96 B6 D2 RI strains D2 (male and female) Lang et al. 2010113 B6 D2 RI strains D2 (male) Lang et al. 2010

    7 3 B6 D2 RI strains B6 (female) Lang et al. 2010 X11 B6 D2 RI strains B6 (female) Gelman et al. 1988 X66 (BALB/cJ B6)

    (C3H D2)BALB (male) Miller et al. 1998 X X

    73 B6 D2 RI strains B6 (female) Lang et al. 2010 X X92 B6 D2 RI strains B6 (female and male) Lang et al. 2010111 (BALB/cJ B6)

    (C3H D2)BALB (male) Miller et al. 1998

    8 15 B6 D2 RI strains B6 (female) Lang et al. 2010 X X26 (LP MOLD)

    (NZW BALB)MOLD (pooled) Klebanov et al. 2001 X X

    111 B6 D2 RI strains B6 (female) Lang et al. 20109 91 (BALB/cJ B6)

    (C3H D2)C3H (male) Jackson et al. 2002;

    Miller et al. 2002aX

    10 48 (BALB/cJ B6) (C3H D2)

    D2 (male) Miller et al. 1998 X

    66 (BALB/cJ B6) (C3H D2)

    D2 (male) Jackson et al. 2002; Miller et al. 2002a

    109 B6 D2 RI strains D2 (male) Lang et al. 2010 X119 (ST B6)

    (CAST D2)CAST (pooled) Klebanov et al. 2001 X

    11a 15 B6 D2 RI strains B6 (female) de Haan et al. 1998 X18 B6 D2 RI strains B6 (male and female) Lang et al. 2010 X35 B6 D2 RI strains B6 (female) Lang et al. 201056 B6 D2 RI strains B6 (male) Lang et al. 2010 X

    12 60 B6 D2 RI strains D2 (female) Gelman et al. 1988105 (BALB/cJ B6)

    (C3H D2)B6/C3H (female and male)

    Jackson et al. 2002; Miller et al. 2002a

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • Volume 52, Number 1 2011 13

    Table 4 (continued)

    ChraPeak (Mb) Cross

    High allele strain (sex) Reference

    Replicated in mice

    Replicated in humans

    16 6 (BALB/cJ B6) (C3H D2)

    BALB (female) Jackson et al. 2002; Miller et al. 1998, 2002a

    32 B6 D2 RI strains B6 (male) Lang et al. 2010 X64 B6 D2 RI strains B6 (female) Lang et al. 2010

    17 34 (B6 D2) D2 B6 (male) Yunis et al. 198418 53 (BALB/cJ B6)

    (C3H D2)D2 (male) Miller et al. 1998

    19 30 (BALB/cJ B6) (C3H D2)

    BALB (female) Miller et al. 1998 X

    32 ILS ISS RI strains ILS (female) Rikke et al. 2010 X47 (BALB/cJ B6)

    (C3H D2)D2 (male) Miller et al. 1998

    X 49 B6 D2 RI strains D2 (female) Lang et al. 2010126 B6 D2 RI strains D2 (female) Lang et al. 2010

    Chr, chromosome; RI, recombinant inbredEach suggestive and signifi cant QTL is listed with the chromosomal peak in Mb (derived from the corrected mouse map [Cox et al. 2009] and the Mouse Map Converter from the Center for Genome Dynamics [http://cgd.jax.org/mousemapconverter/]), the cross in which the QTL was found, the allele conferring longer lifespan, and the reference. The QTL near the bottom of Chr 7 was originally reported with D12Mit38 as peak marker (Miller et al. 1998), but this particular marker was incorrectly mapped; it properly belongs on Chr 7 at Mb 111 and is now called D7Mit1000. aChromosomes 3 and 13-15 are missing because no QTLs affecting lifespan have been reported on them.bAlthough Lang and colleagues (2010) reported QTLs for males and females separately, we combined the two examples for which QTLs were found in both sexes at the same spot (Chr 6 at 96 Mb and Chr 11 at 18 Mb).

    mouse and human genomes. As genes are identifi ed in hu-mans, mouse models will continue to be very useful in ef-forts to investigate underlying mechanisms of the genes that affect aging. We expect to see growing numbers of transla-tional studies demonstrating the relevance of the mouse to human aging. This rise, combined with increasingly refi ned bioinformatic tools and mouse models, will accelerate the identifi cation of genes that delay human aging and extend healthful lifespan.

