Envejecimiento Toward a Control Theory

download Envejecimiento Toward a Control Theory

of 25

Transcript of Envejecimiento Toward a Control Theory

  • 7/31/2019 Envejecimiento Toward a Control Theory

    1/25

    Toward a Control TheoryAnalysis of Aging

    Michael P. Murphy1 and Linda Partridge2

    1Medical Research Council Dunn Human Nutrition Unit, Cambridge CB2 0XY,United Kingdom; email: [email protected]

    2Centre for Research on Aging, University College London, Department of BiologLondon WC1E 6BT, United Kingdom; email: [email protected]

    Annu. Rev. Biochem. 2008. 77:77798

    First published online as a Review in Advance onMarch 4, 2008

    The Annual Review of Biochemistry is online at

    biochem.annualreviews.org

    This articles doi:10.1146/annurev.biochem.77.070606.101605

    Copyright c 2008 by Annual Reviews.All rights reserved

    0066-4154/08/0707-0777$20.00

    Key Words

    cell metabolic history, metabolic control analysis, nonspecific

    damage

    AbstractAging is due to the accumulation of damage over time that affecthe function and survival of the organism; however, it has prove

    difficult to infer the relative importance of the many processes thcontribute to aging. To address this, here we outline an approac

    that may prove useful in analyzing aging. In this approach, the funtion of the organism is described as a set of interacting physiologic

    systems. Degradation of their outputs leads to functional decline andeath as a result of aging. In turn, degradation of the system ou

    puts is attributable to changes at the next hierarchical level dowthe cell, through changes in cell number or function, which are

    turn a consequence of the metabolic history of the cell. Within thframework, we then adapt the methods of metabolic control analy

    (MCA) to determine which modifications are important for aginThis combination of a hierarchical framework and the methodol

    gies of MCA may prove useful both for thinking about aging and fanalyzing it experimentally.

    777

    Click here for quick links to

    Annual Reviews content online,

    including:

    Other articles in this volume

    Top cited articles

    Top downloaded articles

    Our comprehensive search

    FurtherANNUAL

    REVIEWS

    http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/http://showhidebookmarks/
  • 7/31/2019 Envejecimiento Toward a Control Theory

    2/25

    Contents

    INTRODUCTION... . . . . . . . . . . . . . . 778A HIERARCHICAL FRAMEWORK

    FOR CONSIDERINGORGANISMAL AGING. . . . . . . . . 779

    DYSFUNCTION OF

    PHYSIOLOGICAL SYSTEMSDURING AGING . . . . . . . . . . . . . . . 780

    CHANGES IN CELL NUMBER,

    FUNCTION, AND

    PHENOTYPE DURINGAGING . . . . . . . . . . . . . . . . . . . . . . . . . 781

    Changes in Cell NumberDuring Aging . . . . . . . . . . . . . . . . . 783

    Changes in Cell Function andPhenotype During Aging . . . . . . 784

    CELL METABOLIC HISTORY . . . . 784

    Nonspecific Damage . . . . . . . . . . . . . 785Gene Expression

    and Cell Signaling . . . . . . . . . . . . . 786

    Consequences of Cell MetabolicHistory for Cell Survival,

    Replication, and Function. . . . . . 787

    OVERVIEW OF THEHIERARCHICAL

    DESCRIPTION OF AGING . . . . 788QUANTIFICATION OF THE

    FACTORS CONTRIBUTING

    TO AGING . . . . . . . . . . . . . . . . . . . . . 788Metabolic Control Analysis

    a n d A g i n g . . . . . . . . . . . . . . . . . . . . . 7 8 9

    Application of MCA to Aging . . . . . 790Practical Considerations for

    Applying MCA to Aging . . . . . . . 792

    INTRODUCTION

    Extensive biochemical, organismal, popula-tion, and comparative studies on aging have

    focused on qualitative and, sometimes, quan-

    titative assessment of traits that contribute tonormal aging. It is hence evident that aging is

    caused by accumulation of damage, resultingfrom a lack of capacity to protect, maintain,

    and repair somatic tissues over time (17).

    However, attempts to determine which pa

    ticular types of aging-related damage are keto loss of function have been largely unsu

    cessful because the diversity of sources antypes of damage is great and can vary wit

    tissue, organism, and age (2, 810). It coul

    reasonably, be argued that the developmen

    of a general description of aging is prematubecause we lack both detailed descriptive daand a sufficiently mature understanding of a

    ing to produce realistic models of the procesHowever, we believe that there is value in th

    development of conceptual frameworks thhelp direct attention to the kinds of data an

    experiments that could move toward a morquantitativedescription andanalysisof the a

    ing process.Before we outline our approach, we fir

    consider the properties of normal aging thit must accommodate. In nature, avoidindeath often depends on finding food, avoidin

    predators, keeping warm, and surviving infections. However, when these extrinsic ha

    ards are largely eliminated, the intrinsic aging process generally still leads to loss

    function and death (1, 4, 11, 12), althougsome organisms age slowly and, in some case

    seem hardly to age at all (13, 14). Averagand maximal life span under controlled con

    ditions are broadly predictable for a givespecies. This is a familiar fact but, neverth

    less, both intriguing and informative. Agin

    reduces the genetic contribution of an indvidual to the next generation and is hence di

    advantageous; no genes have evolved to caudeath. Instead, aging occurs through we

    and tear that leads to the progressive accumlation of damage. However, different kinds

    organisms evidentlyavoid, repair or withstandamage to different extents and hence diff

    in their rates of aging. This biodiversitycan bdue to different genomes. For instance, ma

    imum life spans can vary by orders of magnitude among different species of mamma

    and birds (4, 1517). In addition, mutationin single genes can greatly increase life spa

    in the budding yeast Saccharomyces cerevisia

    the nematode worm Caenorhabditis elegans,th

    778 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    3/25

    fruitflyDrosophila melanogaster,andthemouse

    (1820). How the genome is expressed withina species can also dramatically affect life span.

    For example, queens of social insects, such asants, bees, and wasps, tend to age much more

    slowly than genetically identical worker castes

    (21).Thus,thegenomeandhowitisexpressed

    constrain mortality and life span.However, genetic constraints on life spanare relatively loose because, even for genet-

    ically identical animals in a standardized en-vironment, there is considerable variability in

    life span (12, 2225), possibly in part owing toheterogeneity in robustness among individu-

    als from stochastic events at the level of thecell, tissue, and organism, despite their sim-

    ilar genotypes and environments (12, 22, 23,25, 26). Finally, several environmental inter-

    ventions, such as decreased temperature (27),lowered oxygen tension (28), and dietary re-striction (DR) (29, 30), can also increase life

    span.A combination of genetic determina-

    tion, environmental variation, and stochasticevents thus contribute to the probability of

    dying at each age, the age-specific mortality,P(t).ThereisnoapriorireasonforP(t)tohave

    anyparticulardependenceonageortobesim-ilar for different organisms. However, as first

    noted for humans by Gompertz (31), in manyorganisms, including the standard laboratory

    model organisms, nematode, fruit fly, and

    mouse, mortality increases roughly exponen-tially over the main part of adulthood (6, 12,

    13). Thus, a Gompertz plot of Ln P(t) againsttime is approximately linear over this region,

    although there are many exceptions (13).The challenges are to explain how normal

    wear and tear at a metabolic level can accountforthegeneralpropertiesoforganismalaging,

    how interventions act to alter life span, andwhy there is such variation in aging within and

    between species. One obstacle is that muta-tions and environmental interventions affect

    organisms at many levels of function, mak-

    ing it difficult to pinpoint how an interven-tion affects aging. To illustrate, consider how

    the poison cyanide leads to death. Is it due

    Dietary restrictio(DR): decreasingthe nutrient supplyto organisms whileavoidingmalnutrition exten

    life span

    Age-specificmortality [P(t)]: tprobability of deathfor an organism atany given age

    Metabolic controlanalysis (MCA): aexperimentalapproach developeto understand howcontrol is distribut

    within metabolicpathways andnetworks

    to inhibition of cytochrome oxidase? Loss of

    mitochondrial proton motive force? Defec-tive ATP synthesis? Loss of control over ion

    gradients in the cell? Defective action of acti-nomyosin? Defective muscle cell contraction?

    Poor blood pumping by the heart? Clearly,

    eveninasimple,acutesituation,itisnotpossi-

    bletosaywhatkilledanorganismwithoutfirstdelineating the interacting biochemical andphysiological entities and considering eachin-

    dependently to determine what precisely canlead to death. Similar complexities in biolog-

    ical processes at different levels of organiza-tion should be addressed to understand how

    normal aging occurs and to link biochemicalalterations to changes in function and in mor-

    tality. A second major issue affecting our un-derstanding of aging is to detemine the rela-

    tive significance or ranking of different typesof damage and biochemical modification fornormal aging. To address these two issues we

    have developed a hierarchical description ofhow the various levels of organization within

    an organism can contribute to aging. We thenuse this framework to show how it may be

    possible to quantify and rank the factors thatcontribute to aging by applying concepts de-

    rived from metabolic control analysis (MCA).Over the next few sections, the description

    of the aging organism as a hierarchical sys-tem with interacting levels of organization is

    developed.

