A MITOCHONDRIAL PARADIGM OF METABOLIC AND...

51
A Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer: A Dawn for Evolutionary Medicine Douglas C. Wallace Center for Molecular and Mitochondrial Medicine and Genetics, Departments of Ecology and Evolutionary Biology, Biological Chemistry, and Pediatrics, University of California, Irvine, California 92697-3940; email: [email protected] Annu. Rev. Genet. 2005. 39:359–407 First published online as a Review in Advance on July 19, 2005 The Annual Review of Genetics is online at http://genet.annualreviews.org doi: 10.1146/ annurev.genet.39.110304.095751 Copyright c 2005 by Annual Reviews. All rights reserved 0066-4197/05/1215- 0359$20.00 Key Words mitochondria, reactive oxygen species, human origins, diabetes, neurodegenerative diseases, aging Abstract Life is the interplay between structure and energy, yet the role of energy deficiency in human disease has been poorly explored by modern medicine. Since the mitochondria use oxidative phospho- rylation (OXPHOS) to convert dietary calories into usable energy, generating reactive oxygen species (ROS) as a toxic by-product, I hypothesize that mitochondrial dysfunction plays a central role in a wide range of age-related disorders and various forms of cancer. Because mitochondrial DNA (mtDNA) is present in thousands of copies per cell and encodes essential genes for energy production, I propose that the delayed-onset and progressive course of the age- related diseases results from the accumulation of somatic mutations in the mtDNAs of post-mitotic tissues. The tissue-specific manifes- tations of these diseases may result from the varying energetic roles and needs of the different tissues. The variation in the individual and regional predisposition to degenerative diseases and cancer may re- sult from the interaction of modern dietary caloric intake and ancient mitochondrial genetic polymorphisms. Therefore the mitochondria provide a direct link between our environment and our genes and the mtDNA variants that permitted our forbears to energetically adapt to their ancestral homes are influencing our health today. 359 Annu. Rev. Genet. 2005.39:359-407. Downloaded from arjournals.annualreviews.org by Medical University of South Carolina - LIBRARIES & LEARNING RES CTR on 08/25/10. For personal use only.

Transcript of A MITOCHONDRIAL PARADIGM OF METABOLIC AND...

Page 1: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

A Mitochondrial Paradigmof Metabolic andDegenerative Diseases,Aging, and Cancer: A Dawnfor Evolutionary MedicineDouglas C. WallaceCenter for Molecular and Mitochondrial Medicine and Genetics, Departments ofEcology and Evolutionary Biology, Biological Chemistry, and Pediatrics,University of California, Irvine, California 92697-3940; email: [email protected]

Annu. Rev. Genet.2005. 39:359–407

First published online as aReview in Advance onJuly 19, 2005

The Annual Review ofGenetics is online athttp://genet.annualreviews.org

doi: 10.1146/annurev.genet.39.110304.095751

Copyright c© 2005 byAnnual Reviews. All rightsreserved

0066-4197/05/1215-0359$20.00

Key Words

mitochondria, reactive oxygen species, human origins, diabetes,neurodegenerative diseases, aging

AbstractLife is the interplay between structure and energy, yet the role ofenergy deficiency in human disease has been poorly explored bymodern medicine. Since the mitochondria use oxidative phospho-rylation (OXPHOS) to convert dietary calories into usable energy,generating reactive oxygen species (ROS) as a toxic by-product, Ihypothesize that mitochondrial dysfunction plays a central role ina wide range of age-related disorders and various forms of cancer.Because mitochondrial DNA (mtDNA) is present in thousands ofcopies per cell and encodes essential genes for energy production,I propose that the delayed-onset and progressive course of the age-related diseases results from the accumulation of somatic mutationsin the mtDNAs of post-mitotic tissues. The tissue-specific manifes-tations of these diseases may result from the varying energetic rolesand needs of the different tissues. The variation in the individual andregional predisposition to degenerative diseases and cancer may re-sult from the interaction of modern dietary caloric intake and ancientmitochondrial genetic polymorphisms. Therefore the mitochondriaprovide a direct link between our environment and our genes and themtDNA variants that permitted our forbears to energetically adaptto their ancestral homes are influencing our health today.

359

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 2: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Contents

LEAN DIETS, LONG LIFE, ANDTHE MITOCHONDRIA. . . . . . . . 360

MITOCHONDRIAL BIOGENESISAND BIOENERGETICS: ANAGING PARADIGM . . . . . . . . . . . . 361

PATHOGENIC mtDNAMUTATIONS . . . . . . . . . . . . . . . . . . 368Pathogenic mtDNA

Rearrangement Mutations . . . . . 368Mitochondrial Defects

in Diabetes . . . . . . . . . . . . . . . . . . . . 369ANCIENT MIGRATIONS AND

CLIMATIC ADAPTATION . . . . . 374ADAPTIVE mtDNA MUTATIONS

IN LONGEVITY ANDDEGENERATIVE DISEASES . . . 378

THE MITOCHONDRIALEITIOLOGY OF AGING . . . . . . . 379

THE MITOCHONDRIALETIOLOGY OFDEGENERATIVE DISEASES . . . 383

SOMATIC AND GERMLINEmtDNA MUTATIONSIN CANCER . . . . . . . . . . . . . . . . . . . . 386Mitochondrial Medicine: An

Integrated MitochondrialModel for the Etiology ofDegenerative Diseases, Aging,and Cancer . . . . . . . . . . . . . . . . . . . 389

Dietary Restriction, DegenerativeDiseases, Longevity, andClimatic Adaptation . . . . . . . . . . . 389

The Mitochondrial Energetics andROS Biology of Cancer . . . . . . . . 391

MITOCHONDRIALADAPTATION ANDEVOLUTIONARY MEDICINE . 392The Interplay between

Environment and Energetics . . . 392CONCLUSION . . . . . . . . . . . . . . . . . . . . 393

LEAN DIETS, LONG LIFE, ANDTHE MITOCHONDRIA

Age-related metabolic and degenerative dis-eases are increasing to epidemic proportionsin all industrialized countries. Diabetes has in-creased sharply over the past century, with aworldwide incidence in 2000 of 151 millioncases and the projected incidence for 2010 of221 million cases (for the United States, 14.2million in 2000 versus 17.5 million by 2010)(73, 257). Today, Alzheimer disease (AD) af-fects about 4.5 million Americans, projectedto increase to 11 to 16 million by 2050 (6,70). Parkinson’s disease (PD) afflicts approx-imately one million persons in the UnitedStates today; 60,000 new cases are diagnosedevery year, and the incidence is projected toquadruple by 2040 (160, 164). The risk ofcancer also increases with age. Prostate can-cer is the most common malignancy amongmen in industrialized countries. In the UnitedStates in 2004, approximately 230,000 newcases were reported, with an annual deathtoll of approximately 30,000 men (31). More-over, the incidence of prostate cancer has beenincreasing steadily in the United States andCanada over the past 30 years (132).

As many of these age-related diseases showsome familial association, a genetic basis hasbeen assumed. Yet, the nature and frequencyof genetic variants in the human popula-tion has not changed significantly in the past50 years, even though the incidence of thesediseases has climbed continuously. Therefore,it isn’t our genes that have changed; it is ourenvironment, and the biggest environmentalchange that we have experienced is in our diet.

For the first time in human history, indi-viduals can live their entire lives free fromhunger. Yet, it has been known for over 70years that laboratory rodents, if maintainedon a restricted calorie diet, will be health-ier, more active, more intelligent, and will live

360 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 3: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

longer and have fewer cancers (69, 126, 127,130, 204).

So what is the link between genetics anddiet for these diseases? Prodigious effort hasbeen expended to identify chromosomal ge-netic loci that are responsible for these prob-lems, but with disappointingly little success.One hint as to why nuclear DNA (nDNA)analysis has been so unfruitful is that all ofthese diseases have a delayed onset and a pro-gressive course. Thus while the most impor-tant risk factor for contracting all of these dis-eases is age, nothing in Mendelian geneticscan provide the needed aging clock. The agingclock must involve the gradual accumulationof sequential changes in thousands of copiesof the same gene or genes, yet each Mendeliangene is present in only two copies.

Furthermore, certain age-related diseasesare preferentially found in distinct popula-tions, suggesting that they are influenced byregional genetic differences. Yet most ancientnDNA polymorphisms are common to allglobal populations. Only one human geneticsystem is known to be present in thousands ofcopies per cell, to exhibit striking regional ge-netic variation, and to be directly involved incalorie utilization: the mitochondrial geneticsystem.

The mitochondria are ancient bacterialsymbionts with their own mitochondrialDNA (mtDNA), RNA, and protein synthe-sizing systems. Each human cell containshundreds of mitochondria and thousands ofmtDNAs. The mtDNA is maternally inher-ited and shows striking regional genetic vari-ation. This regional variation was a majorfactor in permitting humans to adapt to thedifferent global environments they encoun-tered and mastered. Moreover, the mitochon-dria burn the calories in our diet with theoxygen that we breathe to make chemical en-ergy to do work and heat to maintain ourbody temperature. As a by-product of en-ergy production, the mitochondria also gen-erate most of the endogenous reactive oxygenspecies (ROS) of the cell, and these damagethe mitochondria, mtDNAs, and cell. Hence,

mtDNA:mitochondrial DNA

Mitochondrion (s),mitochondria (pl):cellular organelle ofendosymbiotic originthat resides in thecytosol of mostnucleated(eukaryotic) cells andwhich producesenergy by oxidizingorganic acids and fatswith oxygen by theprocess of oxidativephosphorylation(OXPHOS) andgenerates oxygenradicals (reactiveoxygen species,ROS) as a toxicby-product

ROS: reactiveoxygen species,oxygen radicals

OXPHOS:oxidativephosphorylation

MitochondrialDNA (mtDNA):the portion of themitochondrialgenome thatcurrently resides inthe matrix of themitochondrion, as acircular DNAmolecule containingthe mitochondrialrRNA genes, tRNAgenes, and 13subunits of themitochondrialoxidativephosphorylation(OXPHOS) enzymecomplexes

the mitochondria are the only human geneticsystem that embodies the features necessaryto explain the observed characteristics of thecommon age-related diseases (237).

In this review, I make the case that mi-tochondrial decline and mtDNA damage arecentral to the etiology of the age-relatedmetabolic and degenerative diseases, aging,and cancer. This is because the rate of mi-tochondrial and mtDNA damage and thusdecline is modulated by the extent of mi-tochondrial oxidative stress. MitochondrialROS production, in turn, is increased by theavailability of excess calories, modulated byregional mtDNA genetic variation, and reg-ulated by alterations in nDNA expression ofstress response genes.

MITOCHONDRIAL BIOGENESISAND BIOENERGETICS: ANAGING PARADIGM

The proto-mitochondrion entered the primi-tive eukaryotic cell between two and three bil-lion years ago. Initially, the bacterial genomeencoded all of the genes necessary for a free-living organism. However, as the symbiosismatured, many bacterial genes were trans-ferred to extrabacterial plasmids (chromo-somes), such that today the maternally in-herited mtDNA retains only the genes forthe 12S and 16S rRNAs and the 22 tRNAsrequired for mitochondrial protein synthesisplus 13 polypeptides of the mitochondrial en-ergy generating process, oxidative phosphory-lation (OXPHOS) (Figure 1). The remaining∼1500 genes of the mitochondrial genome arenow scattered throughout the chromosomalDNA. These nDNA-encoded mitochondrialproteins are translated on cytosolic ribosomesand selectively imported into the mitochon-drion through various mitochondrial proteinimport systems. For example, certain proteinsdestined for the mitochondrial matrix are syn-thesized with an amino terminal, positivelycharged, amphoteric targeting peptide that iscleaved off once the protein enters the mito-chondrial matrix (237).

www.annualreviews.org • Mitochondrial & Age-Related Diseases 361

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 4: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Figure 1Human mitochondrial DNA map showing representative pathogenic and adaptive base substitutionmutations. D-loop = control region (CR). Letters around the outside perimeter indicate cognate aminoacids of the tRNA genes. Other gene symbols are defined in the text. Arrows followed by continentalnames and associated letters on the inside of the circle indicate the position of defining polymorphismsof selected region-specific mtDNA lineages. Arrows associated with abbreviations followed by numbersaround the outside of the circle indicate representative pathogenic mutations, the number being thenucleotide position of the mutation. Abbreviations: DEAF, deafness; MELAS, mitochondrialencephalomyopathy, lactic acidosis and stroke-like episodes; LHON, Leber hereditary optic neuropathy;ADPD, Alzheimer and Parkinson disease; MERRF, myoclonic epilepsy and ragged red fiber disease;NARP, neurogenic muscle weakness, ataxia, retinitis pigmentosum; LDYS, LHON + dystonia;PC, prostate cancer.