    Acknowledgments

    The authors thank Drs. Kevin Flurkey and James Nelson for their constructive comments on the manuscript, Jesse Hammer for preparation of the fi gure, and Joanne Currer for editing of the manuscript. This work was supported by grants from the Glenn Foundation (BP), the Ellison Medical Founda-tion (BP), and the Nathan Shock Center (grant AG038070; LLP).

    References

    Baker DJ, Jeganathan KB, Cameron JD, Thompson M, Juneja S, Kopecka A, Kumar R, Jenkins RB, de Groen PC, Roche P, van Deursen JM. 2004. BubR1 insuffi ciency causes early onset of aging-associated phe-notypes and infertility in mice. Nat Genet 36:744-749.

    Bluher M, Kahn BB, Kahn CR. 2003. Extended longevity in mice lacking the insulin receptor in adipose tissue. Science 299:572-574.

    Boguski MS. 2002. Comparative genomics: The mouse that roared. Nature 420:515-516.

    Bonafe M, Barbieri M, Marchegiani F, Olivieri F, Ragno E, Giampieri C, Mugianesi E, Centurelli M, Franceschi C, Paolisso G. 2003. Polymor-phic variants of insulin-like growth factor I (IGF-I) receptor and phos-phoinositide 3-kinase genes affect IGF-I plasma levels and human longevity: Cues for an evolutionarily conserved mechanism of life span control. J Clin Endocrinol Metab 88:3299-3304.

    Brown-Borg HM, Borg KE, Meliska CJ, Bartke A. 1996. Dwarf mice and the ageing process. Nature 384:33.

    Chen YF, Wu CY, Kao CH, Tsai TF. 2010. Longevity and lifespan control in mammals: Lessons from the mouse. Ageing Res Rev 9:S28-S35.

    Chiu CH, Lin WD, Huang SY, Lee YH. 2004. Effect of a C/EBP gene re-placement on mitochondrial biogenesis in fat cells. Genes Dev 18:1970-1975.

    Churchill GA, Airey DC, Allayee H, Angel JM, Attie AD, Beatty J, Beavis WD, Belknap JK, Bennett B, Berrettini W, Bleich A, Bogue M, Broman KW, Buck KJ, Buckler E, Burmeister M, Chesler EJ, Cheverud JM, Clapcote S, Cook MN, Cox RD, Crabbe JC, Crusio WE, Darvasi A, Deschepper CF, Doerge RW, Farber CR, Forejt J, Gaile D, Garlow SJ, Geiger H, Gershenfeld H, Gordon T, Gu J, Gu W, de Haan G, Hayes NL, Heller C, Himmelbauer H, Hitzemann R, Hunter K, Hsu HC, Iraqi FA, Ivandic B, Jacob HJ, Jansen RC, Jepsen KJ, Johnson DK, Johnson TE, Kempermann G, Kendziorski C, Kotb M, Kooy RF, Llamas B, Lammert F, Lassalle JM, Lowenstein PR, Lu L, Lusis A, Manly KF, Marcucio R, Matthews D, Medrano JF, Miller DR, Mittleman G, Mock BA, Mogil JS, Montagutelli X, Morahan G, Morris DG, Mott R, Nadeau JH, Nagase H, Nowakowski RS, OHara BF, Osadchuk AV, Page GP, Paigen B, Paigen K, Palmer AA, Pan HJ, Peltonen-Palotie L, Peirce J, Pomp D,

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • 14 ILAR Journal

    Pravenec M, Prows DR, Qi Z, Reeves RH, Roder J, Rosen GD, Schadt EE, Schalkwyk LC, Seltzer Z, Shimomura K, Shou S, Sillanpaa MJ, Siracusa LD, Snoeck HW, Spearow JL, Svenson K, Tarantino LM, Threadgill D, Toth LA, Valdar W, de Villena FP, Warden C, Whatley S, Williams RW, Wiltshire T, Yi N, Zhang D, Zhang M, Zou F. 2004. The Collaborative Cross, a community resource for the genetic analysis of complex traits. Nat Genet 36:1133-1137.

    Conover CA, Bale LK. 2007. Loss of pregnancy-associated plasma protein A extends lifespan in mice. Aging Cell 6:727-729.

    Conti B, Sanchez-Alavez M, Winsky-Sommerer R, Morale MC, Lucero J, Brownell S, Fabre V, Huitron-Resendiz S, Henriksen S, Zorrilla EP, de Lecea L, Bartfai T. 2006. Transgenic mice with a reduced core body temperature have an increased life span. Science 314:825-828.