    A HIERARCHICAL FRAMEWORKFOR CONSIDERINGORGANISMAL AGING

    We first place the physiological and metabolic

    processes of an organism into interacting hi-erarchies so that it is clear how biochemical

    alterations affect aging. In the top level ofthe hierarchy, all of an organisms functions

    are ascribed to a set of physiological systems

    that interact with each other and the environ-ment (Figure 1). Each physiological system

    is considered to be a black box that onlycommunicates with other systems and the en-

    vironment through inputs and outputs. The

    www.annualreviews.org Control Theory Analysis of Aging 779

  • 7/31/2019 Envejecimiento Toward a Control Theory

    4/25

    Organism

    Environmental inputsIndependent outputsOutputs that are functions of inputs

    System A System B

    Figure 1

    Interacting physiological systems are shown schematically as twointeracting systems. These systems are affected by inputs from theenvironment and by inputs from other physiological systems. System

    outputs can originate within the system independently of other inputs orcan be dependent on the inputs from the environment and/or othersystems. Organisms can be divided into physiological systems in a numberof different ways; for example, human physiology can be described using11 systems: skin, respiratory system, circulatory system, central nervoussystem, endocrine system, reproductive system, lymphoid system,musculoskeletal system, urinary system, digestive system, and specialsense organs (33, 34). The increased mortality of the organism as it ages isdue to the decline over time of various systems outputs. These can beintrinsic where the system outputs become inappropriate during agingbecause of alterations within the system. Dysfunction in system outputscan also occur for undamaged systems when the inputs are inappropriate.

    outputs can occur independently of external

    inputs to the system, or they can be a func-tion of inputs from other systems or from

    the environment (Figure 1). This leads to aninteracting network of physiological systems

    that, in principle, gives a complete descrip-tion of the organisms functions. Although

    each system is functionally discrete, interact-ing only through inputs and outputs, physi-

    cal separation is not necessary. For example,the immune system is largely composed of in-

    dividual cells that distribute throughout thebody and that can infiltrate other tissues dur-

    ing inflammation and aging (32) but will still

    only connect with other systems through out-puts such as cytokine release.

    Each system can, in principle, be de-scribed by phenomenological models with-

    out any knowledge of its internal workin

    (Figure 1). System aging occurs over timthrough its output becoming inappropriat

    thus impairing organismal function and elvating mortality rate. Damage within the sy

    tem causes intrinsic dysfunction, leading

    inappropriate system outputs. An undamage

    system can also exhibit extrinsic dysfunctiowhen its outputs are inappropriate solely bcause the inputs from other physiological sy

    tems are inappropriate. For this black box dscription, it is not necessary to understan

    how the damage that leads to intrinsic dyfunction arises.

    The progressive dysfunction with aging the organism is therefore due to dysfunction

    ing of some or all of its systems as defineby their outputs. Of course, the practical di

    ficulties of defining a physiological systemof knowing all the interactions, and of a

    cidentally omitting unknown or unsuspecte

    interactions are enormous. Even so, thinkinof aging in this way can be useful both as

    heuristic exercise and to assist in identifyinfruitful avenues for experimental work. In th

    next section, we consider the types of systedysfunction that can contribute to aging.

    DYSFUNCTION OF

    PHYSIOLOGICAL SYSTEMSDURING AGING

    System function generally declines with aging;examples include humankidney glomer

    larfiltration rate (35); musclestrengthin micflies, and humans (36, 37); mammalian neu

    rological function as measured by memoformation (38, 39); -cell insulin secretio

    in humans (40); and the mammalian immunsystem (41, 42). Studies of functionally iso

    lated systems, such as pancreatic islets (40show that intrinsic system dysfunction can o

    cur, but in vivo it is usually not clear if d

    cline is due to intrinsic damage or defectivinputs. For example, stem cell activity in mi

    declines on aging, but can be ameliorated bparabiotic pairings, whereby a young and a

    old mouse share a circulatory system (43

    780 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    5/25

    suggesting that at least part of thedecline with

    age depends upon systemic factors from otherphysiological systems.

    Important questions are whether all sys-tems within an individual decline and whether

    the relative declines in different systems con-

    tribute similarly to mortality. From an evo-

    lutionary perspective, natural selection mayadjust costly tissue maintenance to maximizereproductive success and thereby lead to sim-

    ilar rates of functional decline for all tissues(44). Many or all physiological systems within

    an organism would then wear out at sim-ilar rates, but with those rates determined

    at the level of the individual system. In ad-dition, similar rates of functional decline of

    systems within an organism could occur be-cause the outputs of a dysfunctioning sys-

    tem caused dysfunction in connected systems(Figure 1), even though their intrinsic ratesof decline were different. An alternative ex-

    planation for parallel rates of aging in differ-ent tissues would be the existence of an ag-

    ing process common to differentphysiologicalsystems (45). Supporting this idea, mutations

    in single genes can extend healthy life spanby ameliorating many forms of aging-related

    damage(e.g.,1820),pointingtotheexistenceof a single common aging process. Further-

    more, the evolutionary conservation of the ef-fects on aging of some of these mutations be-

    tween yeast, worms, flies, and mice raises the

    possibility of a similar underlying aging pro-cess in these very different organisms. Alter-

    natively, the rate of aging of individual phys-iological systems could vary idiosyncratically,

    according to genetic susceptibility and envi-ronmental and stochastic events, with no sin-

    gle biological age attributable to an individualorganism (46). Clearly, some aspects of ag-

    ing can be organ specific and caused, for ex-ample, entirely by differences in environment

    as with skin aging and exposure to sunlight(47). Determining whether physiological sys-

    tems within an individual organism decline in

    function at similar rates, and whether declinein all systems or in only a few key systems

    contributes to aging, is basic to understand-

    Oxidative damagenonspecific damageto biologicalmolecules caused breactive derivativesof oxygen

    ing aging. However, multiple traits in single

    individuals are seldom investigated during ag-ing, and even fewer studies have examined

    functional decline in different physiologicalsystems. Markers of aging have been investi-

    gated, such as common changes in RNA tran-

    script profiles during aging in different tissues

    (37, 48) andthe accumulation of similar mark-ers of oxidative damage (49). However, therelevance of these markers to system function

    is unclear (50).Measurements are needed of the rela-

    tive rates of decline in physiological systemswithin individual organisms over time to com-

    pare how they vary from individual to individ-ual within a population and how they respond

    to interventions that extend life span. An evengreater challenge is in comparing the relative

    importanceofdeclineinthefunctionofdiffer-ent systems to functional decline of the organ-ism and probability of death. At the moment

    there are no methods to quantify, or even torank, the relative roles of the functional de-

    clines of different physiological systems in thefunctioning and probability of death of the

    whole organism. Consequently, although ag-ing can be ascribed to the relative decline in

    various systems, as is outlined in Figure 2,which shows that changes in system function

    lead to alterations in P(t), considerable chal-lenges remain in quantifying how the various

    systems contribute to normal aging. In addi-

    tion, so far each system has been treated asa black box. To understand how dysfunction

    occurs within systems during aging, we lookat the next level down in the hierarchy at the

    constituent cells of the systems.

    CHANGES IN CELL NUMBER,FUNCTION, AND PHENOTYPE

    DURING AGING

    Many kinds of changes to cells occur during

    aging, but only those that affect the functionaloutputs of physiological systems will influence

    aging (Figure 2). Most changes in the outputsof systems are due to alterations in their con-

    stituent cells. Even changes outside cells, such

    www.annualreviews.org Control Theory Analysis of Aging 781

  • 7/31/2019 Envejecimiento Toward a Control Theory

    6/25

    P(t)Age-specificmortality

    Changes in thephysiologicalsystem

    Changes in cellpopulation andfunction

    Cellmetabolichistory

    Changes incell function

    Cell death

    Senescence

    Proliferation

    Changes incell number

    Protectivemechanisms

    Gene expression/cell signaling

    Nonspecific damageOxidative damage

    RadiationProtein unfolding

    Changes insystem function

    Figure 2

    Aging of an organism is due to the decline in function of the top-level physiological systems into whichthe organism has been divided. This dysfunction leads to changes in system outputs that have a greater olesser influence on the age-specific mortality [P(t)], as indicated by the variable width of the arrowslinking to mortality. Dysfunction of each system is in turn due to changes in either the number orfunction of its constituent cells. The changes to cells are caused by their metabolic history and are due tnonspecific damage and to changes in signaling pathways and gene expression. These in turn lead toeffects on cell function and on cell number.