The 13 mtDNA-encoded polypeptidegenes are translated on mitochondrial ri-bosomes and all are structural subunits ofOXPHOS enzyme complexes. These include7 (ND1, 2, 3, 4L, 4, 5, 6) of the 46 polypep-

tides of complex I (NADH dehydrogenase),one (cytochrome b, cytb) of the 11 polypep-tides of complex III (bc1 complex), 3 (COI,II, III) of the 13 polypeptides of complexIV (cytochrome c oxidase), and 2 (ATP 6 &

362 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 5: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

8) of the 16 proteins of complex V (ATPsynthetase) (Figure 1). The nDNA codesfor all other mitochondrial proteins includ-ing all four subunits of complex II (succinatedehydrogenase), the mitochondrial DNApolymerase γ (POLG) subunits, the mito-chondrial RNA polymerase components, themitochondrial transcription factor (mtTFA),the mitochondrial ribosomal proteins andelongation factors, and the mitochondrialmetabolic enzymes.

The organization of the mtDNA is uniquein that its structural genes have no 5′ or 3′ non-coding sequences, no introns, and no spacersbetween the genes. The mtDNA is symmetri-cally transcribed from two promoters, one forthe G-rich heavy (H) stand and the other forthe C-rich light (L) strand. These H- and L-strand promoters (PH and PL) are containedin the approximately 1121-np control region(CR) that encompasses four mtTFA bindingsites, the H-strand origin of replication (OH),and three conserved sequence blocks (CSB I,II, and III). The L-strand origin of replica-tion (OL) is two thirds of the way around themtDNA from OH. Transcription is initiated atPH or PL and progresses around the mtDNA,generating a polycistronic message. ThetRNAs, which punctuate the genes, arecleaved out the mRNAs are then polyadeny-lated (237) (Figure 1).

The mtDNA gene repertoire has remainedrelatively constant since the formation of thefungal-animal lineage. This is because themtDNA genetic code began to drift in thefungi such that the mtDNA genes can nolonger be interpreted by the nuclear-cytosolsystem (233). Consequently, when a mtDNAsequence is transferred to the nDNA, it re-mains as a nonfunctional pseudogene (147).

The mitochondria generate energy by oxi-dizing hydrogen derived from our dietary car-bohydrates (TCA cycle) and fats (β-oxidation)with oxygen to generate heat and ATP(Figure 2). Two electrons donated fromNADH + H+ to complex I (NADH dehy-drogenase) or from succinate to complex II(succinate dehydrogenase, SDH) are passed

CR: mtDNAcontrol region

TCA:mitochondrialtricarboxylic acidcycle

SDH: succinatedehydrogenase

COX: cytochrome coxidase, complex IV

ETC:mitochondrialelectron transportchain, a part of theOXPHOS system

ANT: adeninenucleotidetranslocator

Unc 1,2,3:uncoupling proteins1,2,3

Reactive oxygenspecies (ROS):primarily superoxideanion (O•−

2 ),hydrogen peroxide(H2O2), andhydroxyl radical(•OH), commonlyreferred to as oxygenradicals; generated asa toxic by-product ofoxidative energyproduction byOXPHO; whichdamage themitochondrial andcellular DNA,proteins, lipids, andother moleculescausing oxidativestress

sequentially to ubiquinone (coenzyme Q orCoQ) to give ubisemiquinone (CoQH•) andthen ubiquinol (CoQH2). Ubiquinol trans-fers its electrons to complex III (ubiquinol:cytochrome c oxidoreductase), which trans-fers them to cytochrome c. From cytochromec, the electrons flow to complex IV (cy-tochrome c oxidase, COX) and finally to 1/2O2

to give H2O. Each of these electron transportchain (ETC) complexes incorporates multi-ple electron carriers. Complexes I, II, and IIIencompass several iron-sulfur (Fe-S) centers,whereas complexes III and IV encompass theb + c1 and a + a3 cytochromes, respectively.The mitochondrial TCA cycle enzyme aconi-tase is also an iron-sulfur center protein (234,235, 237).

The energy released by the flow of elec-trons through the ETC is used to pump pro-tons out of the mitochondrial inner mem-brane through complexes I, III, and IV. Thiscreates a capacitance across the mitochondrialinner membrane, the electrochemical gradi-ent (�P = �� + �pH). The potential en-ergy stored in �P is coupled to ATP synthesisby complex V (ATP synthase). As protonsflow back into the matrix through a pro-ton channel in complex V, ADP and Pi arebound, condensed, and released as ATP. Ma-trix ATP is then exchanged for cytosolic ADPby the adenine nucleotide translocator (ANT)(Figure 2).

Because the ETC is coupled to ATPsynthesis through �P, mitochondrial oxygenconsumption rate is regulated by the matrixconcentration of ADP. In the absence of ADP,the consumption of oxygen is regulated byproton leakage across the inner membraneand thus is slow (state IV respiration). How-ever, when ADP is added, it binds to theATP synthase and is rapidly converted intoATP at the expense of �P. As protons flowthrough the ATP synthase proton channel,the proton gradient is depolarized. Stored fatsand carbohydrates are then mobilized to pro-vide electrons to the ETC, which reduce oxy-gen to water and pump the protons back outof the mitochondrial matrix. The resulting

www.annualreviews.org • Mitochondrial & Age-Related Diseases 363

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 6: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Figure 2Diagram showing the relationships of mitochondrial oxidative phosphorylation (OXPHOS) to(a) energy (ATP) production, (b) reactive oxygen species (ROS) production, and (c) initiation ofapoptosis through the mitochondrial permeability transition pore (mtPTP). The OXPHOS complexes,designated I to V, are complex I (NADH: ubiquinone oxidoreductase) encompassing a FMN (flavinmononucleotide) and six Fe-S centers (designated with a cube); complex II (succinate: ubiquinoneoxidoreductase) involving a FAD (flavin adenine dinucleotide), three Fe-S centers, and a cytochrome b;complex III (ubiquinol: cytochrome c oxidoreductase) encompassing cytochromes b, c1 and the RieskeFe-S center; complex IV (cytochrome c oxidase) encompassing cytochromes a + a3 and CuA and CuB;and complex V (H+-translocating ATP synthase). Pyruvate from glucose enters the mitochondria viapyruvate dehydrogenase (PDH), generating acetylCoA, which enters the TCA cycle by combining withoxaloacetate (OAA). Cis-aconitase converts citrate to isocitrate and contains a 4Fe-4S center. Lactatedehydrogenase (LDH) converts excess pyruvate plus NADH to lactate. Small molecules defuse throughthe outer membrane via the voltage-dependent anion channel (VDAC) or porin. The VDAC togetherwith ANT, Bax, and the cyclophilin D (CD) protein are thought to come together at the mitochondrialinner and outer membrane contact points to create the mtPTP. The mtPTP interacts with thepro-apoptotic Bax, anti-apoptotic Bcl2 and the benzodiazepine receptor (BD). The opening of themtPTP is associated with the release of several pro-apoptotic proteins. Cytochrome c (cytc) interactswith and activates cytosolic Apaf-1, which then binds to and activates procaspase-9. The activatedcaspase-9 then initiates the proteolytic degradation of cellular proteins. Apoptosis initiating factor (AIF)and endonuclease G (EndoG) have nuclear targeting peptides that are transported to the nucleus anddegrade the chromosomal DNA. Modified from Reference (237).

364 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 7: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

increased oxygen consumption on addition ofADP is known as state III respiration. �P isalso used for the import of cytosolic proteins,substrates, and Ca2+ into the mitochondrion.Drugs such as 2,4-dinitrophenol (DNP) andnDNA-encoded uncoupler proteins 1, 2, and3 (Unc1, 2, and 3) render the mitochon-drial inner membrane leaky for protons. Thisshort-circuits the capacitor and “uncouples”electron transport from ATP synthesis. Thiscauses the ETC to run at its maximum rate,causing maximum oxygen consumption andheat production, but diminished ATP gener-ation (Figure 2).

The efficiency by which dietary caloriesare converted to ATP is determined by thecoupling efficiency of OXPHOS. If the ETCis highly efficient at pumping protons out ofthe mitochondrial inner membrane and theATP synthesis is highly efficient at convert-ing the proton flow through its proton chan-nel into ATP, then the mitochondria will gen-erate the maximum ATP and the minimumheat per calorie consumed. These mitochon-dria are said to be tightly coupled. By contrast,if the efficiency of proton pumping is reducedand/or more protons are required to makeeach ATP by the ATP synthase, then eachcalorie burned will yield less ATP but moreheat. Such mitochondria are said to be looselycoupled. Therefore, in an endothermic ani-mal, the coupling efficiency determines theproportion of calories utilized by the mito-chondrion to perform work versus those tomaintain body temperature.

As a toxic by-product of OXPHOS, themitochondria generate most of the endoge-nous ROS. ROS production is increased whenthe electron carriers in the initial steps of theETC harbor excess electrons, i.e., remain re-duced, which can result from either inhibitionof OXPHOS or from excessive calorie con-sumption. Electrons residing in the electroncarriers; for example, the unpaired electronof ubisemiquinone bound to the CoQ bind-ing sites of complexes I, II, and III; can bedonated directly to O2 to generate superoxideanion (O•−

2 ). Superoxide O•−2 is converted to

Oxidativephosphorylation(OXPHOS): theprocess by which themitochondriongenerates energythrough oxidation oforganic acids and fatswith oxygen to createa capacitor [electronchemical gradient(�P = �� +�pH)] across themitochondrial innermembrane. This �Pis used as a source ofpotential energy togenerate adenosinetriphosphate (ATP),transport substratesor ions, or produceheat. OXPHOSencompasses fivemultipolypepetidecomplexes I, II, III,IV and V. Complex Iis NADHdehydrogenase orNADH:ubiquinoneoxidoreductase,complex II issuccinatedehydrogenase(SDH) orsuccinate:ubiquinoneoxidoreductase,complex III is thebc1 complex orubiquinole:cytochrome coxidoreductase,complex IV iscytochrome coxidase (COX) orreduced cytochromec: oxygenoxidoreductase, andcomplex V is theATP synthase orproton-translocatingATP synthase.Complexes I, III, IV,and V encompassboth nDNA- andmtDNA-encodedsubunits

H2O2 by mitochondrial matrix enzyme Mnsuperoxide dismutase (MnSOD, Sod2) or bythe Cu/ZnSOD (Sod1), which is located inboth the mitochondrial intermembrane spaceand the cytosol. Import of Cu/ZnSOD intothe mitochondrial intermembrane space oc-curs via the apoprotein, which is metallatedupon entrance into the intermembrane spaceby the CCS metallochaperone (166, 207).H2O2 is more stable than O•−

2 and can dif-fuse out of the mitochondrion and into the cy-tosol and the nucleus. H2O2 can be convertedto water by mitochondrial and cytosolic glu-tathione peroxidase (GPx1) or by peroxisomalcatalase. However, H2O2, in the presence ofreduced transition metals, can be convertedto the highly reactive hydroxyl radical (•OH)(Figure 2). Iron-sulfur centers in mitochon-drial enzymes are particularly sensitive toROS inactivation. Hence, the mitochondriaare the prime target for cellular oxidative dam-age (241, 242).

For tightly coupled mitochondria, in thepresence of excess calories and in the ab-sence of exercise, the electrochemical gra-dient (�P) becomes hyperpolarization andthe ETC chain stalls. This is because with-out ADP the ATP synthase stops turningover, thus blocking the flow of protons backacross the mitochondrial inner membranethrough the proton channel of the ATP syn-thase. However, the ETC will continue todraw on the excess calories for electrons topump protons out of the mitochondrial innermembrane until the electrostatic potential of�P inhibits further proton pumping. At thispoint, the ETC stalls and the electron car-riers become maximally occupied with elec-trons (maximally reduced). These electrons(reducing equivalents) can then be transferredto O2 to generate O2 and thus increased ROSand oxidative stress.