    Coschigano KT, Holland AN, Riders ME, List EO, Flyvbjerg A, Kopchick J. 2003. Deletion, but not antagonism, of the mouse growth hormone receptor results in severely decreased body weights, insulin, and insulin-like growth factor I levels and increased life span. Endocrinology 144:3799-3810.

    Cox A, Ackert-Bicknell CL, Dumont BL, Ding Y, Bell JT, Brockmann GA, Wergedal JE, Bult C, Paigen B, Flint J, Tsaih SW, Churchill GA, Broman KW. 2009. A new standard genetic map for the laboratory mouse. Genetics 182:1335-1344.

    de Haan G, Gelman R, Watson A, Yunis E, Van Zant G. 1998. A putative gene causes variability in lifespan among genotypically identical mice. Nat Genet 19:114-116.

    de Magalhaes JP, Cabral JA, Magalhaes D. 2005. The infl uence of genes on the aging process of mice: A statistical assessment of the genetics of aging. Genetics 169:265-274.

    Dellagnello C, Leo S, Agostino A, Szabadkai G, Tiveron C, Zulian A, Prelle A, Roubertoux P, Rizzuto R, Zeviani M. 2007. Increased longev-ity and refractoriness to Ca(2+)-dependent neurodegeneration in Surf1 knockout mice. Hum Mol Genet 16:431-444.

    Flurkey K, Papaconstantinou J, Miller RA, Harrison DE. 2001. Lifespan extension and delayed immune and collagen aging in mutant mice with defects in growth hormone production. Proc Natl Acad Sci U S A 98:6736-6741.

    Flurkey K, Papaconstantinou J, Harrison DE. 2002. The Snell dwarf mutation Pit1(dw) can increase life span in mice. Mech Ageing Dev 123:121-130.

    Fontana L, Klein S. 2007. Aging, adiposity, and calorie restriction. JAMA 297:986-994.

    Fontana L, Partridge L, Longo VD. 2010. Extending healthy life span: From yeast to humans. Science 328:321-326.

    Garrett MR, Pezzolesi MG, Korstanje R. 2010. Integrating human and ro-dent data to identify the genetic factors involved in chronic kidney dis-ease. J Am Soc Nephrol 21:398-405.

    Gelman R, Watson A, Bronson R, Yunis E. 1988. Murine chromosomal re-gions correlated with longevity. Genetics 118:693-704.

    Goodrick CL. 1975. Life-span and the inheritance of longevity of inbred mice. J Gerontol 30:257-263.

    Grubb SC, Maddatu TP, Bult CJ, Bogue MA. 2009. Mouse phenome data-base. Nucleic Acids Res 37:D720-D730.

    Hakimi P, Yang J, Casadesus G, Massillon D, Tolentino-Silva F, Nye CK, Cabrera ME, Hagen DR, Utter CB, Baghdy Y, Johnson DH, Wilson DL, Kirwan JP, Kalhan SC, Hanson RW. 2007. Overexpression of the cyto-solic form of phosphoenolpyruvate carboxykinase (GTP) in skeletal muscle repatterns energy metabolism in the mouse. J Biol Chem 282:32844-32855.

    Harper JM, Wilkinson JE, Miller RA. 2010. Macrophage migration inhibi-tory factor-knockout mice are long lived and respond to caloric restric-tion. FASEB J 24:2436-2442.

    Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandez E, Miller RA. 2009. Rapamycin fed late in life extends lifespan in ge-netically heterogeneous mice. Nature 460:392-395.

    Heikkinen S, Argmann C, Feige JN, Koutnikova H, Champy MF, Dali-Youcef N, Schadt EE, Laakso M, Auwerx J. 2009. The Pro12Ala PPARgamma2 variant determines metabolism at the gene-environment interface. Cell Metab 9:88-98.

    Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC, Cervera P, Le Bouc Y. 2003. IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421:182-187.

    Jackson AU, Galecki AT, Burke DT, Miller RA. 2002. Mouse loci associ-ated with life span exhibit sex-specifi c and epistatic effects. J Gerontol A Biol Sci Med Sci 57:B9-B15.

    Kemnitz J. 2011. Caloric restriction and aging in nonhuman primates. ILAR J 52:66-77.

    Kenyon CJ. 2010. The genetics of ageing. Nature 464:504-512.Klebanov S, Astle CM, Roderick TH, Flurkey K, Archer JR, Chen J,

    Harrison DE. 2001. Maximum life spans in mice are extended by wild strain alleles. Exp Biol Med (Maywood) 226:854-859.

    Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y, Kurabayashi M, Kaname T, Kume E, Iwasaki H, Iida A, Shiraki-Iida T, Nishikawa S, Nagai R, Nabeshima YI. 1997. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 390:45-51.

    Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, McGuinness OP, Chikuda H, Yamaguchi M, Kawaguchi H, Shimomura I, Takayama Y, Herz J, Kahn CR, Rosenblatt KP, Kuro-o M. 2005. Suppres-sion of aging in mice by the hormone Klotho. Science 309:1829-1833.

    Kwan KY, Wang JC. 2001. Mice lacking DNA topoisomerase IIIbeta de-velop to maturity but show a reduced mean lifespan. Proc Natl Acad Sci U S A 98:5717-5721.

    Lang DH, Gerhard GS, Griffi th JW, Vogler GP, Vandenbergh DJ, Blizard DA, Stout JT, Lakoski JM, McClearn GE. 2010. Quantitative trait loci (QTL) analysis of longevity in C57BL/6J by DBA/2J (BXD) recombi-nant inbred mice. Aging Clin Exp Res 22:8-19.

    Larsson NG. 2010. Somatic mitochondrial DNA mutations in mammalian aging. Annu Rev Biochem 79:683-706.

    Li Q, Ren J. 2007. Infl uence of cardiac-specifi c overexpression of insulin-like growth factor 1 on lifespan and aging-associated changes in cardiac intracellular Ca2+ homeostasis, protein damage and apoptotic protein expression. Aging Cell 6:799-806.

    Liao CY, Rikke BA, Johnson TE, Diaz V, Nelson JF. 2010. Genetic variation in the murine lifespan response to dietary restriction: From life exten-sion to life shortening. Aging Cell 9:92-95.

    Liu X, Jiang N, Hughes B, Bigras E, Shoubridge E, Hekimi S. 2005. Evolu-tionary conservation of the clk-1-dependent mechanism of longevity: Loss of mclk1 increases cellular fi tness and lifespan in mice. Genes Dev 19:2424-2434.

    Migliaccio E, Giorgio M, Mele S, Pelicci G, Reboldi P, Pandolfi PP, Lanfrancone L, Pelicci PG. 1999. The p66shc adaptor protein controls oxidative stress response and life span in mammals. Nature 402:309-313.

    Miller RA, Chrisp C, Jackson AU, Burke D. 1998. Marker loci associated with life span in genetically heterogeneous mice. J Gerontol A Biol Sci Med Sci 53:M257-M263.

    Miller RA, Chrisp C, Jackson AU, Galecki AT, Burke DT. 2002a. Coordi-nated genetic control of neoplastic and nonneoplastic diseases in mice. J Gerontol A Biol Sci Med Sci 57:B3-B8.

    Miller RA, Harper JM, Dysko RC, Durkee SJ, Austad SN. 2002b. Longer life spans and delayed maturation in wild-derived mice. Exp Biol Med 227:500-508.

    Miller RA, Harper JM, Galecki A, Burke DT. 2002c. Big mice die young: Early life body weight predicts longevity in genetically heterogeneous mice. Aging Cell 1:22-29.

    Miller RA, Harrison DE, Astle CM, Floyd RA, Flurkey K, Hensley KL, Javors MA, Leeuwenburgh C, Nelson JF, Ongini E, Nadon NL, Warner HR, Strong R. 2007. An Aging Interventions Testing Program: Study design and interim report. Aging Cell 6:565-575.

    Morley JE, Chahla E, Alkaade S. 2010. Antiaging, longevity and calorie restriction. Curr Opin Clin Nutr Metab Care 13:40-45.

    Moskovitz J, Bar-Noy S, Williams WM, Requena J, Berlett BS, Stadtman ER. 2001. Methionine sulfoxide reductase (MsrA) is a regulator of antioxidant defense and lifespan in mammals. Proc Natl Acad Sci U S A 98:12920-12925.

    Mounkes LC, Kozlov S, Hernandez L, Sullivan T, Stewart CL. 2003. A progeroid syndrome in mice is caused by defects in A-type lamins. Nature 423:298-301.

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from

  • Volume 52, Number 1 2011 15

    Nadon NL, Strong R, Miller RA, Nelson J, Javors M, Sharp ZD, Peralba JM, Harrison DE. 2008. Design of aging intervention studies: The NIA Interventions Testing Program. Age (Dordr) 30:187-199.