    782 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    7/25

    as in the extracellular matrix or exoskeleton,

    can often be ascribed to changes in cell func-tion. For example, the accumulation of ex-

    tracellular debris such as fatty plaques can beassigned to the dysfunction of macrophages.

    Bone is a dynamic systemcontrolled by the ac-

    tivity of osteoclasts and osteoblasts. The form

    of structures that no longer contain livingcells, such as tooth enamel or hair, are ascrib-able to the cells that constructed them. Even

    so, some age-related modifications, such asglycation-mediated loss of elasticity of blood

    vessel walls or damage to lens proteins, may bedifficult to ascribe to cell function. However,

    these modifications occur at the same hierar-chical level as cell changes; therefore, they af-

    fect system function in much the same way aschanges to cell function. Thus, the functional

    declines of physiological systems during agingare caused by changes in cell number or func-tion or by changes in noncellular components

    of the systems.

    Changes in Cell NumberDuring Aging

    During aging many tissues undergo changes

    in cell number owing to cell death and to dis-ruption of the mechanisms that maintain cell

    number (51). The change in cell content withaging varies tremendously with tissue and or-ganism, with both cell loss and hyperplasia

    possible (51), as well as infiltration of somecell types, such as adipocytes or lymphocytes,

    into other tissues (32, 42, 52).In postmitotic tissues, including most tis-

    sues in adult flies and all tissues in nematodesapart from the gonad, cells that are lost are not

    replaced (5356). In mammals, therefore, thenumber of cells in many postmitotic tissues

    decreases with age, and this cell loss often cor-relates with a decline in system function. For

    example, the loss of functioning glomeruli in

    aging kidney correlates with decreased organfunction with age (35). However, cell loss with

    aging is not general for all postmitotic tissues.One example is the mammalian brain where

    there is no evidence for neuron loss with age

    (38,39),eventhoughintheadultbrainthereisvery limited capacity to replace lost cells (57),

    and this is restricted to the dentate gyrus andhippocampus (58, 59).

    In mitotic tissues when differentiated cellsare lost, they can be replenished by divi-

    sion of other differentiated cells (e.g., mam-malian liver) or from a pool of pluripotent,self-replenishing stem cells (e.g., mammalian

    gut endothelium, fly ovary) (60, 61). How-ever, many differentiated cells in mitotic tis-

    sues divide less readily with age in vivo, withan increasing proportion entering replicative

    senescence (51, 6264). For example, in oldbaboons, more than 15% of skin fibroblasts

    exhibit markers of senescence (62). Yet, theextent to which the accumulation of senescent

    cells contributes to diminished mitotic capac-ity and to decreased cell function with aging is

    uncertain. In addition, mammalian stem cells

    are less effective at replacing lost cells as theorganism ages (57, 60, 65). However, it is un-

    clear if decline in stem cell function causes de-cline in number of differentiated cells. There-

    fore in mitotic tissues, cell number can declineon aging through increased cell death, dimin-

    ished replacement of lost cells, or both.Cell number can also increase with age.

    For example, the number of adipocytes in hu-man visceral adipose tissue increases (52), and

    the increased tissue mass can raise produc-tion of proinflammatory cytokines that influ-

    ence the function of other systems (52). Cell

    hyperplasia also occurs in many tissues dur-ing aging, producing nodules that can dis-

    rupt system function (e.g., 66, 67) and thatcan also develop into cancers. Indeed, aging

    has been described as the most potent of allcarcinogens. Metastatic cancers can undergo

    extensive genetic and epigenetic alterations,change their location in the body, and inter-

    fere in diverse ways with the function of otherphysiological systems. Similarly, some cells

    of the immune systems also infiltrate tissues,and during aging a proinflammatory state de-

    velops (e.g., 6870) whereby several tissues

    www.annualreviews.org Control Theory Analysis of Aging 783

  • 7/31/2019 Envejecimiento Toward a Control Theory

    8/25

    are invaded by various classes of immune

    cells, whichcontribute to pathogenesis duringaging (e.g., 71, 72).

    Change in cell number during aging de-pends on physiological context and on the

    signals and contacts received in vivo and is

    hence difficult to study in vitro. More experi-

    ments are needed to measure change in cellnumber in tissues in different systems dur-ing aging as well as in different individuals

    and species. It is also important to determinewhether changes are due to increased loss or

    defective replacement, which is often unclear.Most importantly, it is vital to determine how

    much cell loss or gain can occur before it im-pairs system functions and thereby determine

    how important change in cell number is fornormal aging.

    Changes in Cell Functionand Phenotype During Aging

    Changes in cell function with age could also

    affect system outputs. A decline in cell func-tion with age is often found, for example, in

    the synaptic transmission in neurons (38) andthe rates of contraction by musculoskeletal

    motor units (36). The finding that decline inneuronal function in the aging mammalian

    brain is associated with decreased numbersof synaptic connections and conduction, butnot with decreased cell number, indicates

    the importance of loss of function indepen-dent of cell loss (38). Cell phenotype has

    been investigated more often than has cellfunction. For example, muscle fiber size de-

    creases, with a decrease in myofilament num-bers and poor packing of sarcomeres in both

    fly and human muscle (73). Neurons shrinkand have fewer spines and dendrites, lower

    synapse concentrations, and myelin dystro-phy (38). Changes in gene expression, in-

    creased numbers of senescent cells, morpho-

    logical changes, and accumulation of damagemarkers with age (e.g., 49) can also occur.

    With age, there is a gradual divergence of cellphenotypes within a tissue, perhaps because

    of stochastic events (24, 74, 75). For example,

    gene expression varies more between individ

    ual cardiomyocytes in aging mouse heart thin young cells (73, 76), and small changes

    expression of many genes in the kidney cumulatively correlate with a small change

    cell function (32). However, the relevance these markers to loss of cell function is un

    clear. More importantly, the ways in whicdeclines in cell function cause systemdysfuntion have not been investigated systematical

    Changes in cell number and functiowithin a system are probably intimately r

    lated. Loss or dysfunction of cells with agcould have deleterious effects on the remain

    ing cells in the system, because they are likeon average to be working harder, spendin

    more time trying to restore themselves thomeostasis, and thus increasing the prob

    bility of cell death or dysfunction. In addtion, senescent and other damaged cells ca

    have a deleterious bystander effect by secre

    ing factors that enhance local inflammatioand tissue structure changes that may lead

    more cell death (51). These mechanisms mexplain why loss and dysfunction of cells in

    system should accelerate with age.Therefore, changes in cell number, fun

    tion, or phenotype can contribute to aginby altering the function of their physiolo

    ical systems (Figure 2). However, there aconsiderable uncertainties as to how these a

    terations may contribute to system functioand thus to mortality. To understand the bio

    chemical processes that lead to the loss of cel

    and the change in function that occur durinaging, we have to drop down to the next lev

    in the hierarchy, that of the individual cell.

    CELL METABOLIC HISTORY

    Cell dysfunction and death are attributabto the cells metabolic history, which is

    combination of the initial state of the ce

    and subsequent cumulative changes. Thewill lead on to the changes in cell fun

    tion and number that affect system functioand thereby mortality (Figure 2). The in

    tial state of the cell is due to its genom

    784 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    9/25

    developmental history, physical niche occu-

    pied within the organism, and epigenetic fac-tors that affect genome expression (77). All

    these factors combine to lead to a particu-lar state of its functional components. The

    cells subsequent metabolic history can cause

    permanent DNA sequence modification, al-

    ter gene expression, and change the func-tional machinery of the cell by nonspecificdamage, posttranslational modification, and

    environmental factors. These factors com-bine to determine the cells intrinsic proba-

    bility of dying, proliferating, or malfunction-ing over time. Similar factors affect the func-

    tion of noncellular components of systems,such as the extracellular matrix, that change

    their function on aging. Extrinsic factors fromother cells and the environment will also alter

    the cells chances of dying or malfunctioning.Factors clearly identified as important in thecells metabolic history are nonspecific dam-

    age and changes in gene expression and sig-naling pathways.

    Nonspecific Damage

    Accumulation of diverse forms of nonspecific

    damage to biomolecules with age is a majorcontributor to cell loss and dysfunction. Ox-

    idative damage has attracted the most inter-est (49, 78, 79), but processes such as thermaldenaturation, misincorporation of monomers

    into biopolymers, radiation, and inappropri-ate chemical reactions are also likely to be

    important. These damage processes are aninevitable consequence of carrying out thou-

    sands of chemical reactions in an enclosedspace containing many reactive molecules, re-

    sulting in a range of damaged andpoorlyfunc-tioning biomolecules and subcellular struc-

    tures during aging.A range of processes reduces formation

    and duration of damaging agents, protects

    the cell against the damage, and repairsor degrades the altered target biomolecules.