By contrast, in individuals who actively ex-ercise and generate ADP, the ATP synthasekeeps �P hypopolarized and electrons con-tinue to flow through the ETC to sustain �P.Consequently, the electron carriers retain fewelectrons (remain oxidized). The low electron

www.annualreviews.org • Mitochondrial & Age-Related Diseases 365

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 8: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

MnSOD (Sod2):mitochondrial matrixsuperoxide dismutase

Cu/ZnSOD (Sod1):mitochondrialintermembranespace and cytosolicsuperoxide dismutase

mtPTP:mitochondrialpermeabilitytransition pore

Apoptosis: aprocess ofprogrammed celldeath resulting in theactivation of caspaseenzymes andintracellularnucleases thatdegrade the cellularproteins and nDNA.Apoptosis can beinitiated via themitochondrionthrough theactivation of themitochondrialpermeabilitytransition pore(mtPTP) in responseto energy deficiency,increased oxidativestress, excessiveCa2+, and/or otherfactors

density of the ETC electron carriers limitsROS production and reduces oxidative stress.

This same effect can be achieved if the mi-tochondria become partially uncoupled, ei-ther by decreasing the number of protonspumped per electron pair oxidized or by per-mitting protons to flow back through theinner membrane without making ATP. Un-coupling can be achieved by disconnectingelectron transport from proton pumping byalterations of complexes I, III, or IV; by in-creasing the number of protons required bythe ATP synthase to make an ATP; or by ex-pression of an alternative proton channel suchas an uncoupling protein (UCP).

The mitochondria are also the major reg-ulators of apoptosis, accomplished via themitochondrial permeability transition pore(mtPTP). The mtPTP is thought to be com-posed of the inner membrane ANT, the outermembrane voltage-dependent anion channel(VDAC) or porin, Bax, Bcl2, and cyclophilinD. The outer membrane channel is thoughtto be VDAC, but the identity of the innermembrane channel is unclear since elimina-tion of the ANTs does not block the chan-nel (98). The ANT performs a key regulatoryrole for the mtPTP (98). When the mtPTPopens, �P collapses and ions equilibrate be-tween the matrix and cytosol, causing the mi-tochondria to swell. Ultimately, this resultsin the release of the contents of the mito-chondrial intermembrane space into the cy-tosol. The released proteins include a numberof cell death-promoting factors including cy-tochrome c, AIF, latent forms of caspases (pos-sibly procaspases-2, 3, and 9), SMAD/Diablo,endonuclease G, and the Omi/HtrA2 serineprotease 24. On release, cytochrome c ac-tivates the cytosolic Apaf-1, which activatesthe procaspase-9. Caspase 9 then initiates aproteolytic cascade that destroys the proteinsof the cytoplasm. Endonuclease G and AIFare transported to the nucleus, where theydegrade the chromatin. The mtPTP can bestimulated to open by the mitochondrial up-take of excessive Ca2+, by increased oxidativestress, or by deceased mitochondrial �P, ADP,

and ATP. Thus, disease states that inhibitOXPHOS and increase ROS production in-crease the propensity for mtPTP activationand cell death by apoptosis (Figure 2) (241,242).

The mtDNA is maternally inherited (63)and semi-autonomous. The genetic inde-pendence of the mtDNA has been demon-strated by enucleating cells harboring a pu-tative mtDNA marker, such as resistance tothe mitochondrial ribosome inhibitor chlo-ramphenicol (CAPR), and fusing the cytoplas-mic fragments containing the donor mito-chondria and mtDNA to recipient cells thatlack the genetic marker, e.g., CAPS recipientcells (26, 240). The resulting transmitochon-drial cybrids have the mtDNA of the donorand thus inherit the donor’s mitochondrialphenotype (CAPR), yet they have the nDNAof the recipient, demonstrating that mtDNAinheritance can be independent of nDNAinheritance.

The mtDNAs of a donor cell can morecompletely repopulate the recipient cells ifthe recipient cells are depleted, or cured, oftheir resident mtDNAs. The recipient cellscan be depleted of their mtDNA by treatmentwith the mitochondrial poison rhodamine-6-G (220) or they can be cured of theirmtDNAs by previous selection in ethidiumbromide plus glucose, pyruvate and uri-dine, resulting in cells that are permanentlymtDNA-deficient (ρ0 cells) (94).

The mtDNA has a very high mutation rate,presumably due to its chronic exposure to mi-tochondrial ROS. When a new mtDNA mu-tation arises in a cell, a mixed intracellularpopulation of mtDNAs is generated, a stateknown as heteroplasmy. As the heteroplasmiccell undergoes cytokinesis, the mutant andnormal molecules are randomly distributedinto the daughter cells. As a consequence ofthis replicative segregation, the proportion ofmutant and normal mtDNAs can drift to-ward homoplasmic mutant or wild type. Fur-thermore, in post-mitotic tissues, mtDNAsharboring deleterious mutations have beenfound to be preferentially, clonally, amplified

366 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 9: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

within cells. This may be a consequence ofthe nucleus attempting to compensate foran energy deficiency by making more mito-chondria. Therefore, cells with defective mi-tochondria are preferentially stimulated toreplicate their mitochondria and mtDNAs.As the proliferating mitochondria are turnedover, presumably by autophagy (2, 116, 136),the mutant mtDNA can become enriched insome cells by genetic drift. As the percentageof mutant mtDNAs increases, the mitochon-drial energetic output declines, ROS produc-tion increases, and the propensity for apop-tosis increases. As cells are progressively lostthrough apoptosis, tissue function declines,ultimately leading to symptoms. Thus the ac-cumulation of mutant mtDNA creates the ag-ing clock (241, 242) (Figure 3).

Clinical symptoms appear when the num-ber of cells in a tissue declines below the min-

imum necessary to maintain function. Thetime when this clinical threshold is reachedis related to the rate at which mitochondrialand mtDNA damage accumulates within thecells, leading to activation of the mtPTP andcell death, and to the number of cells presentin the tissue at birth in excess of the min-imum required for normal tissue function.Given that the primary factor determining cellmetabolism and tissue structure is reproduc-tive success, it follows that each tissue musthave sufficient extra cells at birth to make itlikely that that tissue will remain functionaluntil the end of the human reproductive pe-riod, or about 50 years. If the mitochondrialROS production rate increases, the rate ofcell loss will also increase, resulting in earlytissue failure and age-related disease. How-ever, if mitochondrial ROS production is re-duced, then the tissue cells will last longer and

Figure 3Mitochondrial and cellular model of aging. The upper line of cells diagrams the mitochondrial role in theenergetic life and death of a cell. The bottom diagram represents the loss of cells in a tissue over the lifeof an individual through mitochondrial-mediated death, black cells. The minimum number of cells forthe tissue to function normally is indicated by the dashed line.

www.annualreviews.org • Mitochondrial & Age-Related Diseases 367

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 10: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

CPEO/KSS:chronic progressiveexternalophthalmoplegia/Kearn-Sayresyndrome

NARP: neurogenicmuscle weakness,ataxia, and retinitispigmentosa

LHON: Leber’shereditary opticneuropathy

age-related symptoms will be deferred (236,238, 241) (Figure 3).

PATHOGENIC mtDNAMUTATIONS

The first evidence that mtDNA mutationsmight be a key factor in aging and age-related degenerative diseases came with theidentification of systemic diseases caused bymtDNA mutations. Pathogenic mtDNA mu-tations fall into three categories: rearrange-ment mutations (74), polypeptide gene mis-sense mutations (243), and protein synthe-sis (rRNA and tRNA) gene mutations (200,244). As the number of mtDNA-associateddiseases identified increased, it became clearthat mitochondrial diseases commonly havea delayed onset and progressive course andthat they result in the same clinical prob-lems as observed in age-related diseases and inthe elderly. Clinical manifestations that havebeen linked to mtDNA mutations affect thebrain, heart, skeletal muscle, kidney, and en-docrine system, the same tissues affected inaging. Specific symptoms include forms ofblindness, deafness, movement disorders, de-mentias, cardiovascular disease, muscle weak-ness, renal dysfunction, and endocrine disor-ders including diabetes (51, 149, 237, 241,242).

Pathogenic mtDNA RearrangementMutations

Systemically distributed mtDNA rearrange-ment mutations can be either inherited orspontaneous. Inherited mtDNA rearrange-ment mutations are primarily insertions. Thefirst inherited insertion mutation to be identi-fied caused maternally inherited diabetes anddeafness (13, 14).

Spontaneous rearrangement mutations,primarily deletions, generally result in a re-lated spectrum of symptoms, irrespective ofthe position of the deletion end points. Thisis because virtually all deletions remove atleast one tRNA and thus inhibit protein syn-

thesis (154). Thus the nature and severity ofthe symptoms from mtDNA deletion rear-rangements is not a consequence of the natureof the rearrangement, but rather of the tis-sue distribution of the rearranged mtDNAs.Rearrangements that are widely disseminatedprevent bone marrow stem cells from prolifer-ating and lead to the frequently lethal child-hood pancytopenia, known as Pearson mar-row pancreas syndrome. Widely distributedmtDNA rearrangements that spare the bonemarrow result in mitochondrial myopathywith ragged red fibers (RRF). Mitochon-drial myopathy is frequently associated withophthalmoplegia and ptosis, which is re-ferred to as chronic progressive external oph-thalmoplegia (CPEO). More severe, earlier-onset cases with multisystem involvementare known as the Kearns-Sayre syndrome(KSS) (241).

Pathogenic mtDNA missense mutations.Missense mutations in mtDNA polypeptidegenes can also result in an array of clinicalmanifestations. A mutation in the mtDNAATP6 gene at np 8993 (T to G) is associatedwith neurogenic muscle weakness, ataxia, andretinitus pigmentosum (NARP) when presentat lower percentages of mutant (75) and lethalchildhood Leigh syndrome when present athigher percentages of mutant (213). This mu-tation causes a marked inability of the ATPsynthase to utilize the electrochemical gra-dient to make ATP (219) and an associatedincrease in mitochondrial ROS production(128).

Missense and nonsense mutations in thecytb gene have been increasingly linked toprogressive muscle weakness (8, 52). Rarenonsense or frameshift mutants in COI havebeen associated with encephalomyopathies(25, 37).

Missense mutations in complex I geneshave been linked to Leigh syndrome (205),generalized dystonia and deafness (88), and toLeber hereditary optic neuropathy (LHON),a form of midlife, sudden-onset blindness(22, 23, 243). Surprisingly, the phenotype of

368 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 11: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

LHON can be caused by mutations in a num-ber of ND genes that change amino acids, withvery different amino acid conservation result-ing in varying severities of complex I defects(22). This anomaly has been explained by thediscovery that the mildest LHON mtDNAmutations, particularly those in ND6 at np14,484 and ND4L at np 10,663, are usuallyfound on a particular mtDNA background,the European mtDNA lineage J (21, 23). Lin-eage J mtDNAs have been found to harbormtDNA missense mutations that partially un-couple OXPHOS, thus exacerbating the ATPdefect of the pathogenic mutations (194).

Pathogenic mtDNA proteins synthesismutations. Base substitutions in mtDNAprotein synthesis genes can also result inmultisystem disorders with a wide range ofsymptoms, including mitochondrial myopa-thy, cardiomyopathy, deafness, mood disor-ders, movement disorders, dementia, dia-betes, intestinal dysmotility, etc. As with theATP6 np 8993 (T > G) mutation, studies onthe tRNALys np 8344 A to G mutation associ-ated with myoclonic epilepsy and ragged redfiber (MERRF) disease have revealed that thelevel of mutant heteroplasmy plus the age ofthe patient influence the severity of the clini-cal symptoms (200, 244).