    Neumann CA, Krause DS, Carman CV, Das S, Dubey DP, Abraham JL, Bronson RT, Fujiwara Y, Orkin SH, Van Etten RA. 2003. Essential role for the peroxiredoxin Prdx1 in erythrocyte antioxidant defence and tu-mour suppression. Nature 424:561-565.

    Newman AB, Walter S, Lunetta KL, Garcia ME, Slagboom PE, Christensen K, Arnold AM, Aspelund T, Aulchenko YS, Benjamin EJ, Christiansen L, DAgostino RB Sr, Fitzpatrick AL, Franceschini N, Glazer NL, Gudnason V, Hofman A, Kaplan R, Karasik D, Kelly-Hayes M, Kiel DP, Launer LJ, Marciante KD, Massaro JM, Miljkovic I, Nalls MA, Hernandez D, Psaty BM, Rivadeneira F, Rotter J, Seshadri S, Smith AV, Taylor KD, Tiemeier H, Uh HW, Uitterlinden AG, Vaupel JW, Walston J, Westendorp RG, Harris TB, Lumley T, van Duijn CM, Murabito JM. 2010. A meta-analysis of four genome-wide association studies of survival to age 90 years or older: The Cohorts for Heart and Aging Research in Genomic Epidemiology Consortium. J Gerontol A Biol Sci Med Sci 65:478-487.

    Paigen K. 1995. A miracle enough: The power of mice. Nat Med 1:215-220.

    Peters LL, Robledo RF, Bult CJ, Churchill GA, Paigen BJ, Svenson KL. 2007. The mouse as a model for human biology: A resource guide for complex trait analysis. Nat Rev Genet 8:58-69.

    Petkova SB, Yuan R, Tsaih SW, Schott W, Roopenian DC, Paigen B. 2008. Genetic infl uence on immune phenotype revealed strain-specifi c varia-tions in peripheral blood lineages. Physiol Genomics 34:304-314.

    Rikke BA, Liao CY, McQueen MB, Nelson JF, Johnson TE. 2010. Genetic dissection of dietary restriction in mice supports the metabolic effi -ciency model of life extension. Exp Gerontol 45:691-701.

    Sahin E, Depinho RA. 2010. Linking functional decline of telomeres, mito-chondria and stem cells during ageing. Nature 464:520-528.

    Schriner SE, Linford NJ, Martin GM, Treuting P, Ogburn CE, Emond M, Coskun PE, Ladiges W, Wolf N, Van Remmen H, Wallace DC, Rabinovitch PS. 2005. Extension of murine life span by overexpression of catalase targeted to mitochondria. Science 308:1909-1911.

    Selman C, Lingard S, Choudhury AI, Batterham RL, Claret M, Clements M, Ramadani F, Okkenhaug K, Schuster E, Blanc E, Piper MD, Al-Qassab H, Speakman JR, Carmignac D, Robinson IC, Thornton JM, Gems D, Partridge L, Withers DJ. 2008a. Evidence for lifespan exten-sion and delayed age-related biomarkers in insulin receptor substrate 1 null mice. FASEB J 22:807-818.

    Selman C, Lingard S, Gems D, Partridge L, Withers DJ. 2008b. Comment on Brain IRS2 signaling coordinates life span and nutrient homeosta-sis. Science 320:1012; author reply 1012.

    Selman C, Tullet JM, Wieser D, Irvine E, Lingard SJ, Choudhury AI, Claret M, Al-Qassab H, Carmignac D, Ramadani F, Woods A, Robinson IC, Schuster E, Batterham RL, Kozma SC, Thomas G, Carling D, Okkenhaug K, Thornton JM, Partridge L, Gems D, Withers DJ. 2009. Ribosomal protein S6 kinase 1 signaling regulates mammalian life span. Science 326:140-144.

    Storer JB. 1966. Longevity and gross pathology at death in 22 inbred mouse strains. J Gerontol 21:404-409.

    Strong R, Miller RA, Astle CM, Floyd RA, Flurkey K, Hensley KL, Javors MA, Leeuwenburgh C, Nelson JF, Ongini E, Nadon NL, Warner HR, Harrison DE. 2008. Nordihydroguaiaretic acid and aspirin increase lifespan of genetically heterogeneous male mice. Aging Cell 7:641-650.