    Increased steady-state levels of damagedbiomolecules could reflect elevated genera-

    tion of damage, decreasedrepair and turnover,

    or a combination of both. Some types of dam-

    age, such as a misfolded protein, can in prin-ciple be dealt with by the cell, whereas oth-

    ers cannot, such as fixation of a DNA mu-tation or accumulation of damaged material

    that can be neither broken down nor removedfrom the cell. With damage repair, there is a

    steady-state balance between impact of dam-age and its avoidance, repair, removal, orsequestration. Thissteady statecould theoret-

    ically be set to prevent accumulation of non-specific damage to a level that affects function

    by devoting a sufficientamount of thecells re-sources to maintain itself indefinitely. For ir-

    reversible damage, which cannot be repaired,the cell could also decrease damage by com-

    mitting more resources to prevention. How-ever, there may be no evolutionary advantage

    in devoting resources to maintain an undam-aged somatic cell indefinitely at the expense of

    reproduction, and mechanisms that prevent,

    repair, or degrade damage are hence limiting,leading to cell dysfunction during aging.

    Damage accumulation to cellular lipids,proteins, and nucleic acids during aging is

    abundantly documented (e.g., reviewed in 49,8083). Furthermore, a role for nonspecific

    damage in normal aging is supported by stud-ies where life span is increased by reducing

    damage by, for example, overexpressing heatshock proteins in worms (84, 85) and increas-

    ing antioxidant defenses in the mouse (86).Autophagy is essential for some forms of life

    span extension in C. elegans (87). However,

    caution in interpretation is warranted becauseit is often unproven that these manipulations

    increase life span by decreasing damage accu-mulation, rather than by altering other pro-

    cesses such as signaling pathways.Nonspecific damage, such as oxidative

    damage, can impair the activities of enzymes,the fluidity of membranes, or the activity of

    organelles (49), and thus impair cell function.However, to demonstrate a role in normal ag-

    ing is less straightforward because we needto know whether damage affects cell survival

    or function in vivo sufficiently to affect the

    outputs of its physiological system and hence

    www.annualreviews.org Control Theory Analysis of Aging 785

  • 7/31/2019 Envejecimiento Toward a Control Theory

    10/25

    IGF: insulin-likegrowth factor

    aging.For example, mutation to nuclear DNA

    is an important candidate contributor to ag-ing because it is irreversible and it does oc-

    cur during aging. However, its contributionto cellular systems and organismal aging is

    still debated (88). Mutations to mitochondrial

    DNA also accumulate with age (89), but their

    role in cell dysfunction and organismal agingis more questionable because the normal mu-tation load to mitochondrial DNA during ag-

    ing may be insufficient to explain functionaldecline (90). Yet, there have been few detailed

    studies of the chain of events from accumu-lation of damage to biomolecules, to the ef-

    fects on cell functions, and hence cell sur-vival or function, through to the physiological

    system and aging. Cells may have consider-able thresholds for the accumulation of dam-

    age before function or survival is impaired,and because a major consequence for cells ofdamage accumulation is death, damage could

    have a major impactbut leave no obvious traceamong living cells in the organism.

    Gene Expression and Cell Signaling

    Changes in gene expression and cell signaling

    during normal aging could contribute to ag-ing by affecting the rate of accumulation of

    cell nonspecific damage or by independentlyaltering pathways that directly affect the abil-ity of the cell to function or survive. These

    changes would then act to influence the out-puts of physiological systems in such a way as

    to affect mortality (Figure 2).There is an extensive literature showing

    changes in gene expression and cell signal-ing pathways in cells during aging (9196).

    RNA transcript profiles have revealed a num-ber of changes in gene expression in mouse

    muscle (91), including decreases in expres-sion of genes encoding proteins involved in

    energy metabolism and increases in expres-

    sion of stress response genes (91). In additionthere is increased stochasticity and variabil-

    ity in expression between cells (73, 76). Thereare no systematic decreases in the expression

    of defense, protective, or repair pathways, and

    in fact, increases are often seen instead (91

    consistent with a response to increased damage on aging. Thus, a systematic downregu

    lation of protective pathways does not seeto account for aging, although it is possib

    that increased variability in gene expressiomay lead to stochastic decreases in protectiv

    systems in individual cells.Cellular signaling pathways are intimateinvolved in extension of organismal life spa

    by single gene mutations and environmental interventions. For example, DR, a mod

    erate reduction in food intake while avoiding malnutrition, extends life span in diver

    organisms, including budding yeast, nemtodes, fruit flies, and rodents (97). Furthe

    more, intensive study of DR in rodents hshown that it delaysor ameliorates theimpac

    of multiple forms of damage, dysfunctioand disease (29). Although it is unclear if th

    mechanisms by which DR extends life spa

    are evolutionarily conserved, recent work himplicated several evolutionarily conserve

    signaling pathways in the response to DRincluding the nutrient-sensing target of r

    pamycin signaling pathway (98100) and thinsulin/insulin-like growth factor (IGF) pat

    ways (101, 102). Mutations in genes encodincomponents of these same signaling pathwa

    can also extend healthy life span in yeast (e.g103105), C. elegans(19), Drosophila (99, 106

    and mouse (107, 108).The implication is that the altered acti

    ity of these pathways ameliorates the kinds

    damage that are normally limiting for organismal life span. For example, extension of li

    span by reduced insulin/IGF signaling is ofteassociated with up-regulation of cellular pat

    ways that increase the activity of stress resitance and cellular detoxification pathways

    C. elegans, Drosophila, and mouse (e.g., 109111). However, interventions such as DR ar

    associated with decreased damage accumulation, but this does not seem to be cause

    by an increase in defensive pathways, manof which actually decline during DR (112

    114). These interventions might also alt

    the threshold for the amount of cell damag

    786 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    11/25

  • 7/31/2019 Envejecimiento Toward a Control Theory

    12/25

    tumor load (51). The relationship between

    cell damage, function, and survival and theireffects on aging are complex. For example, al-

    though oxidative damage is often assumed tobe a major contributor to aging, in some mod-

    els such as in mice heterozygous for the mito-

    chondrial antioxidant enzyme manganese su-

    peroxide dismutase(130)andin the long-livedrodent the naked mole rat (131), the relation-ship between oxidative damage and life span

    is the opposite of that predicted. Thus, whatis required is a way of linking types of damage

    and changes in cell signaling and gene expres-sion within cells to changes in cell survival and

    cell function. In turn, these alterations need tobe linked to changes in system functions that

    affect mortality (Figure 2). It seems probablethat, for example, large amounts of damage to

    some cell types may be unimportant for mor-tality because they can withstand higher levelsof damage or because the damage does not af-

    fect critical cell functions and system outputsthat affect aging. In contrast, small amounts

    of damage in other cells may be critical foraging. More studies are required to measure

    how factors such as damage affect outputs andhow these in turn affect aging.

    OVERVIEW OF THE

    HIERARCHICAL DESCRIPTIONOF AGING

    The hierarchical description of aging is sum-marized in Figure 2. The increase in P(t) of

    the organism with time is due to system dys-functions that lead to changes over time in

    their functional outputs. These changes havea greater or lesser influence on the P(t), as in-

    dicated by the variable width of the arrowslinking to mortality. System dysfunction is,

    in turn, due to either changes in the numberor in the function of its constituent cells or

    equivalent components, owing to metabolic

    history. Interventions can only affect agingby changing system functions that alter the

    P(t) of that organism, and many changes thatoccur during aging will have no impact on

    mortality.

    The hierarchical approach helps clarify th

    contribution of various factors to aging, provides a framework for discussing aging that

    internally consistent, and accommodates thfact that different tissues and organisms ma

    age through quite different pathways. Eve

    so, considerable challenges remain, particu

    larly in determining which factors withinhierarchy are most critical for aging and thuwhere we should focus interventions and e

    perimental effort. Approaches that can quantify, or at least rank, the contributions of va

    ious factors to aging at various levels of thhierarchies are required. How the hierarch

    cal description can be extended to be quantfied in ways that are useful for experimenta

    ists investigating aging is the topic of the nesection.

    QUANTIFICATION OF THEFACTORS CONTRIBUTING

    TO AGING

    The hierarchical description of aging prvides a framework that helps us pose appr

    priate questions about the kinds of processthat contribute to aging and clarifies how bi

    chemical alterations within cells impact oaging through their effects on physiologic

    systems (Figure 2). However, even when cosidering the changes that occur during aging in a hierarchical context, we continual

    come up against the problem of how to dtermine whether a process contributes to ag

    ing or not. Even if it does, the challenges athen to quantify or rank the relative contribu

    tions of different processes, at different levein the hierarchy, to aging within an organis

    and determine how these contributions vawithin and between species and are affected b

    interventions that affect life span. There armany questions central to aging that requir

    a quantitative answer. For example, we wou

    like to be able to assess the relative impotance of different physiological systems an

    system outputs for aging and to know whethcell dysfunction or loss of cells during aging

    more important. For cell loss, is accumulatio

    788 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    13/25

    of senescent cells, loss of stem cells, or the

    increased rate of cell loss most important?Which processes within a cell contribute most

    to its loss of function or inabilityto replicate asit ages? What forms of nonspecific damageare

    of most importance to the decline in cell func-

    tion during normal aging? What is required

    is a way of incorporating quantification, or atleast ranking, into the hierarchical approachwe have described to determine which system

    or process does contribute to aging, whichis most important, and, hence, where inter-

    ventions are likely to have most impact onaging.