Perhaps the most common mtDNA pro-tein synthesis mutation is at np 3243 (A >

G) in the tRNALeu(UUR) gene (64). This mu-tation is remarkable in the variability of itsclinical manifestations. When present at rel-atively low levels (10%–30%) in the patient’sblood, the patient may manifest only type IIdiabetes with or without deafness (223). Thisis the most common known molecular causeof type II diabetes, purportedly accountingfor between 0.5% and 1% of all type II dia-betes worldwide (241). By contrast, when theA3243G mutation is present in >70% of themtDNAs, it does not cause diabetes, but in-stead causes more severe symptoms includ-ing short stature, cardiomyopathy, CPEO,and mitochondrial encephalomyopathy, lac-

MERRF: myoclonicepilepsy and raggedred fiber disease

MELAS:mitochondrialencephalomyopathy,lactic acidosis, andstroke-like episodes

tic acid and stroke-like episodes, the MELASsyndrome (64).

Mitochondrial Defects in Diabetes

The genetic linkage of mtDNA rearrange-ment (13, 14) and tRNA mutations (223) totype II diabetes directly implicated mitochon-drial defects in the etiology of diabetes and themetabolic syndrome. Evidence that mtDNAdefects are a common factor in the etiol-ogy of diabetes comes from the observationthat as the age-of-onset of the proband in-creases, the probability that the mother wasthe affected parent also increases, reachinga ratio of 3:1 for patients with a mean age-of-onset of 46 years (241). These observa-tions have been linked to the larger humanmetabolic syndrome through the identifica-tion of a mtDNA tRNAIle mutation at np4291 (T > C) that causes hypertension, hyper-cholesterolemia, and hypomagnesemia (renalductal convoluted tubule defect). This mu-tation was found in a maternal pedigree inassociation with reduced mitochondrial ATPproduction and the secondary clinical find-ings of migraine, hearing loss, hypertrophiccardiomyopathy, and mitochondrial myopa-thy (248).

Consistent with these genetic results, stud-ies of patients with type II diabetes have re-vealed that mitochondrial function and geneexpression are generally down-regulated.Insulin-resistant offspring of type II dia-betic patients have been found to have im-paired mitochondrial energetics, as assessedby 31P-MR spectroscopy (176). Furthermore,type II diabetes patients consistently show adown-regulation in the expression of nDNA-encoded mitochondrial genes, in associa-tion with alterations in the levels of theperoxisome-proliferation-activated receptorγ (PPARγ )-coactivator 1 (PGC-1) (153, 175),a major regulator of mitochondrial biogenesisand fat oxidation (105, 251).

Diabetes mellitus is also seen in Friedreichataxia. Patients with Friedreich ataxia mani-fest cerebellar ataxia, peripheral neuropathy,

www.annualreviews.org • Mitochondrial & Age-Related Diseases 369

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 12: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

PGC-1: PPARγ

(peroxisome-proliferating-activated receptor γ )coactivator 1

hypertrophic cardiomyopathy, and diabetesas the result of inactivation of the frataxingene on chromosome 9q13. Frataxin bindsiron in the mitochondrial matrix, thus min-imizing mitochondrial •OH production. Theloss of the frataxin protein results in excessiveROS generation which inactivates all mito-chondrial iron-sulfur-containing enzymes in-cluding complex I, II, III, and aconitase. Thusincreased mitochondrial ROS production anddecreased mitochondrial OXPHOS must bethe cause of diabetes in Friedreich ataxia (103,192, 249).

Type II diabetes has also been associatedwith a Pro121A polymorphism in the PPARγ

gene (5) and with a Gly482Ser polymorphismin the PGC-1 gene in Danish (53) and PimaIndian (156) populations.

Maturity onset of diabetes in the young(MODY) is an early-onset autosomal dom-inant form of type II diabetes that can alsobe linked to mitochondrial dysfunction. Themolecular defects of four forms of MODYhave been identified. MODY 2 is the resultof mutations in glucokinase, MODY 1 is dueto mutations in the hepatocyte nuclear factor(HNF)-1α, MODY 3 to mutations in HNF-4α, and MODY 4 to mutations in insulin pro-moter factor (IPF)-1.

Glucokinase has a very high Km for glucoseand is thought to be the glucose sensor. HNF-1α is a transcription factor and mutationsin its gene are associated with post-pubertaldiabetes, obesity, dyslipidemia, and arterialhypertension, all features of mitochondrialdiseases. HNF-1α is also important in reg-ulating nDNA-encoded mitochondrial geneexpression and the expression of the GLUT 2glucose transporter (250). HNF-4α, a mem-ber of the steroid/thyroid hormone receptorsuperfamily, acts as an upstream regulator ofHNF-1α (231).

The importance of mitochondrial defectsin β cell insulin secretion deficiency has beenconfirmed in two mouse models. In the first,the mitochondrial transcription factor Tfamwas inactivated in the pancreatic β cells. Thisresulted in increased blood glucose in both

fasting and nonfasting states, and the pro-gressive decline in β cell mass by apoptosis(201). In the second, the ATP-dependent K+-channel (KATP) affinity for ATP was reduced,resulting in a severe reduction in serum in-sulin, severe hyperglycemia with hypoinsu-linemia, and elevated D-3-hydroxybutyratelevels (102). These models demonstrate thatmitochondrial ATP production is critical inthe signaling system of the β cell to per-mit insulin release (238). Thus pancreaticβ cell mitochondrial defects are importantin both glucose sensing through glucokinaseand insulin release through the KATP channel(Figure 4).

Type II diabetes thus involves muta-tions in energy metabolism genes includingthe mtDNA and glucokinase; mutations inthe transcriptional control elements PPARγ ,PGC-1, HNF-1α, HNF-4α, and IPF-1; andalterations in insulin signaling. These seem-ingly disparate observations can be unifiedthrough the energetic interplay between thevarious organs of the body.

The human and mammalian organs canbe divided into four energetic categories: theenergy-utilization tissues including skeletalmuscle, heart, kidney and brain; the energy-storage tissues including brown adiposetissue (BAT) and white adipose tissue (WAT);the energy-homeostasis tissue, liver; and theenergy-sensing tissues including the α and β

cells of the pancreatic Islets of Langerhans.All of these tissues interact to coordinate theutilization and storage of energy, based on theavailability of calories in the environment.For our hunter-gather ancestors, the primaryvariation in available dietary calories was dueto the cyclic growing seasons of edible plantscaused by either warm versus cold or wet ver-sus dry seasons. During the growing season,plants convert the Sun’s energy into glucose,which the plants store as starch. Whenhumans and animals ingest these plant tissuesthe concentration of glucose in their bloodrises. Hence, serum glucose is the metabolicsurrogate for monitoring plant calorieabundance.

370 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 13: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Figure 4Model of pancreatic β cell showing the mitochondrial regulation of insulin secretion. The light yellowblob associated with VDAC represents glucokinase. The green double squares in the plasma membraneon the left side represent the glucose transporter. The two pairs of double gold squares on the right sideof the plasma membrane represent: below and labeled KATP the ATP gated K+ channel i and above andlabeled VDCa++ the voltage-dependent Ca++ channel. The circles with the internal “i” representinsulin containing vesicles. Other abbreviations as in Figure 2. Reprinted from Reference 25.

When plant calories are abundant andconsumed, the elevated serum glucose isdetected by the energy-sensing pancreatic β

cells, which respond by secreting insulin intothe blood stream. The insulin signal then in-forms the energy-utilizing heart and muscletissues to down-regulate mitochondrial en-ergy utilization, since food-seeking behavioris less pressing. It also informs the energy-storing WAT and BAT tissues to store theexcess calories as fat for when the seasonchanges and plant calories again become lim-ited. When plant calories do become limited,insulin secretion declines and the pancreaticα cells secrete glucagon. These low bloodsugar hormonal signals inform the energy-

utilization tissues to up-regulate the mito-chondrial OXPHOS system, thus enhancingfood-seeking capacity. They also mobilizethe energy-storage tissues to transfer thestored triglycerides into the blood to fuelthe increased mitochondria OXPHOS. Fur-thermore, low blood glucose stimulates theenergy-homeostasis tissue, liver, to synthe-sis glucose to maintain the basal level ofblood sugar, which is particularly critical forbrain metabolism. The molecular basis of thisprimeval system for adapting to energy fluctu-ation can now be partially understood throughrecent discoveries pertaining to the tran-scriptional regulation of mitochondrial geneexpression.

www.annualreviews.org • Mitochondrial & Age-Related Diseases 371

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 14: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

FOXO: mammalianforkheadtranscription factor

The energy-sensing tissue, pancreatic β

cells, detects abundant plant calories when theblood glucose level exceeds the high Km ofthe β cell glucokinase. Since glucokinase isbound to the mitochondrial VDAC and ANTthrough the mitochondrial inner and outermembrane contact points, the ATP bind-ing site of glucokinase is continuously occu-pied (3, 60, 125). The binding of glucose byglucokinase immediately generates glucose-6-phosphate (G-6-P) and then pyruvate.Pyruvate is oxidized by the mitochondria togenerate ATP. The elevated ATP productionincreases the ATP/ADP ratio, which results inthe closure of the KATP channel, depolarizingthe β cell plasma membrane. The depolarizedplasma membrane opens the voltage-sensitiveCa2+ channel. The influx of Ca2+ leads tothe formation of the exocytosis core complexand activation of protein kinases (255). Theinsulin-containing vesicles then fuse to theplasma membrane and release their stored in-sulin into the blood (Figure 4).

Thus, when blood glucose is high, theinsulin concentration in serum increases.The insulin binds to the insulin receptorof target cells throughout the body, acti-vating the insulin receptor tyrosine proteinkinase to phosphorylate insulin receptor sub-strates. These activate the phosphotidyli-nositol 3-kinase (PI3K), which phosphory-lates phosphotidylinositol 2-phosphate (PI2)to phophatidylinositol 3-phosphate (PI3). PI3then activates the AKT 1/2 kinases, whichphosphorylate the FOXO forkhead transcrip-tion factors FOXO1, FOXO3A, and FOXO4in the target tissues. Phosphorylation of theFOXOs results in their transport out of thenucleus and the transcriptional inactivationof genes whose promoters contain insulin re-sponse elements (IRE) (1, 59). The FOXOscan also be removed from the nucleus byacetylation via Cbp or p300 (1) and reactivatedby deacetylation via the NAD+-dependentSIRT1 (Sirtuin 1) (24).

When blood sugar is low, the pancreaticα cells secrete glucagon, which binds to theglucagon receptor in target cells. This stim-

ulates the production of cAMP, which ac-tivates protein kinase A (PKA). PKA phos-phorylates and activates the cAMP responseelement binding protein (CREB), which acti-vates transcription of genes harboring a cAMPresponse element (CRE) (48).

The FOXOs and CREB regulate numer-ous genes through the IRE and CRE ciselements. The PGC-1α promoter containsthree IREs that bind unphosphorylated anddeacetylated FOXO1, plus one CRE thatbinds phosphorylated CREB (48). PGC-1α

together with its companion genes PGC-1β

and PRC (PERC) are transcriptional co-activators that regulate genes for mitochon-drial biogenesis, thermogenesis, and fatty acidoxidation (92, 196).

PGC-1α was cloned through its interac-tion with PPARγ . It is strongly induced inBAT in response to cold acting through thecAMP-mediated β-adrenergic and thyroidhormone (TR) systems. The up-regulationof PGC-1α in turn induces UCP-1, whichthen creates a proton channel through themitochondrial inner membrane, uncouplingOXPHOS. This causes the rapid burningof the stored fats in BAT, generating heatto maintain body temperature (184). PGC-1α also induces mitochondrial biogenesisand UCP-2 in muscle cells (C2C12), act-ing through the nuclear respiratory factors 1and 2 (NRF1 and NRF2) (251). NRF1 andNRF2, in turn, activate the transcription ofa wide range of nDNA-encoded mitochon-drial genes, including components of OX-PHOS such as the ATP synthase β subunit(ATPsynβ) and cytochrome c (cytc) and themtTFA, which regulates mtDNA transcrip-tion (92, 196).