    Sugiyama F, Churchill GA, Higgins DC, Johns C, Makaritsis KP, Gavras H, Paigen B. 2001. Concordance of murine quantitative trait loci for salt-induced hypertension with rat and human loci. Genomics 71:70-77.

    Suh Y, Atzmon G, Cho MO, Hwang D, Liu B, Leahy DJ, Barzilai N, Cohen P. 2008. Functionally signifi cant insulin-like growth factor I receptor mutations in centenarians. Proc Natl Acad Sci U S A 105:3438-3442.

    Sundberg JP, Sundberg BA, Schofi eld P. 2008. Integrating mouse anatomy and pathology ontologies into a phenotyping database: Tools for data capture and training. Mamm Genome 19:413-419.

    Sutter NB, Bustamante CD, Chase K, Gray MM, Zhao K, Zhu L, Padhukasahasram B, Karlins E, Davis S, Jones PG, Quignon P, Johnson GS, Parker HG, Fretwell N, Mosher DS, Lawler DF, Satyaraj E, Nordborg M, Lark KG, Wayne RK, Ostrander EA. 2007. A single IGF1 allele is a major determinant of small size in dogs. Science 316:112-115.

    Taguchi A, White MF. 2008. Response to Comment on Brain IRS2 sig-naling coordinates life span and nutrient homeostasis. Science 320:1012.

    Taguchi A, Wartschow LM, White MF. 2007. Brain IRS2 signaling coordi-nates life span and nutrient homeostasis. Science 317:369-372.

    Threadgill D, Miller D, Churchill G, de Villena FP. 2011. The Collaborative Cross: Recombinant inbred mouse population for the systems genetic era. ILAR J 52:24-31.

    Trifunovic A, Wredenberg A, Falkenberg M, Spelbrink JN, Rovio AT, Bruder CE, Bohlooly YM, Gidlof S, Oldfors A, Wibom R, Tornell J, Jacobs HT, Larsson NG. 2004. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature 429:417-423.

    Tsaih SW, Pezzolesi MG, Yuan R, Warram JH, Krolewski AS, Korstanje R. 2009. Genetic analysis of albuminuria in aging mice and concordance with loci for human diabetic nephropathy found in a genome-wide as-sociation scan. Kidney Int 77:173-175.

    Wang X, Paigen B. 2005. Genetics of variation in HDL cholesterol in hu-mans and mice. Circ Res 96:27-42.

    Wooley CM, Xing S, Burgess RW, Cox GA, Seburn KL. 2009. Age, experi-ence and genetic background infl uence treadmill walking in mice. Phys-iol Behav 96:350-361.

    Xing S, Tsaih SW, Yuan R, Svenson KL, Jorgenson LM, So M, Paigen BJ, Korstanje R. 2009. Genetic infl uence on electrocardiogram time inter-vals and heart rate in aging mice. Am J Physiol Heart Circ Physiol 296:H1907-H1913.

    Yamaza H, Komatsu T, Wakita S, Kijogi C, Park S, Hayashi H, Chiba T, Mori R, Furuyama T, Mori N, Shimokawa I. 2010. FoxO1 is involved in the antineoplastic effect of calorie restriction. Aging Cell 9:372-382.

    Yan L, Vatner DE, OConnor JP, Ivessa A, Ge H, Chen W, Hirotani S, Ishikawa Y, Sadoshima J, Vatner SF. 2007. Type 5 adenylyl cyclase dis-ruption increases longevity and protects against stress. Cell 130:247-258.

    Yang X, Doser TA, Fang CX, Nunn JM, Janardhanan R, Zhu M, Sreejayan N, Quinn MT, Ren J. 2006. Metallothionein prolongs survival and an-tagonizes senescence-associated cardiomyocyte diastolic dysfunction: Role of oxidative stress. FASEB J 20:1024-1026.

    Yuan R, Tsaih SW, Petkova SB, Marin de Evsikova C, Xing S, Marion MA, Bogue MA, Mills KD, Peters LL, Bult CJ, Rosen CJ, Sundberg JP, Harrison DE, Churchill GA, Paigen B. 2009. Aging in inbred strains of mice: Study design and interim report on median lifespans and circulat-ing IGF1 levels. Aging Cell 8:277-287.

    Yunis EJ, Watson AL, Gelman RS, Sylvia SJ, Bronson R, Dorf ME. 1984. Traits that infl uence longevity in mice. Genetics 108:999-1011.

    at Universidad A

    utnoma de G

    uerrero on March 6, 2015

    http://ilarjournal.oxfordjournals.org/D

    ownloaded from