    If they existed, robust, quantifiable mod-els of aging might be useful to determine the

    processes of greatest significance. However,modeling aging is problematic at several lev-

    els. An immediate hurdle is that our lack ofdetailed knowledge of the processes involvedmakes modeling of aging premature. A further

    challenge is the dynamic aspect; during aging,the systems themselves change with time, and

    any modeling approach has to accommodatethis. In addition, time lags are likely to be im-

    portant. Processes such as cell loss may occurdecades before they affect function or proba-

    bility of death. Therefore, although there area number of interesting approaches under de-

    velopment to model aging (e.g., 2, 810), ro-bust and quantifiable models of aging that can

    rank the importance of systems, cells, and cel-

    lular components to aging are not yet on thehorizon.

    Therefore, we require an empirical ap-proach that would enable us to identify the

    factors that contribute to aging and to deter-mine the relative importance of these factors

    to aging in experimental animals, despite ourincomplete knowledge of the system and lim-

    ited means of intervening to alter the rate ofaging. We think a promising way to do this is

    to adapt aspects of metabolic control analysis(MCA). In the following sections, we describe

    MCA and show how it may be used to design

    and interpretexperiments, usingcurrent tech-nologies to answer important questions about

    aging.

    Metabolic Control Analysisand Aging

    MCA was initially developed independently

    by Kacser & Burns (132, 133) and Heinrich& Rappoport (134) and has since been de-

    veloped and used to describe the control and

    regulation of a range of metabolic pathwaysand networks (135139). In considering the

    control of a metabolic pathway by MCA, thefirst step is to develop an explicit definition

    of the limits of the system and of the measur-ablevariables,such as metabolicintermediates

    and pathway fluxes. Importantly, apart fromclearly defining its limits, there is no require-

    ment for a complete description of the system,and sections of it can be treated as black boxes

    to accommodate measurable variables. Once

    these measurable variables and their interac-tions are defined, the system is manipulated in

    small ways, andthe changing relationships be-tween the variables reveal the extent to which

    each step is controlling.Consider the analysis by MCA of a sim-

    ple metabolic pathway of intermediates con-nected by enzyme-catalyzed reactions to ad-

    dress which enzymatic steps exert control overthe overall pathway flux. To do this, the activ-

    ity of each enzymatic step in the pathway isvaried very slightly, independently of changes

    in other components of the pathway, and theeffect of this on the overall flux is determined.

    This simple example yields several interesting

    conclusions. A major one is a simple defini-tion of control, where the greater the change

    in the overall flux on altering the activity ofan enzyme, then the greater the control of

    that enzyme over flux. However, this changein overall flux will be the result of the change

    in enzymeactivityin thecontextof the system,as altering its activity impacts on the over-

    all flux by changing the concentrations of themetabolic intermediates that link it to the rest

    of the pathway. Thus, the control over fluxis a property of the pathway, not of the en-

    zyme in isolation. An important consequence

    of control being a system property is that sev-eral steps in a pathway can share control, and

    www.annualreviews.org Control Theory Analysis of Aging 789

  • 7/31/2019 Envejecimiento Toward a Control Theory

    14/25

  • 7/31/2019 Envejecimiento Toward a Control Theory

    15/25

    Increasing levelVariableDecreasing level

    ABCIncreasingmortality

    Mortalityreadout

    Decreasingmortality

    Levels lower thanseen in normal aging

    Range of levelsin normal aging

    Levels higher thanseen in normal aging

    Normal aging

    Figure 3

    Analysis of aging by metabolic control analysis. Here a generic indicator of mortality, the mortalityreadout, is plotted against a variable that is both decreased and increased relative to its level in normalpopulations. Each value of the mortality readout is determined for a separate population in which thevalue of the variable is altered. The central shaded area indicates how the variable alters in normal aging.Three scenarios are shown. In curve A, the variable has no impact on aging as varying it over the rangethat occurs in normal aging does not affect mortality. At high and low levels, it does impact on mortality,

    but because these occur outside the range found in normal aging, they do not contribute to aging. CurveB shows a variable that contributes to aging; increasing it in the central shaded area raises the level ofmortality, whereas decreasing it lowers mortality. In contrast, in curve C, increasing the variable in thecentral shaded area decreases mortality, and decreasing it increases mortality, as might happen if thisvariable were protective. For both curves B and C, the steepness of the slope as the curve passes throughthe point of unmodulated aging gives an indication of how controlling the two processes are over aging.In this example, the process described by curve B is more controlling over aging than that described bycurve C.

    at either end of curve A. However, as this

    increase in mortality occurs outside the rangeof values for that variable during normal ag-

    ing, illustrated by the central shaded area, this

    factor does not contribute to normal aging.The effect of a variable that is harmful to the

    organism is shown in curve B. Decreasing theamount of this variable increases longevity,

    whereas increasing it will lead to increasedmortality. Most importantly, the changes in

    the amount of the variable that affect agingoccur within the normal range of the vari-

    able during aging. At some point, decreasingthis variable further will impact on mortality,

    leading to increasing mortality, but this occursoutside the normal range of variation during

    aging. The effect of a variable that protects

    against aging is shown in curve C. Decreasingthe amount of the variable is harmful, but in-

    creasing it is protective, and most importantly,the changes in the amount of the variable that

    are sufficient to affect the mortality readout

    occur during normal aging. Many combina-

    tions of these three curves are possible, butthese illustrate the critical aspects. The most

    important point is that if a factor controls ag-ingthen thecurveof mortalityreadout against

    the variable of interest has a measurable slope

    as it passes through the region of the normallyaging population.

    Figure 3 indicates how ideas from MCAcan be used to determine whether or not a

    process contributes to aging. There is a well-developed mathematical apparatus for MCA

    that can be used to quantify and rank the con-tribution of various factors to the control of

    metabolic fluxes (135139). It is clear that thegreater the control of a process over aging the

    steeper the slope of curves such as B and C asthey pass through the point of normal aging in

    Figure 3. For example, it is clear that allowing

    for appropriate normalization, the process incurve B has more control over aging than that

    in curve C. Quantification and comparison

    www.annualreviews.org Control Theory Analysis of Aging 791

  • 7/31/2019 Envejecimiento Toward a Control Theory

    16/25

    of the relative control of different factors in

    MCA are done by comparing the normalizedfractional changes in an output, such as a flux,

    with the normalized fractional changes in thefactors being varied. This leads to dimension-

    less quantities called control coefficients that

    enable the proportion of control over a flux

    to be assigned to each process. Analysis ofthe control of aging requires similar carefulnormalization of the changes relative to the

    endogenous levels of the variable, requiring adetailed development of the parallels between

    the mathematical formalism of MCA and thecontrol of aging that will be described in fu-

    ture publications. Even so, the approach out-linedin Figure 3 showshowMCAcanbeused

    to determine if an intervention affects normalaging and to rank and quantify the contribu-

    tions of factors to aging. In the next section,the practical aspects of carrying out these ex-periments are considered.

    Practical Considerationsfor Applying MCA to Aging

    Here we consider how studies, such as thosedescribed in Figure 3, could be done using

    currently available animal models and tech-nologies. This approach requires (a) factors

    that may impact on aging to be modulated bya series of small increments and decrementsin different populations of experimental ani-

    mals and (b) the effects of these changes ona mortality readout for each population to be

    determined. In the first instance, such experi-ments can be done with populations of nema-

    todes or Drosophila because these are alreadyroutinely used in aging research and have the

    advantages of a short life span as well as easeof genetic manipulation and measurement of

    mortality.A number of mortality readouts could be

    chosen,butmeasuringtheslopeofaplotofLn

    P(t) against time for a population has a num-ber of attractions. Over much of the life span,

    it is linear; consequently, a large number of in-dividuals in a given population contribute to

    thereadout,it is already routinelymeasuredin

    aging research in worms and flies, and it gen

    erates a single number for each populatioNevertheless, other readouts of aging, such

    median life span or the intercept of curves Ln P(t) against time with the y-axisor dea

    within or by a certain time interval, may alprove useful.