The PGC-1 family of transcriptional coac-tivators interacts with a broad spectrum oftissue-specific transcription factors. This pro-vides the tissue-specific response to the gen-eralized insulin and glucagon hormonal sig-nals. In the energy utilization tissue, muscle,PGC-1 induces mitochondrial biogenesisthrough the interaction with NRF1, PPARβ,PPARδ, MEF2, ERR2 (estrogen-related

372 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 15: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

receptor 2), HDAC5, and Gabpa/b (GA-repeat binding protein) (47, 78, 117, 152).In heart, PGC-1α interacts with PPARα andNRF1 (78, 111). In the brain, the transcrip-tional partners of PGC-1α have not yet beenidentified (118). In the energy-storage tis-sues, PGC-1α interacts with PPARγ , PPARα,and the thyroid hormone receptor (TR) inBAT to induce mitochondrial biogenesis andUCP-1 during cold stress (78, 251), whilein 3T3-Li preadipocytes, PGC-1α interactswith PPARα to stimulate fatty acid oxidation,including induction of the medium-chain acylCoA dehydrogenase (MCAD) (230). Finally,in the energy-homeostasis tissue, liver, PGC-1α interacts with FOXO1, HNF-4α, andthe glucocorticoid receptor (GR) to inducegluconeogenesis enzymes, including phos-phoenolpyruvate carboxylase (PEPCK) andglucose-6-phosphatase (G6Pase) to maintainbasal blood glucose levels (78, 121, 187, 254).

PGC-1β is abundantly expressed in BAT,heart, kidney, and skeletal muscle but alsoin stomach and in white adipose tissue. In3T3-L1 preadipocytes, PGC-1β is inducible,though PGC-1α is not, and PGC-1β theninteracts with the ERRs to induce MCADand fatty acid oxidation (89). Thus bothinsulin through the FOXOs and glucagonthrough cAMP modulate mitochondrial en-ergy metabolism in response to the availabilityof carbohydrates via the regulation of mem-bers of the PGC-1 co-activator family.

Mitochondrial energy metabolism is fur-ther modulated by PGC-1α through deacety-lation by SIRT1. PC12 cells that overexpressSIRT1 experience a 25% reduction in oxy-gen utilization (161). Hepatocytes that over-express SIRT1 induce the gluconeogenesisgenes PEPCK and G6Pase via interaction be-tween PGC-1α and HNF-4α (187). Further-more, upon caloric restriction, the SIRT1 infat cells binds to the nuclear receptor core-pressor (NCoR), blocking its interaction withPPARγ . This inhibits fat storage and en-hances fat mobilization (178, 179).

These observations link all of the knowngenetic mutations associated with diabetes to

defects in mitochondria bioenergetics and en-ergy metabolism. Mutations in the mtDNAand in glucokinase would result in ener-getic dysfunction. Mutations in PPARγ andPGC-1α would reduce mitochondrial geneexpression. Mutations in HNF-4α and its tar-get HNF-1α would result in loss of glucosehomeostasis. Thus, diabetes is an energy de-ficiency disease centered upon mitochondrialbioenergetics.

Given a mitochondrial etiology for type IIdiabetes, the various stages in the progres-sion of type II diabetes can be understood.In individuals harboring a partial defect inOXPHOS, the capacity of the energy-utilization cells to oxidize carbohydrates andfats to make ATP is reduced. Given a highcaloric diet, individuals with partial OXPHOSdefects overload their mitochondrial ETCwith excessive calories (reducing equivalents),hyperpolarizing �P, stalling the ETC, andblocking the tissue utilization of glucose. Asa result, the nonmetabolized glucose remainsin the blood. The chronically high serum glu-cose signals the β cells to secrete insulin, cre-ating concurrently elevated glucose and in-sulin: the hallmark of insulin resistance.

The excessive reduction of the mitochon-drial ETC electron carriers in the energy-utilization tissues maximizes mitochondrialROS production. The high serum insulin acti-vates their Akt pathway, which phosphorylatesthe FOXOs. The departure of the FOXOsfrom the nucleus stops transcription of thestress response genes, including the mito-chondrial antioxidant enzymes. It also sup-presses PGC-1α transcription, which down-regulates mitochondrial OXPHOS, furtherexacerbating the mitochondrial energy defi-ciency. The resulting chronic mitochondrialoxidative stress erodes mitochondrial functionand increases insulin resistance.

The excess of reducing equivalents also in-creases the NADH/NAD+ ratio. The conver-sion of NAD+ to NADH inhibits SIRT1, re-sulting in increased acetylation of the FOXOsand their removal from the nucleus. This fur-ther down-regulates PGC-1α and OXPHOS.

www.annualreviews.org • Mitochondrial & Age-Related Diseases 373

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 16: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

The sustained high serum glucose and in-sulin also activates the PGC-1α and PGC-1β

in the energy-storage tissues BAT and WATto store the excess calories as fats. Also, theliver accumulates lipid, and this can lead tononalcoholic steatohepatitis (NASH), whichis observed in over 30% of individuals withindications of insulin resistance, hypertriglyc-eridemia, and hypercholesterolemia (174).

In the β cells, the excessive mitochondriaROS inhibits mitochondrial ATP production,eventually leading to a decline in insulin se-cretion due to inadequate ATP for glucoki-nase and a low ATP/ADP ratio that cannotactivate the KATP channel. The resultinghigh glucose but reduced serum insulin istermed insulin-independent diabetes. Con-tinued calorie overload in the pancreatic β

cells and associated mitochondrial ROS pro-duction ultimately activates the β cell mtPTP,resulting in β cell death by apoptosis, produc-ing insulin-dependent diabetes.

Chronic mitochondrial oxidative stress onthe peripheral tissues subsequently damagesthe retina, vascular endothelial cells, periph-eral neurons, and the nephrons, leading tothe clinical sequelae of end-stage diabetes.Thus chronic mitochondrial dysfunction canexplain all of the features of type II diabetes,and is thus the likely cause of the disease.

ANCIENT MIGRATIONS ANDCLIMATIC ADAPTATION

In addition to having to adapt to changingcaloric availability due to seasonal changes,ancient human hunter-gathers had to adapt tothe rigors of different climatic zones. Whileinduction of UCP-1 in BAT and UCP-2 inmuscle permits acute adaptation to thermalstress in rodents, this is not an adequate mech-anism for long-term cold adaptation by hu-mans. UCP-1 induction in BAT cannot gen-erate sufficient heat to effectively regulatethe temperature of the much larger humanbody. Therefore, as humans migrated into themore northern latitudes they had to adapt tothe chronic cold using another mitochondrial

strategy. It now appears that this global cli-matic adaptive strategy was achieved by theacquisition of mtDNA mutations that par-tially uncoupled OXPHOS and thus resultedin perpetually increased mitochondrial heatproduction.

The first clear evidence that ancientmtDNA polymorphisms influence humanphysiology came from the observation thatLHON patients with mild complex I mtDNAmissense mutations frequently harbored thesame European mtDNA lineage, J. It is nowappears that lineage J mtDNAs harbor spe-cific “uncoupling mtDNA variants” that re-duce mitochondrial ATP output in favor ofheat production. Thus this lineage exacer-bates the partial ATP defect generated by thepathogenic mtDNA mutations.

Phylogeographic studies of the humanmtDNAs have revealed a remarkable corre-lation between mtDNA lineages and the ge-ographic origins of indigenous populations.These regional mtDNA lineages are groupsof related individual mtDNA sequences (hap-lotypes) known as haplogroups. The variousregional haplogroups form the branches ona single human dichotomous mtDNA phylo-genetic tree, generated by the accumulationof sequential mtDNA mutations on radiat-ing maternal lineages. The human mtDNAtree is rooted in Africa, and it has specificbranches radiating into different geographicregions that we now believe to have been con-strained by the climatic zones (28, 86, 144,239) (Figure 5).

African mtDNAs are the most diverse andthus most ancient, with an overall age of about150,000 to 200,000 YBP. African mtDNAs fallinto four major haplogroups: L0 (oldest), L1,L2, and L3 (youngest). L0, L1, and L2 rep-resent about 76% of all sub-Saharan AfricanmtDNAs and are defined by a HpaI restric-tion site at np 3592 [macro-haplogroup L(L0, L1, and L2)] (in Figure 1). In northeast-ern Africa, two mtDNA lineages, M and N,arose from L3 about 65,000 YBP. These werethe only mtDNA lineages that succeeded inleaving sub-Saharan Africa and radiating into

374 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 17: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Figure 5Diagram outlining the migratory history of the human mtDNA haplogroups. Homo sapiens mtDNAsarose in Africa about 150,000 to 200,000 years before present (YBP), with the first African-specifichaplogroup branch being L0, followed by the appearance in Africa of lineages L1, L2, and L3. Innortheastern Africa, L3 gave rise to two new lineages, M and N. Only M and N mtDNAs succeeded inleaving Africa and colonizing all of Eurasia about 65,000 YBP. In Europe, N gave rise to the H, I, J, Uk,T, U, V, W, and X haplogroups. In Asia, M and N gave rise to a diverse range of mtDNA lineagesincluding A, B, and F from N and C, D, and G from M. A, C, and D became enriched in northeasternSiberia and crossed the Bering land bridge about 20,000 to 30,000 YBP to found the Paleo-Indians. At15,000 YBP, haplogroup X came to central Canada either from across the frozen Atlantic or by an Asianroute of which there are no clear remnants today. At 12,000 to 15,000 YBP, haplogroup B entered theAmericas, bypassing Siberia and the arctic, likely by moving along the Beringian Coast. Next at 7000 to9000 YBP, a migration bringing a modified haplogroup A moved from the northeastern Siberia intonorthwestern North America to found the Na-Dene (Athebaskins, Dogrib, Apaches, and Navajos).Finally, relatively recently, derivatives of A and D moved along the Arctic Circle to found the Eskimos.These observations revealed two major latitudinal discontinuities in mtDNA variation: one between theAfrica L haplogroups and the Eurasia and N and M derivatives and the other between the plethora ofCentral Asian mtDNA lineages and the almost exclusive presence of lineages A, C, and D northeasternSiberia, the latter spawning the first Native American migrations. Since these discontinuities correspondto the transitions from tropical and subtropical to temperate and from temperate to arctic, we haveproposed that these discontinuities were the result of climatic selection of specific mtDNA mutationsthat permitted certain female lineages to prosper in the increasingly colder northern latitudes. Reprintedfrom http://www.mitomap.org.

Eurasia to give all of the Eurasian mtDNAs.In Europe, haplogroups L3 and N gave rise tohaplogroups H (about 45% of EuropeanmtDNAs), (H in Figure 1), T, U, V, W, andX (about 2%) as well as I, J (about 9%), andK(Uk). Europeans separated from Africansabout 40,000–50,000 YBP. In Asia, lineages

M and N radiated to give rise to a plethoraof mtDNA lineages. These include from N,haplogroups A, B, F, and others, and fromM, haplogroups C, D, G, and others (239)(Figure 5).

As Asians migrated northeast into Siberia,haplogroups A, C, D (A, C, D in Figure 1)

www.annualreviews.org • Mitochondrial & Age-Related Diseases 375

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 18: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

became progressively enriched, such that theybecame the predominant mtDNA lineagesin the indigenous peoples of extreme north-eastern Siberia, Chukotka. When the Beringland bridge appeared about 20,000 to 30,000YBP, people harboring these mtDNA hap-logroups were in a position to migrate intothe New World, where they founded thePaleo-Indians. After the land bridge sub-merged, haplogroup G arose in Central Asiaand moved into northeastern Siberia to pop-ulate the area around the Sea of Okhotsk.Later, a migration carrying haplogroup B (B inFigure 1) started from eastern Central Asiaand moved along the coast to the New World,by-passing Siberia. Haplogroup B then mixedwith A, C, and D in southern North Amer-ica, Central America, and northern SouthAmerica to generate the Paleo-Indians. Inaddition, haplogroup X was brought to theNew World in a migration that took placeabout 15,000 YBP. These immigrants settledin the Great Lakes region, and haplogroup XmtDNAs are found in 25% of the OjibwamtDNAs today. Since haplogroup X is foundprimarily in northeastern Europe, it has beenspeculated that an ancient European mi-gration carrying this haplogroup might alsohave contributed to the Paleo-Indian pop-ulations, perhaps bringing the progenitorsof the Clovis lithic culture to the Americas(239).

Later migrations from northeasternSiberia, carrying a modified lineage of hap-logroup A, founded the Na-Dene populationsabout 9500 YBP. More recently, immigrantsfrom Siberia bearing derived lineages ofhaplogroups A and D moved along the ArcticCircle to found the Eskimos and Aleuts (239).