    The variable whose effect on aging is beininvestigated will have to be increased and dcreased very slightly (perhaps by as little as

    few percent) and incrementally. The mortaity readout would be determined in a seri

    of populations of flies or nematodes whethe variable was modified slightly. In add

    tion, the range of values of the variable durinnormal aging would have to be measured.

    the variable was a protein, then in nematodand Drosophila, its expression level could b

    modulated downward by standard RNAi approaches and modified so as to decrease e

    pression of the protein by only a few pe

    cent. For a small increase in expression the same protein, a number of current ap

    proaches can be adapted to generate strainwith slightly increased expression levels of th

    target protein. After generating several diffeent populations, each with a slight variatio

    in the expression level of the protein of interest, their mortality readouts would then b

    measured and plotted against the level of thprotein of interest. If a type of damage, suc

    as oxidative damage or accumulation of mifolded protein was of interest, then this cou

    be increased slightly by addition of pharm

    cological or environmental stressors or bdecreasing expression of protective enzyme

    Damage could be decreased by addition protective agents or by increasing expressio

    of protective proteins. These approaches cabe extended by selectively expressing the pr

    teinsinonlyonetissueorphysiologicalsysteor by only changing expression at differen

    stages during the subjects life spans. Similarthe effect of cell number in a system cou

    be addressed by increasing or decreasing thexpression of toxic or protective proteins t

    modulate slightly the number of cells in

    tissue.

    792 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    17/25

    Consider some examples:the DAF2 recep-

    tor is known to impact on the life span of ne-matodes, and its activity is thought to corre-

    late inversely with life span. If the amount ofthis receptor was altered and compared with

    a mortality readout, we might predict a re-

    sult, similar to curve B in Figure 3. However,

    it could be that the effects of changing theamounts of these receptors on life span onlyoccur with large-scale changes, and the curve

    might look more like curve A. With increasedantioxidant defenses we might predict a curve

    such as C in Figure 3; however, it could bethat the thresholds for effects on mortality

    are such that there is no change relative tothe normal range over aging. Another inter-

    esting type of damage is increased mtDNAmutation load, where high levels clearly lead

    to an aging phenotype, but it is unclear if thisonly occurs because the dependence of mor-

    tality on mtDNA damage is a curve of type

    A. The approaches outlined should help de-termine whether a single factor can impact

    on aging. Many interesting questions in ag-ing research arise from environmental inter-

    ventions, such as DR, that alter life span, but

    it is difficult to determine which of the many

    changes that occur during DR are importantfor aging and which are not. This issue can beaddressed by the MCA approach by selecting

    plausible factors that change in DR and ma-nipulating these independently to see if they

    contribute to the changes in aging seen duringDR.

    Thus, by developing the MCA approachesand applying them to currently available ex-

    perimental models of aging using experimen-tal approaches that are already developed, we

    should be able to address a number of criticalquestions about the factors that control aging.

    SUMMARY POINTS

    1. Aging arises from the accumulation of damage resulting from a lack of capacity to

    protect, maintain, and repair somatic tissues over time. Accumulation of damage leads

    to loss of function and, ultimately, death.

    2. The rate of aging of individuals can vary as a result of genetic, epigenetic, and envi-

    ronmental variation as well as of stochastic events.3. The accumulation of damage during aging occurs at multiple levels, from the physi-

    ological system, through organs, to cells, and individual biomolecules. Not all of the

    changes that occur with age are likely to be causal in loss of function and increasedlikelihood of death, and it is often difficult to determine which factors are important

    for aging.

    4. One way of clarifying causality with events occurring at multiple levels during aging

    is to make explicit the hierarchical level under consideration and its relationshipto other levels. The mortality of the individual is ultimately due to the change in

    function of its physiological systems. These system changes are caused by changes in

    the number or function of its component cells. Cell changes are themselves due to themetabolic history of the cell and to its impact on the ability of the cell to function and

    survive. The aspects of the metabolic history of the cell that are important for agingare the accumulation of nonspecific damage and changes in cell signaling and gene

    expression.

    www.annualreviews.org Control Theory Analysis of Aging 793

  • 7/31/2019 Envejecimiento Toward a Control Theory

    18/25

    5. Even when the hierarchical description has been adapted, the remaining critical ques-

    tion in aging research is to develop methodologies that will enable the relative con-

    tributions of various metabolic changes to aging to be quantified and related to eachother. What is required is a methodology that can be used to address these questions

    in the experimental animal systems currently in use using technologies now avail-able. We suggest that adaptation of MCA to aging will enable significant progress in

    determining the relative importance of the factors that contribute most to aging.

    FUTURE ISSUES

    1. Can we successfully adapt the methodologies of MCA to aging in model organisms

    such as worms, flies, and mice so as to determine the relative contribution of variousfactors to aging? In doing so, is it possible to use current approaches such as RNAi

    to manipulate the levels of factors that are thought to contribute to aging? Is it alsopossible to use this approach to determinethe pathways through which changes during

    interventions such as DR occur?

    2. If the MCA approach proves fruitful in aging research, is it able to contribute toward

    answering critical questions, including: Can we quantify or rank the relative impor-tance of the functional outputs of different physiological systems that are important

    for aging? Can we quantify the contribution of changes in cell number and functionto the alteration in a systems functional outputs over aging? Is it possible to quantify

    or rank the importance of nonspecific damage and changes in gene expression and cellsignaling pathways as well as to determine how they affect cell function and survival

    in vivo?

    3. If quantification of the contribution of various processes to aging proves feasible, then

    are the critical factors for aging similar or different for individuals within a population

    and also between different species? Is the hierarchical description and application ofthe MCA approach helpful in developing new insights into aging and in suggestingnovel interventions that may affect aging? Can this approach be usefully extended to

    aging-associated degenerative diseases and to other complex, multifactorial diseases?

    DISCLOSURE STATEMENT

    The authors are not aware of any biases that might be perceived as affecting the objectivity this review.

    ACKNOWLEDGMENTS

    We thank Meredith Ross for drawing the figures and Martin Brand, Judith Campisi, HelenCocheme, David Gems, Aubrey de Gray, Andrew James, Nils-Goran Larsson, George Marti

    Richard Miller, Meredith Ross, and Thomas Von Zglinicki for helpful advice. We are gratefto the BBSRC, MRC, Wellcome Trust, and the European Communitys sixth Framewor

    Program for Research, Contract LSHM-CT-2004-503116, for financial support.

    794 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    19/25

    LITERATURE CITED

    1. Holliday R. 1995. Understanding Ageing. Cambridge: Cambridge Univ. Press2. Kirkwood TBL. 2005. Cell120:437473. Partridge L, Gems D. 2002. Nat. Rev. Genet. 3:165754. Partridge L, Gems D. 2006. Trends Ecol. Evol. 21:334405. Kirkwood TBL, Austad SN. 2000. Nature 408:233386. Finch CE. 1990. Longevity, Senescence and the Genome. Chicago: Univ. Chicago Press

    7. Nemoto S, Finkel T. 2004. Nature 429:149528. Kirkwood TB, Boys RJ, Gillespie CS, Proctor CJ, Shanley DP, Wilkinson DJ. 2003.Nat.

    Rev. Mol. Cell Biol. 4:243499. Gavrilov LA, Gavrilova NS. 2001. J. Theor. Biol. 213:52745

    10. Gavrilov LA, Gavrilova NS. 2004. Ann. NY Acad. Sci. 1019:5091211. Comfort A. 1979. The Biology of Senescence. London: Churchill Livingstone. 3rd ed.12. Finch CE, Kirkwood TBL. 2000. Chance, Development and Ageing. Oxford: Oxford Univ.

    Press13. Vaupel JW, Baudisch A, Dolling M, Roach DA, Gampe J. 2004. Theor. Popul. Biol. 65:339

    5114. Baudisch A. 2005. Proc. Natl. Acad. Sci. USA 102:826368

    15. Sibly RM, Collett D, Promislow DEL, Peacock DJ, Harvey PH. 1997. J. Zool. 243:11216. Ricklefs RE, Scheuerlein A. 2001. Exp. Gerontol. 36:8455717. Austad SN. 1997. Exp. Gerontol. 32:233818. Tatar M, Bartke A, Antebi A. 2003. Science 299:13465119. Kenyon C. 2005. Cell120:4496020. Piper MD, Skorupa D, Partridge L. 2005. Exp. Gerontol. 40:8576221. Keller L, Genoud M. 1997. Nature 389:9586022. Vaupel JW, Johnson TE, Lithgow GJ. 1994. Science 266:82623. Vaupel JW, Carey JR, Christensen K, Johnson TE, Yashin AI, et al. 1998. Science 280:855

    6024. Herndon LA, Schmeissner PJ, Dudaronek JM, Brown PA, Listner KM, et al. 2002.Nature

    419:8081425. Wu D, Rea SL, Yashin AI, Johnson TE. 2006. Exp. Gerontol. 41:2617026. Rea SL, Wu D, Cypser JR, Vaupel JW, Johnson TE. 2005. Nat. Genet. 37:8949827. Partridge L, Pletcher SD, Mair W. 2005. Mech. Ageing Dev. 126:354128. Honda Y, Honda S. 2002. Ann. NY Acad. Sci. 959:4667429. Masoro EJ. 2005. Mech. Ageing Dev. 126:9132230. Barger JL, Walford RL, Weindruch R. 2003. Exp. Gerontol. 38:13435131. Gompertz B. 1825. Philos. Trans. R. Soc. Lond. Ser. A 115:5138532. Rodwell GEJ, Sonu R, Zahn JM, Lund J, Wilhelmy J, et al. 2004. PLoS Biol. 2:e42733. Hunter PJ, Borg TK. 2003. Nat. Rev. Mol. Cell Biol. 4:2374334. Hunter P, Smith N, Fernandez J, Tawhai M. 2005. Mech. Ageing Dev. 126:18792

    35. Hoang K, Tan JC, Derby G, Blouch KL, Masek M, et al. 2003. Kidney Int. 64:14172436. Vandervoort AA. 2002. Muscle Nerve 25:172537. Zahn JM, Sonu R, Vogel H, Crane E, Mazan-Mamczarz K, et al. 2006. PLoS Genet.