This phylogeographic distribution ofmtDNA haplogroups reveals two striking dis-continuities in human mtDNA diversity. Thefirst occurs between sub-Saharan Africa andEurasia, in which all of the sub-SaharanAfrican mtDNA diversity remained in Africaand only derivatives of lineages M and N col-onized temperate Eurasia. The second occursbetween temperate Central Asia and arctic

Siberia, where the plethora of Asian mtDNAtypes is markedly reduced to only three an-cient mtDNA lineages (A, C, and D). The re-sulting fivefold enrichment of haplogroups A,C, D, and G in the arctic over Central Asiais unlikely to be the result of genetic bottle-necks, since there are no obvious geographicbarriers that separate Asia and Siberia. It ismore plausible that many Asian mtDNA lin-eages entered the arctic, but only a few sur-vived the intense cold to become permanentresidents. A similar logic could also apply tothe African-Eurasian discontinuity.

Evidence that climatic adaptation hasinfluenced the geographic distribution ofmtDNA diversity was first obtained from an-alyzing the amino acid replacement (nonsyn-onymous, NS) (Ka) to silent (synonymous,S) (Ks) mutation ratios (Ka/Ks) from the 13mtDNA open reading frames (83, 239). Thisrevealed that the amino acid sequence of theATP6 gene was highly variable in the arc-tic, but was strongly conserved in the tropicsand temperate zone; cytb was hypervariable intemperate Europe, but conserved in the trop-ics and arctic; and COI was variable in thetropical Africa, but invariant in the temper-ate and arctic regions. Regional variation wasalso observed in multiple ND subunits (148).Such regional gene-specific variation wouldnot be expected if all mtDNA mutations wererandom and neutral.

The geographic constraints on mtDNAprotein variation were further validated bypositioning all of the mtDNA variants from1125 complete mtDNA coding sequences col-lected from around the world and organizedinto a sequentially mutational tree. This treecould be assembled because the maternally in-herited mtDNA can only change by sequen-tial mutations along radiating female lines.Therefore, mutations shared between differ-ent mtDNAs must be derived from a commonfemale ancestor, and those that are confinedto a single mtDNA haplotype must be newmutations.

Analysis of the mtDNA nucleotide vari-ants revealed three different categories of

376 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 19: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

variants: (a) neutral, including synonymousand weakly conserved nonsynonymous aminoacid substitutions; (b) deleterious, alteringhighly conserved amino acids but located atthe tips of the branches of the tree, indicat-ing they are recent; and (c) adaptive, alter-ing highly conserved amino acids but locatedwithin the internal branches of the tree, in-dicating that they are ancient. The mutationsin class (c) must be adaptive because they alterhighly conserved amino acids, yet they havepersisted in the face of intense purifying se-lection for tens of thousands of years. Hence,as these variants are not neutral or deleterious,hence, they must be adaptive.

To quantify the potential effect of selec-tion on particular mtDNA variants, the inter-specific amino acid conservation [Conserva-tion Index (CI)] was determined for 22 knownpathogenic mtDNA replacement mutations(194). This gave an average CI for deleteriousmutations of 93.3 ± 13.3%. Using two stan-dard deviations around the mean CI of thepathogenic missense mutations to distinguishbetween functionally important versus neutralreplacement mutations, 26% of the internalbranch replacement mutations were found tobe functionally significant, with a mean CI =85.1 ± 9.2%. The remaining 74% were foundto be essentially neutral, with a mean CI =23.3 ± 14.9%.

Since these conserved internal branch mis-sense mutations frequently initiate new limbsof the mtDNA tree, they must have permit-ted individuals carrying these mutations tosurvive and multiply in new geographic re-gions, generating the region-specific mtDNAhaplogroups. The most obvious environmen-tal factor that differentiates tropical and sub-tropical sub-Saharan Africa from temperateEurasia and temperate Central Asia from arc-tic Siberia is temperature. This implies thatan important functional effect of many of theconserved internal branch missense mutationswas to permit adaptation to cold.

That these adaptive internal branch mu-tations were important in human radiationwas confirmed by analyzing the ratio of NS

to S missense mutations in the three majorclimatic zones. The internal branch NS/S ra-tio for the tropical and subtropical African Lhaplogroups was 0.31. The internal NS/S ra-tio for the temperate Eurasian haplogroupsM and N(R) were 0.42 and 0.44, whereasthe internal NS/S ratio for the primarilyarctic macro-haplogroup N(nonR) was 0.62.Thus, the farther north that the mtDNAswere found, the more missense mutations theyharbored.

The excess of internal branch missensemutations in the colder latitudes is particu-larly striking for haplogroups that reside inthe arctic and subarctic. The mean internalbranch NS/S ratio for northeastern Siberian-North American haplogroups A, C, D, andX was 0.61, much higher than the mean forthe non-arctic haplogroups of 0.39, the meanfor the African L haplogroups of 0.31, or themean for the Native American haplogroup B,which by-passed the cold selection of Siberia,of 0.38.

The internal branch replacement muta-tions of the arctic and subarctic haplogroupsalso have a higher average CI. The internalbranch CIs of haplogroups A, C, D, and Xwas 51%; that for the remaining global hap-logroups was 39%; that of L was 36%; andthat of B was 31%.

The European haplogroup internal branchreplacement mutations were also higher thanthe African L value of 0.31. Haplogroup Hwas 0.48, J was 0.66, and IWX was 0.63.By contrast, haplogroup T was only 0.31.However, this is because T harbors a singlehighly conserved founding replacement mu-tation in the ND2 gene (np 4917), whichcontributed virtually all of the adaptive ad-vantage to this lineage. Hence, the CI ofhaplogroup T was 20.3, the highest of any Eu-ropean haplogroup. Thus, adaptive changesfall into two categories, either the lineage ac-cumulates multiple missense mutations, eachchanging a somewhat less conserved aminoacid, or the lineage changes only a few aminoacids, each change of which is highly evolu-tionarily conserved.

www.annualreviews.org • Mitochondrial & Age-Related Diseases 377

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 20: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Examples of the adaptive mtDNA muta-tions that occur in the arctic mtDNAs in-clude two replacement mutations [ND2 np4824G (T119A) and ATP6 np 8794T (H90Y)]for haplogroup A; two replacement vari-ants [ND4 np 11969A (A404T) and cytb np15204C (I153T)] for haplogroup C; a ND2np 5178A (L237M) variant for haplogroup D;and a ND5 np 13708A (A458T) variant for asublineage of haplogroup X. This latter vari-ant also appears in European haplogroup J.

The European sister-haplogroups J andT provide the clearest example of the twoclasses of adaptive mutation strategies: severalless conserved mutations versus a few highlyconserved mutations. J and T share a commonroot involving two amino acid substitutions:ND1 np 4216C (Y304H) and cytb np 15452A(L236I). J and T then diverge. Haplogroup Tis founded by the single nodal adaptive mu-tation: ND2 np 4917G (N150D), the mostconserved ND2 polymorphism found (194).

Haplogroup J has two replacement muta-tions at its root: ND3 np 10398G (T114A) andND5 np 13708A (A458T), the second beingthe same variant found in haplogroup X. Hap-logroup J then splits into to sub-haplogroupsJ1 and J2, each defined by a major cytb mu-tation. The J2 cytb variant is at np 15257A(D171N) and the J1 cytb mutation is at np14798C (F18L). The np 14789C mutation isalso found at the root of sub-haplogroup Uk.The np 15,257 and np 14,789 variants alterwell-conserved amino acids with CIs of 95%and 79%, respectively. The 15,257 variant al-ters the outer coenzyme Q binding site (Qo) ofcomplex III, which contacts the Rieske iron-sulfur protein, while the np 14,798 site altersthe inner coenzyme Q binding site (Qi) ofcomplex III (194). Since the Qo and Qi bind-ing sites are essential for complex III protonpumping via the Q-cycle, the np 14,798 and15,257 variants are both likely to have discon-nected electron flow through complex III withproton pumping. This would reduce the cou-pling efficiency of mitochondrial OXPHOSby one third and proportionately increase heatgeneration.

That the internal branch mtDNA mis-sense mutations are functionally relevant hasbeen demonstrated by comparing the spermmobility of males harboring the differentmtDNA haplogroups. Sperm flagellar motionis driven primarily by ATP generated from themitochondria in the mid-piece. Therefore,sperm with partially uncoupled mitochondrialshould swim slower that those with coupledmitochondria. As expected, sperm from hap-logroup H subjects swam significantly fasterthan those from T subjects (193). Thus thefunctional mtDNA variants that founded spe-cific mtDNA lineages affect mitochondrialphysiological functions.

ADAPTIVE mtDNA MUTATIONSIN LONGEVITY ANDDEGENERATIVE DISEASES

That these adaptive mutations are clini-cally relevant has become apparent fromstudies that correlated mtDNA haplogroupswith longevity and degenerative disease. Inan Italian study, mtDNA haplogroup J wasfound to be overrepresented in centenarians(50, 189). Similarly, in an Irish centenarianstudy, J2 was overrepresented (191), and ina Finnish study of individuals over 90 yearsJ2, Uk, and WIX were enriched (163). InJapanese centenarians a sublineage of hap-logroup D was enriched (210, 211). Hence,specific mtDNA lineages from Europe andAsia are protective against the ravages ofaging.

Some of these same mtDNA lineages havealso been found to be protective against neu-rodegenerative diseases. Haplogroups J andUk are underrepresented in Parkinson disease(PD) (225) and haplogroup T is underrepre-sented in AD patients (32, 224). The repeatedassociation of haplogroups J1 and Uk withlongevity and neuro-protection is particularlyilluminating because both haplogroups en-compass the same cytb mutation at np 14,798.Such convergent evolution provides strongsupport for the functional importance of thecytb mutations.

378 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 21: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

In other studies, haplogroup J has beenfound to increase the penetrance of the milderLHON mutations (20). Haplogroup T hasbeen found to be overrepresented in bipolaraffective disorder (BPAD) (133).

How can the same variants be associatedwith increased life span and protection againstcertain diseases on the one hand, yet increasethe predilection of developing other degener-ative diseases on the other? The answer lieswith the interdependence of mitochondrialenergy production, ROS production, and themtPTP activation of apoptosis.

The cold-adapted mtDNA uncouplingmutations would generate less ATP per calo-rie consumed. Hence, uncoupled mitochon-dria would be more prone to clinical prob-lems resulting from energy insufficiency suchas LHON and bipolar mental illness. How-ever, individuals with uncoupled mitochon-dria would burn calories more rapidly to gen-erate both the required ATP plus increasedheat. As a result, the ETCs of uncoupled in-dividuals would be generally more oxidized,thus minimizing the production of mitochon-drial ROS. Reduction in mitochondrial oxida-tive stress would reduce the activation of themtPTP, thus preserving cells and protectingthe individuals from cellular loss leading toneurological and visceral tissue degenerationand aging.

THE MITOCHONDRIALEITIOLOGY OF AGING

Since mtDNA haplogroups that harbor adap-tive mtDNA uncoupling mutations have a re-duced rate of aging and neurodegenerativedisease, the mitochondria must be an impor-tant factor in the aging process. Aging is thedecline of structure and function over time,which is a natural consequence of entropy, thetendency for all complex systems to decay withtime. To counter entropy requires energy torepair or rebuild the damaged functions.

Since uncoupling mtDNA mutations areassociated with increased human life span and

would have reduced mitochondrial ROS pro-duction, ROS damage to the mitochondria,mtDNA, and host cell must be one of themost important entropic factors in determin-ing age-related cellular decline. To counterendogenous sources of oxidative damage, thenucleus must make enzymes that limit mito-chondrial ROS damage, thus preserving themitochondria and mtDNAs.

An important role of mitochondrial ROSproduction in aging and degenerative diseasesis congruent with the life-extending capacityof caloric restriction (69, 126, 127, 130, 204).Reduction of available calories will starve themitochondrial ETC for electrons, thus reduc-ing ROS and protecting the mitochondria andmtDNAs. Caloric restriction can be achievedby direct dietary restriction or by blocking theretention of excess calories. This may explainwhy mice in which the insulin receptor genewas inactivated in adiposities had an 18% in-crease in life span (16).

In rodent studies, mtDNA base oxidation(8oxo-dG) and rearrangement mutation lev-els have been found to increase with age. Di-etary restriction will inhibit the accumula-tion of both forms of mtDNA damage (67,139). Furthermore, aging alters gene expres-sion in muscle and brain, but calorie restric-tion prevents many of these changes (106,107). In muscle, two classes of genes areaffected in aging, those for mitochondrialenergy metabolism and those for antioxidantdefenses. Calorie restriction normalizes theexpression of many of these genes (106).

Drosophila life span can be extended bydietary restriction, and the functional basisof the life extension appears to overlap withinsulin-like growth factor defects (35). Thechico (small boy) dwarf mutant of Drosophila hasa defect in an insulin-like growth factor recep-tor substrate protein and an increased life span(36). That this relates to energy metabolismwas suggested by DNA microarray analysesthat revealed the down-regulation with ageof genes involved in metabolism, cell growth,and reproduction (180).

www.annualreviews.org • Mitochondrial & Age-Related Diseases 379

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 22: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Evidence for a role for mitochondrialenergy metabolism in Drosophila aging alsocomes from the Indy (I’m not dead yet) mu-tant. This mutation inactivates a dicarboxylatecotransporter expressed in the midgut, fatbody, and oenocytes of the fly. This enzymeprobably mediates the uptake or reuptake ofdi- and tricarboxylic acid TCA cycle interme-diates (188). Since Drosophila lives on decayingfruit, which is rich in the di- and tricarboxylicacids that are substrates for the mitochon-drion, the Indy mutant could be restricting theavailability of calories to the mitochondrion.

Evidence that ROS toxicity is a limitingfactor for life span in Drosophila was directlydemonstrated by showing that transgenicDrosophila expressing increased Cu/Zn SODand catalase lived longer (168). Drosophila lifespan extension was also seen in flies in whichMnSOD was overexpressed in motor neurons(173) and systemically in Drosophila adults af-ter heat shock induction of the MnSOD trans-gene (208). These observations support theconclusion that mitochondrial ROS toxicityis an important factor in limiting Drososphilalife span.

A mitochondrial role in longevity has alsobeen observed in the nematode Caenorhabdi-tis elegans. A number of the C. elegans mutantsthat extend life span specifically affect mito-chondrial function. These include the clk-1mutant, which affects a gene in the biosyn-thetic pathway of coenzyme Q (71, 87, 150),the isp-1 mutant, which alters the Rieske ironsulfur protein of respiratory complex III (56),a mitochondrial leucyl-tRNA synthetase mu-tant (110), and a mitochondrial tRNA wobbleU modifying enzyme, isopentenylpyrophos-phate:tRNA transferase mutant (114). Sincethe isp-1 mutation has been shown to bothincrease longevity and reduce mitochondrialROS production, it seems likely that mito-chondrial ROS must be modulating C. eleganslife span (56).

The importance of the mitochondria inregulating C. elegans life span was further con-firmed by an RNAi scan of the genes lo-cated on chromosomes I and II. Of the genes

whose inactivation extended life span, 15%were identified as having mitochondrial func-tions. Since only about 1.5% of the genes ofthe C. elegans genome have been proposedto have a mitochondrial function, mitochon-drial functions must have a much greater in-fluence on longevity than other physiologicalprocesses (109, 110).

The life span of C. elegans is also regu-lated by the insulin-like growth factor signaltransduction pathway. The metabolic func-tions associated with key longevity muta-tions include the insulin-like receptor (IRL)(daf-2), the catalytic subunit of PI3Kinase(age-1), the PTEN-like lipid phosphatase(daf-18), the Akt-1/2 kinases, and the tar-get of Akt 1/2 phosphorylation, the fork-head transcription factor (daf-16). Phos-phorylated forkhead transcription factor iscytosolic and transcriptionally inactive. Thusmutational inactivation of the insulin-likegrowth factor pathway must extend life spanby blocking phosphorylation of the fork-head transcription factor and rendering itconstitutively transcriptionally active (65, 93,165, 215). Two genes that are up-regulatedby activation of the daf-16 forkhead tran-scription factor via dephosphorylation areMnSOD and catalase (122). Hence, one im-portant function of the insulin-like signalpathway in C. elegans appears to be the nega-tive regulation of the stress response and an-tioxidant enzymes that protect the cell fromthe deleterious effects of mitochondrial ROSproduction.

Life span in C. elegans as well as in yeast canalso be extended by the overexpression of theNAD+-dependent protein deacetylase Sir2.Sir2 acts, in part, through the deacetylationand activation of the forkhead transcriptionfactors, favoring nuclear relocation and tran-scription activation (61, 214). In yeast, Sir2also connects the insulin-like signaling path-way to caloric restriction. Sir2 requires NAD+

as a co-reactant. NADH will not suffice andeven acts as a competitive inhibitor of yeast.Since caloric restriction would reduce thecellular NADH to NAD+ ratio, this should

380 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 23: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

activate Sir2, causing the deacetylation andactivation of the forkhead transcription fac-tors and thus increasing expression of the an-tioxidant and stress response genes (119, 120).By contrast, defects in mitochondrial OX-PHOS should increase the NADH/NAD+

ratio, inhibit SIR2, and favor the acetylation ofthe forkhead transcription factors. This wouldturn off antioxidant and stress response geneexpression, thereby reducing antioxidant de-fenses and life span.

This overlap between the insulin-like sig-naling pathway and the mitochondrion hasbeen demonstrated in C. elegans by combiningdaf-2 and isp-1 mutants and showing that theydo not have an additive effect on longevity.This is logical since inactivation of the daf-2 pathway would increase mitochondrial an-tioxidant defenses while the isp-1 mutationwould reduce mitochondrial ROS produc-tion (56). Hence, the two classes of mutationswould have the same effect.

That mitochondrial ROS production is animportant factor affecting C. elegans life spanhas been confirmed by the mev-1 mutationand the effects of SOD mimetics. The mev-1mutation affects the cytb-containing subunitof complex II. This mutation has a markedlyincreased rate of mitochondrial ROS produc-tion and a substantially shortened life span(84, 198). This reduced longevity can be re-versed by treatment with the catalytic antiox-idant mimetic EUK134, a salen Mn complex.Furthermore, treatment of wild-type C. el-egans with EUK134 can increase the meanand maximum life span of C. elegans to an ex-tent similar to that of the long-lived C. ele-gans mutant age-1 (140). Therefore, the lifespan of C. elegans may be modulated by ei-ther decreasing mitochondrial ROS produc-tion or increasing mitochondrial antioxidantdefenses.

Corroborative data that mitochondrialROS limits the life span of mammals havecome from studies on mutant and transgenicmice. Mice in which the MnSOD wasgenetically inactivated die at a mean age ofeight days of a dilated cardiomyopathy, in as-

sociation with the inactivation of iron-sulfurcenter containing enzymes including complexI, II, III, and aconitase (115, 137). The mito-chondria of these animals have reduced stateIII respiration and hypersensitized mtPTPs(97), and they can be rescued from theircardiomyopathy by treatment with the SODmimetic MnTBAP [Mn 5, 10, 15, 20-tetrakis(4-benzoic acid) porphyrin]. However,MnTBAP does not readily cross the blood-brain barrier and whereas the treated miceavoid the cardiomyopathy, they developmovement disorders together with corticaland brainstem spongiform vacuolization andbrainstem astrocytosis (142). Again, both thesystemic and neurological pathology can beameliorated by treatment with the salen Mncomplex compounds EUK8, EUK134, andEUK189, which do cross the blood-brainbarrier and are protective against both thevisceral effects and the neurological patholo-gies (138). Thus mitochondrial ROS damagemust be important in the age-related declineof both mammalian visceral and centralnervous system cells and tissues.

Although mice heterozygous for the Mn-SOD mutation live to old age, they havea chronically reduced liver mitochondrialmembrane potential, reduced state III respi-ration equivalent to that of an old animal, anda striking rise in mitochondrial lipid perox-idation by middle age that drops to belowthe wild-type levels in older mice. This strik-ing drop in lipid peroxidation is associatedwith a marked increase in the sensitivity ofthe mtPTP to Ca2+ activation and a strik-ing increase in number of liver apoptotic cellsin older heterozygous animals. The MnSODheterozygotes also showed a striking induc-tion of complex IV (COX) in the older animals(97). Aging MnSOD heterozygous C57Bl/6(B6) mice also show a significant increase inoxidative damage (8oxo-dG) to nDNA andmtDNA, and a 100% increased tumor inci-dence, though their life span is not diminished(226).

Proof that mitochondrial ROS limit mam-malian life span has come from the targeting

www.annualreviews.org • Mitochondrial & Age-Related Diseases 381

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 24: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

of the human catalase to the mouse mito-chondria in transgenic mice and observingan increase in life span. To import catalaseto the mitochondrial matrix, the C-terminalperoxisomal targeting peptide was inacti-vated, and the catalase coding sequence wasfused to the N-terminal mitochondrial target-ing peptide of the mitochondrial matrix en-zyme ornithine transcarbamylase. This con-struct was then expressed in transgenic miceusing an α-actin promoter and CMV en-hancer. The modified catalase was shown tobe imported into the mitochondrion and tostrikingly increase catalase activity in heartand muscle. Moreover, heart mitochondriafrom the transgenic animals showed a dra-matically increased resistance of mitochon-drial aconitase to inactivation by H2O2,and the skeletal muscle mtDNAs showed asignificant reduction in the age-related ac-cumulation of mtDNA rearrangement muta-tions. These protective effects to mitochon-drial oxidation and mtDNA mutations wereassociated with a 21% increase in mean lifespan and a 10% increase in maximum life spanin both male and female mitochondrial cata-lase transgenic animals (197).

The importance of mitochondrial ROS inlongevity has also been supported by the se-lective inactivation of the p66shc splice vari-ant of the p52shc/p46shc gene. This mutantincreases mouse life span by 30%, resultsin increased resistance to oxidative stresscaused by paraquat, and reduces mitochon-drial ROS production (146). p66Shc has beenshown to become incorporated within the mi-tochondria, where it interacts with HSP70and affects mitochondrial function and DNAoxidative damage (169). It is also a down-stream target of p53 and is required for stress-activated p53 to induce intracellular ROS pro-duction, cytochrome c release, and apoptosis(218).

That increased mitochondrial ROS pro-duction accelerates mtDNA mutation ratewas shown in mice deficient in the nDNA-encoded heart-muscle (and brain) isoform ofthe ANT gene (Ant1). These animals exhibit

severe fatigability and develop a hypertrophiccardiomyopathy. Since an ANT defect blocksthe import of cytosolic ADP into the mi-tochondria, and the ATP synthase withoutADP cannot utilize �P to make ATP, then�P becomes hyperpolarized, the ETC stalls,and the excess electrons are transferred to O2

to generate ROS. Consequently, the ANT1-deficient mice have greatly increased heart,muscle, and brain mitochondrial H2O2 pro-duction. This increased mitochondrial ROScorrelates with a dramatic increase in mtDNArearrangements in the heart by middle age(54).

That mtDNA mutations are one of the pri-mary factors that limit mouse life span wasdemonstrated by the knock-in of a mutantform of the mtDNA polymerase γ (POLG)subunit A in which the proofreading func-tion was inactivated by the mutation D257A.These mice had a shortened life span and de-veloped a premature aging phenotype involv-ing weight loss, reduction of subcutaneousfat, hair loss (alopecia), curvature of the spine(kyphosis), osteoporosis, anemia, reduced fer-tility, and heart enlargement. This was associ-ated with an age-related decline in respiratorycomplexes I and IV and in mitochondrial ATPproduction rates in the heart. Analysis of themtDNA revealed that the knock-in animalshad a three- to fivefold increase in mtDNAbase substitution mutations in brain, heart,and liver, with a higher number of mutationsin the coding region cytb gene than in the CR(216). Thus, increased mtDNA mutations candirectly increase the aging rate, implying thatthe mtDNA is one of the most important tar-gets for mitochondrial ROS damage in aging.