    2:10586938. Dickstein DL, Kabaso D, Rocher AB, Luebke JI, Wearne SL, Hof PR. 2007. Aging Cell

    6:2758439. Morrison JH, Hof PR. 1997. Science 278:4121940. Chiu KC, Lee NP, Cohan P, Chuang LM. 2000. Clin. Endocrinol. 53:56975

    www.annualreviews.org Control Theory Analysis of Aging 795

  • 7/31/2019 Envejecimiento Toward a Control Theory

    20/25

    41. Lamberts SW, Van Den Beld AW, Van Der Lely AJ. 1997. Science 278:41924

    42. Gruver AL, Hudson LL, Sempowski GD. 2007. J. Pathol. 211:1445643. Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weismann IL, Rando TA. 2005. Natu

    433:7606444. Rose M. 1991. Evolutionary Biology of Aging. Oxford: Oxford Univ. Press

    45. Miller RA, Bookstein F, Van Der Meulen J, Engle S, Kim J, et al. 1997. J. Gerontol.

    52:B3947

    46. Masoro EJ. 1995. InHandbook of Physiology, ed. EJ Masoro, pp. 55590. New York: OxforUniv. Press47. Wulf HC, Sandby-Mller J, Kobayasi T, Gniadecki R. 2004. Micron 35:18591

    48. Fu C, Hickey M, Morrison M, McCarter R, Han ES. 2006.Mech. Ageing Dev. 127:90549. Beckman KB, Ames BN. 1998. Physiol. Rev. 78:54781

    50. Butler RN, Sprott R, Warner H, Bland J, Feuers R, et al. 2004. J. Gerontol. A 59:B560651. Campisi J. 2005. Cell120:51322

    52. Slawik M, Vidal-Puig AJ. 2006. Ageing Res. Rev. 5:1446453. Sulston JE. 1983. Cold Spring Harb. Symp. Quant. Biol. 48:44352

    54. Micchelli CA, Perrimon N. 2006. Nature 439:4757955. Ohlstein B, Spradling A. 2006. Nature 439:47074

    56. Ohlstein B, Spradling A. 2007. Science 315:9889257. Rando TA. 2006. Nature 441:10808658. Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, et al. 1998. Na

    Med. 4:13131759. Kuhn HG, Dickinson-Anson H, Gage FH. 1996. J. Neurosci. 16:202733

    60. Van Zant G, Liang Y. 2003. Exp. Hematol. 31:6597261. Fuller MT, Spradling AC. 2007. Science 316:4024

    62. Herbig U, Ferreira M, Condel L, Carey D, Sedivy JM. 2006. Science 311:125763. Herbig U, Sedivy JM. 2006. Mech. Ageing Dev. 127:1624

    64. Dimri GP, Lee X, Basile G, Acosta M, Scott G, et al. 1995. Proc. Natl. Acad. Sci. US92:936367

    65. Kirkwood TBL. 2004. Mech. Ageing Dev. 125:9111566. Hornsby PJ. 2002. Ageing Res. Rev. 1:22942

    67. Untergasser G, Madersbacher S, Berger P. 2005. Exp. Gerontol. 40:12128

    68. Franceschi C, Capri M, Monti D, Giunta S, Olivieri F, et al. 2007. Mech. Ageing De128:92105

    69. Finch CE, Crimmins EM. 2004. Science 305:17363970. Crimmins EM, Finch CE. 2006. Proc. Natl. Acad. Sci. USA 103:498503

    71. Aoshiba K, Nagai A. 2007. FEBS Lett. 581:35121672. Zheng F, Cheng QL, Plati AR, Ye SQ, Berho M, et al. 2004. Am. J. Pathol. 165:17899

    73. Bahar R, Hartmann CH, Rodriguez KA, Denny AD, Busuttil RA, et al. 2006. Natu441:101114

    74. Dolle ME, Snyder WK, Gossen JA, Lohman PH, Vijg J. 2000. Proc. Natl. Acad. Sci. US97:84038

    75. Vijg J. 2004. Mech. Ageing Dev. 125:74753

    76. Busuttil R, Bahar R, Vijg J. 2007. Neuroscience 145:13414777. Chambers SM, Shaw CA, Gatza C, Fisk CJ, Donehower LA, Goodell MA. 2007. PLo

    Biol. 5:e20178. Balaban RS, Nemoto S, Finkel T. 2005. Cell120:48395

    79. Harman D. 1956. J. Gerontol. 11:298300

    796 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    21/25

    80. Ahmed N, Thornalley PJ. 2007. Diabetes Obes. Metab. 9:2334581. Barja G. 2004. Trends Neurosci. 27:59560082. Ayala V, Naudi A, Sanz A, Caro P, Portero-Otin M, et al. 2007. J. Gerontol. A 2:3526083. Sohal RS, Mockett RJ, Orr WC. 2002. Free Rad. Biol. Med. 33:5758684. Hsu AL, Murphy CT, Kenyon C. 2003. Science 300:11424585. Walker GA, Lithgow GJ. 2003. Aging Cell2:1313986. Schriner SE, Linford NJ, Martin GM, Treuting P, Ogburn CE, et al. 2005. Science

    308:19091187. Melendez A, Talloczy Z, Seaman M, Eskelinen EL, Hall DH, Levine B. 2003. Science

    301:13879188. Vijg J. 2007. Aging of the Genome: The Dual Role of DNA in Life and Death . Oxford: Oxford

    Univ. Press89. Michikawa Y, Mazzucchelli F, Bresolin N, Scarlato G, Attardi G. 1999. Science 286:774

    7990. Vermulst M, Bielas JH, Kujoth GC, Ladiges WC, Rabinovitch PS, et al. 2007.Nat. Genet.

    39:5404391. Lee CK, Klopp RG, Weindruch R, Prolla TA. 1999. Science 285:13909392. Werner T. 2007. Mech. Ageing Dev. 128:1687293. Boehm M, Slack FJ. 2006. Cell Cycle 5:83740

    94. Han SN, Adolfsson O, Lee CK, Prolla TA, Ordovas J, Meydani SN. 2006. J. Immunol.177:605261

    95. Lener T, Moll PR, Rinnerthaler M, Bauer J, Aberger F, Richter K. 2006. Exp. Gerontol.41:38797

    96. Droge W, Schipper HM. 2007. Aging Cell6:3617097. Kennedy BK, Steffen KK, Kaeberlein M. 2007. Cell Mol. Life Sci. 64:13232898. Kaeberlein M, Powers RW 3rd, Steffen KK, Westman EA, Hu D, et al. 2005. Science

    310:11939699. Kapahi P, Zid BM, Harper T, Koslover D, Sapin V, Benzer S. 2004. Curr. Biol. 14:88590

    100. Hansen M, Taubert S, Crawford D, Libina N, Lee SJ, Kenyon C. 2007. Aging Cell6:95110

    101. Clancy DJ, Gems D, Hafen E, Leevers SJ, Partridge L. 2002. Science 296:319102. Bonkowski MS, Rocha JS, Masternak MM, Al Regaiey KA, Bartke A. 2006. Proc. Natl.

    Acad. Sci. USA 103:79015103. Fabrizio P, Pozza F, Pletcher SD, Gendron CM, Longo VD. 2001. Science 292:28890104. PowersRW,Kaeberlein M, Caldwell SD, Kennedy BK,Fields S. 2006. GenesDev.20:174

    84105. Bonawitz ND, Chatenay-Lapointe M, Pan Y, Shadel GS. 2007. Cell Metab. 5:26577106. Giannakou ME, Partridge L. 2007. Trends Biochem. Sci. 32:18088107. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, et al. 2003.Nature 421:182

    87108. Bluher M, Kahn BB, Kahn CR. 2003. Science 299:57274

    109. Spindler SR, Dhahbi JM. 2007. Annu. Rev. Nutr. 27:193217110. McElwee JJ,Schuster E, Blanc E, Piper MD, Thomas JH, et al. 2007. Genome Biol. 8:R132111. Amador-Noguez D, Dean A, Huang W, Setchell K, Moore D, Darlington G. 2007.Aging

    Cell6:45370112. Sohal RS, Weindruch R. 1996. Science 273:5963113. Lopez-Torres M, Gredilla R, Sanz A, Barja G. 2002. Free Radic. Biol. Med. 32:88289114. Gong X, Shang F, Obin M, Palmer H, Scrofano MM, et al. 1997. Mech. Ageing Dev.

    99:18192

    www.annualreviews.org Control Theory Analysis of Aging 797

  • 7/31/2019 Envejecimiento Toward a Control Theory

    22/25

    115. Napoli C, Martin-Padura I, de Nigris F, Giorgio M, Mansueto G, et al. 2003. Proc. NaAcad. Sci. USA 100:211216

    116. Pinton P, Rimessi A, Marchi S, Orsini F, Migliaccio E, et al. 2007. Science 315:65963

    117. Navarro A, Boveris A. 2007. Am. J. Physiol. Cell. Physiol. 292:C67086118. McCarroll SA, Murphy CT, Zou S, Pletcher SD, Chin CS, et al. 2004. Nat. Gene

    36:197204

    119. Shigenaga MK, Hagen TM, Ames BN. 1994. Proc. Natl. Acad. Sci. USA 91:1977178

    120. Boffoli D, ScaccoSC,Vergari R, Solarino G, Santacroce G, Papa S. 1994. Biochim. BiophyActa 1226:7382121. Kujoth GC, Hiona A, Pugh TD, Someya S, Panzer K, et al. 2005. Science 309:48184

    122. Trifunovic A, Wredenberg A, Falkenberg M, Spelbrink JN, Rovio AT, et al. 2004. Natu429:41723

    123. Nicotera P, Ankarcrona M, Bonfoco E, Orrenius S, Lipton SA. 1997.Adv. Neurol. 72:95101

    124. Thompson CB. 1995. Science 267:145662125. Wyllie AH, Kerr JFR, Currie AR. 1980. Int. Rev. Cytol. 68:251306

    126. Von Zglinicki T, Martin-Ruiz CM, Saretzki G. 2005. Signal Transduct. 5:10314127. Passos JF, Saretzki G, Ahmed S, Nelson G, Richter T, et al. 2007. PLoS Biol. 5:11385

    128. Horvath MM, Wang X, Resnick MA, Bell DA. 2007. PLoS Genet. 3:e127129. Gatza C, Moore L, Dumble M, Donehower LA. 2007. Cell Cycle 6:5255130. VanRemmenH,IkenoY,HamiltonM,PahlavaniM,WolfN,etal.2004.Physiol. Genom

    16:2937131. Andziak B, OConnor TP, Qi W, Dewaal EM, Pierce A, et al. 2006. Aging Cell5:4637

    132. Kacser H, Burns JA. 1973. Symp. Soc. Exp. Biol. 27:65104133. Kacser H, Burns JA. 1979. Biochem. Soc. Trans. 7:114960

    134. Heinrich R, Rappoport TA. 1974. Eur. J. Biochem. 42:8995135. Fell D. 1996. Understanding the Control of Metabolism. London: Portland

    136. Fell DA. 1992. Biochem. J. 286:31330137. Brown GC. 1992. Biochem. J. 284:113

    138. Brand MD. 1997. J. Exp. Biol. 200:193202139. Brand MD. 1996. J. Theor. Biol. 182:35160

    798 Murphy Partridge

  • 7/31/2019 Envejecimiento Toward a Control Theory

    23/25

    Annual Review of

    Biochemistry

    Volume 77, 2008Contents

    Prefatory Chapters

    Discovery of G Protein Signaling

    Zvi Selinger p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p1

    Moments of Discovery

    Paul Berg p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 14

    Single-Molecule Theme

    In singulo Biochemistry: When Less Is More

    Carlos Bustamante p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 45

    Advances in Single-Molecule Fluorescence Methods

    for Molecular Biology

    Chirlmin Joo, Hamza Balci, Yuji Ishitsuka, Chittanon Buranachai,

    and Taekjip Ha p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 51

    How RNA Unfolds and Refolds

    Pan T.X. Li, Jeffrey Vieregg, and Ignacio Tinoco, Jr. p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 77

    Single-Molecule Studies of Protein FoldingAlessandro Borgia, Philip M. Williams, and Jane Clarke p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 101

    Structure and Mechanics of Membrane Proteins

    Andreas Engel and Hermann E. Gaub p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p127

    Single-Molecule Studies of RNA Polymerase: Motoring Along

    Kristina M. Herbert, William J. Greenleaf, and Steven M. Block p p p p p p p p p p p p p p p p p p p p149

    Translation at the Single-Molecule Level

    R. Andrew Marshall, Colin Echeverra Aitken, Magdalena Dorywalska,

    and Joseph D. Puglisi p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p177

    Recent Advances in Optical Tweezers

    Jeffrey R. Moffitt, Yann R. Chemla, Steven B. Smith, and Carlos Bustamante p p p p p p 205

    Recent Advances in Biochemistry

    Mechanism of Eukaryotic Homologous Recombination

    Joseph San Filippo, Patrick Sung, and Hannah Klein p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 229

    v

  • 7/31/2019 Envejecimiento Toward a Control Theory

    24/25

    Structural and Functional Relationships of the XPF/MUS81

    Family of Proteins

    Alberto Ciccia, Neil McDonald, and Stephen C. West p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 25

    Fat and Beyond: The Diverse Biology of PPAR

    Peter Tontonoz and Bruce M. Spiegelman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 28

    Eukaryotic DNA Ligases: Structural and Functional Insights

    Tom Ellenberger and Alan E. Tomkinsonp p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p

    31

    Structure and Energetics of the Hydrogen-Bonded Backbone

    in Protein Folding

    D. Wayne Bolen and George D. Rose p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 33

    Macromolecular Modeling with Rosetta

    Rhiju Das and David Baker p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 36

    Activity-Based Protein Profiling: From Enzyme Chemistry

    to Proteomic Chemistry

    Benjamin F. Cravatt, Aaron T. Wright, and John W. Kozarich p p p p p p p p p p p p p p p p p p p p p p 38

    Analyzing Protein Interaction Networks Using Structural Information

    Christina Kiel, Pedro Beltrao, and Luis Serrano p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 41

    Integrating Diverse Data for Structure Determination

    of Macromolecular Assemblies

    Frank Alber, Friedrich Frster, Dmitry Korkin, Maya Topf, and Andrej Sali p p p p p p p p 44

    From the Determination of Complex Reaction Mechanisms

    to Systems Biology

    John Ross p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 47

    Biochemistry and Physiology of Mammalian SecretedPhospholipases A2Gerard Lambeau and Michael H. Gelb p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p49

    Glycosyltransferases: Structures, Functions, and Mechanisms

    L.L. Lairson, B. Henrissat, G.J. Davies, and S.G. Withers p p p p p p p p p p p p p p p p p p p p p p p p p p p 52

    Structural Biology of the Tumor Suppressor p53

    Andreas C. Joerger and Alan R. Fersht p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p55

    Toward a Biomechanical Understanding of Whole Bacterial Cells

    Dylan M. Morris and Grant J. Jensen p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p58

    How Does Synaptotagmin Trigger Neurotransmitter Release?

    Edwin R. Chapman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p61

    Protein Translocation Across the Bacterial Cytoplasmic Membrane

    Arnold J.M. Driessen and Nico Nouwen p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 64

    vi C on te nt s

  • 7/31/2019 Envejecimiento Toward a Control Theory

    25/25

    Maturation of Iron-Sulfur Proteins in Eukaryotes: Mechanisms,

    Connected Processes, and Diseases

    Roland Lill and Ulrich Mhlenhoff p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 669

    CFTR Function and Prospects for Therapy

    John R. Riordan p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 701

    Aging and Survival: The Genetics of Life Span Extension

    by Dietary RestrictionWilliam Mair and Andrew Dillin p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 727

    Cellular Defenses against Superoxide and Hydrogen Peroxide

    James A. Imlay p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 755

    Toward a Control Theory Analysis of Aging

    Michael P. Murphy and Linda Partridge p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 777

    Indexes

    Cumulative Index of Contributing Authors, Volumes 7377p p p p p p p p p p p p p p p p p p p p p p p p

    799

    Cumulative Index of Chapter Titles, Volumes 7377 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 803

    Errata

    An online log of corrections to Annual Review of Biochemistry articles may be found

    at http://biochem.annualreviews.org/errata.shtml