The relationship between mtDNA mu-tations and longevity has been shown in awide variety of animal species including hu-mans. mtDNA rearrangement mutations havebeen shown to accumulate in proportion tolife span in organisms as diverse as mouse,chimpanzee, and human (43, 139, 141, 143).When the level of mtDNA rearrangementswas quantified in various human brain regionsby the analysis of the common 5-kb mtDNA

382 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 25: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

deletion, mtDNA rearrangements were foundto accumulate to the highest levels in thebasal ganglia, followed by the cerebral cortex,with the least mutant being found in thecerebellum (39, 206). Base substitution mu-tations also accumulate in human tissues withage. A mtDNA CR mutation, T414G, in themtTFA binding site of the PL accumulateswith age in diploid fibroblasts (145); a T408Apromoter mutation and a A189G OH muta-tion accumulate with age in skeletal muscle(245); and a T150C OH mutation accumu-lates with age in white blood cells (256). ThesemtDNA CR mutations appear to be highlytissue specific. The T414G mutation can bedetected at low levels in skeletal muscle byusing a highly specific and sensitive proteinnucleic acid (PNA)-clamping PCR method.However, this same method has been unableto detect the T414G mutation in the brains ofeven very elderly people (157). These mtDNAmutations can be attributed to oxidative dam-age, at least in the brain, since the human brainmtDNA shows a marked age-related increasein oxidative damage (135).

All of these data are consistent with thehypothesis that aging is the result of mito-chondrial decline due to oxidative damageto the mitochondria and mtDNAs. This hy-pothesis, when combined with our currentunderstanding of caloric regulation of mito-chondrial function, can neatly explain whycaloric restriction is able to extend mam-malian life span. Reduction of dietary calo-ries reduces serum glucose, thus inhibitinginsulin secretion by the pancreatic β cells.This results in the dephosphorylation andactivation of the FOXO forkhead transcrip-tion factors in the energy-utilizing tissues.Further, caloric restriction reduces cellularNADH/NAD+ ratios, activating the NAD+-dependent SIRT1, which further activates theFOXOs by deacetylation (24, 61, 155). Theactivated FOXOs then activate the transcrip-tion of the stress response genes includingthe antioxidant enzymes MnSOD and cata-lase. They also induce, along with glucagonthrough CREB, the transcriptional induction

of PGC-1α. This increases the mitochon-drial OXPHOS capacity, mobilizes the WATtriglyceride stores for mitochondrial oxida-tion, and activates gluconeogenesis to main-tain the basal serum glucose level. Hence,caloric restriction induces the mitochondriato more completely oxidize dietary reduc-ing equivalents and the antioxidant defensesystems to more effectively eliminate ROS.Thus, dietary restriction should substantiallyreduce mitochondrial oxidative stress. In ad-dition, activation of SIRT1 by reduction ofthe NADH/NAD+ ratio will stimulate thedeacetylate p53, and deacetylation of p53 re-duces its apoptotic potential. Hence, caloricrestriction also protects the mtPTP from p53-mediated activation and apoptosis (82, 104,123, 229). Therefore, caloric restriction ex-tends life span in three ways: by decreasingmitochondrial generation of ROS, by increas-ing the cellular antioxidant defenses, and byinhibiting cell loss via apoptosis.

THE MITOCHONDRIALETIOLOGY OF DEGENERATIVEDISEASES

The level of mtDNA rearrangement muta-tions has also been found to be strikinglyincreased in a variety of age-related degener-ative diseases. The 5-kb mtDNA rearrange-ment is increased between 7- and 2200-foldin the hearts of patients with chronic coro-nary artery disease (41, 42). Since coronaryartery disease is associated with recurrent car-diac ischemia and reperfusion, which gener-ates bursts on mitochondrial ROS (49), thecardiac rearrangement mutations may also becaused by mitochondrial oxidative damage.

Similarly, mtDNA rearrangements arefound in patients with AD. In the brains ofAD patients who died prior to age 75, the5-kb deletion is increased on average 15-fold,whereas in the brains of patients who diedafter age 75, the 5-kb deletion is present atone fifth the level of that in control brains(40). The increased mtDNA rearrangementmutations in AD brains are associated with

www.annualreviews.org • Mitochondrial & Age-Related Diseases 383

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 26: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

APP: amyloidprecursor protein

increased mtDNA oxidative damage (134).The sudden drop in the level of mtDNA dele-tions in older AD brains might be the resultof loss of the brain cells containing the mostmutated mtDNA through apoptosis. This isconsistent with the increased neuronal apop-tosis observed in AD brains (30).

AD, is a progressive neurodegenerativedisease resulting in dementia associated withthe deposition of Aβ amyloid peptide plaquesand neurofibrillary tangles in the brain. Early-onset, familial AD has been associated withmutations in the Aβ precursor protein (APP)and the presenilin-containing APP peptideprocessing complexes (212). Although themolecular defects that cause the late-onset,sporadic AD cases have not been determined(203), the inheritance of the ApoE ε4 allelehas been identified as a major risk factor fordeveloping late-onset AD (190, 203).

Mitochondrial OXPHOS defects have alsobeen detected in a variety of tissues from ADpatients. The mtDNA of AD patients has beenindirectly shown to harbor genetic defects byfusing blood platelets from AD patients to hu-man ρ0 cell lines (94), with the resulting cy-brids being found to have OXPHOS defects(17, 29, 45, 96, 158, 159, 217, 222, 232). Theconcept that mtDNA mutations might con-tribute to late-onset AD has been bolsteredby the observation that a mtDNA tRNAGln

gene mutation at np 4336 is present in about5% of late-onset AD patients and in 7% ofcombined AD plus PD patients, but in 0.4%of normal controls (81, 199).

The association between mitochondrialdeficiency and AD was further strengthenedrecently by the demonstration, using PNA-clamping PCR, that the somatic mtDNA CRT414G mutation in the PL mtTFA bindingsite can be detected in 65% of all AD brainsbut not in any control brains. Moreover, thelevel of somatic mtDNA CR mutations inAD brains is increased by 63% overall, withthe elevation of somatic mtDNA CR mu-tations in AD patients over 80 years being120%. Furthermore, in the AD brains, thesomatic mtDNA mutations preferentially oc-

cur within the known mtDNA transcriptionand replication regulatory elements, whereasvery few of the mtDNA CR mutations foundin the control brains occur in functional el-ements. Finally, AD brains have an average50% reduction in the ND6/ND2 mRNA ra-tio and a comparable reduction in the levels ofthe mtDNA/nDNA ratio, suggesting the in-hibition of l-strand transcription and mtDNAreplication (44).

These data suggest that the accumulationof somatic mtDNA CR mutations in ADbrains that damage key mtDNA regulatoryelements may be a central feature in the eti-ology of the disease. The accumulation ofthese mutations with age would result in theprogressive decline in mitochondrial energyproduction and an associated increase in mi-tochondrial ROS production. The increasedROS would ultimately lead to the activation ofthe mtPTP and the loss of neuronal synapsesthrough mitochondrial induced cytochrome crelease and caspase activation.

By this logic, individuals harboringmtDNAs that generate tightly coupled mi-tochondria would experience chronicallyincreased ROS production on a high calo-rie diet. This would increase the probabil-ity of mtDNA CR mutations and thus theprobability of developing AD. By contrast,individuals who harbor mtDNAs with uncou-pling polymorphisms would have reduced mi-tochondrial ROS and be less prone to somaticmtDNA CR mutations and AD. Populationstudies have confirmed these predictions (32,224).

Mitochondrial ROS production might alsoaccount for the connection between the ApoEε4 allele and AD, since the presence of theApoE ε4 allele has been associated with in-creased brain oxidative damage (151, 185,186). Aβ peptide toxicity might also be re-lated to mitochondrial dysfunction. The Aβ

peptide has recently been reported to enterthe mitochondria, bind to mitochondrial al-cohol dehydrogenase, and increase mitochon-drial ROS production (46, 124). Moreover,incubation of rat brain mitochondria with the

384 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 27: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Aβ25–35 fragment results in a selective anddose-independent inhibition of complex IV(27). Consistent with the possibility thatAβ enters the mitochondrion, the insulin-degrading enzyme (IDE) has been discoveredto have a mitochondrially directed form gen-erated by initiating translation at an AUGcodon 41 amino acids upstream from thecanonical IDE peptide start codon. Since theIDE is thought to degrade both cerebral Aβ-peptide and plasma insulin, it could functionin mitochondrial Aβ turnover (113).

The Aβ peptide is generated by cleavageof the APP by the presenilin-containing γ -secretase complex. Recently, the presenilin-1(PS1) protein has been located in the innermembrane of rat brain and liver mitochon-dria (9). The other components of the prese-nilin γ -secretase including nicastrin, APH-1,and PEN-2 have also been found in a high-molecular-weight complex within the mito-chondrial inner membrane. Although the im-port mechanism for these proteins is not fullyunderstood, the nicastrin protein has beenfound to harbor both ER and mitochondrialN-terminal targeting signals (68). APP hasalso been found in the mitochondrion, but it isthought to be located in the outer mitochon-drial membrane, a position that would not beconducive to its processing into Aβ by the mi-tochondrial inner membrane γ -secretase (7).Therefore, the γ -secretase complex proteinsmust have other functions in the mitochon-drion. Given that γ -secretase is unique in itsability to cleave peptide bonds within mem-branes, the PS1 complex may well be process-ing mitochondrial inner membrane proteinssuch as those of the respiratory complexes.Hence mutations in the presinilin complex as-sociated with early-onset AD might also causemitochondrial defects.

A mitochondrial etiology of AD couldalso explain the near-universal association ofAβ amyloid plaques with AD. At the con-centrations found in biological fluids, Aβ1–40 and Aβ1–42 act as antioxidant and anti-apoptotic polypeptides, presumably becauseof their tendency to chelate transition metals,

particularly copper. However, when the Aβsaggregate, they become pro-oxidants, perhapsthrough placing the transition metal in thepresence of the redox-active Met 35 (99–101).Given this perspective on the Aβ peptides,it could follow that genetic variants whichchronically increase mitochondrial ROS pro-duction would overwhelm the antioxidant ca-pacity of basal levels of Aβ. To compen-sate, the brain would increase Aβ production.However, this would eventually lead to excessAβ, which would then aggregate into a pro-oxidant form (80) and thereby exacerbate theproblem. Consistent with this scenario, thebrains of late-onset sporadic AD patients havebeen observed to have increased oxidativedamage, increased activated caspase activity(62, 183), and increased numbers of TUNEL-(terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling) positive cells (45).Increased mitochondrial ROS would also in-crease the mtDNA somatic mutation rateand exacerbate mitochondrial oxidative stress,mtPTP activation, and synaptic loss.

Early-onset AD could also fit within thisframework. Mutations in the Aβ processingpathway would increase the rate of Aβ pro-duction and aggregation, creating an activepro-oxidant that would increase synaptic ox-idative stress, damage the mitochondria, anddestroy the synapses.

A similar mitochondrial scenario couldbe applied to other age-related neurodegen-erative diseases. Amyotrophic lateral sclero-sis (ALS) is a neurodegenerative disease thatspecifically affects the motor neurons. In fa-milial cases, ALS is caused by mutations in theCu/ZnSOD (Sod1). Transgenic mice that ex-press a human mutant Cu/ZnSOD developmotor neuron disease associated with vac-uolate mitochondria containing high con-centrations of Cu/ZnSOD (85). The mutantCu/ZnSOD mice also accumulate 8oxo-dGin cytoplasmic granules, possibly represent-ing mtDNA nucleoids (247), and in culturedneuroblastoma cells, the toxicity of a mu-tant Cu/ZnSOD can be ameliorated by theincreased expression of MnSOD (57). The

www.annualreviews.org • Mitochondrial & Age-Related Diseases 385

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 28: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

mitochondrial uptake of the three commonfamilial ALS Cu/ZnSOD mutants (G37R,G41D, and G93A) is blocked by the heatshock proteins Hsp70, Hsp27, and Hsp25(167). Consequently, familial ALS appearsto be due to a failure in the Cu/ZnSOD’sability to detoxify mitochondrial superoxideanion, which is released into the mitocho-ndrial inner membrane space from ubisemi-quinone bound to the Qo site of complex III.

Idiopathic PD has also consistently beenreported to be associated with mitochon-drial defects, particularly in respiratory com-plex I. Evidence supporting a mitochon-drial etiology of PD has been previouslyreviewed but includes the selective destruc-tion of the dopaminergic neurons in humansand rodents by the mitochondrial complex Iinhibitor 1-methyl-4-phenyl-1,2,3,6-tetrahy-dropyridine (MPTP); the detection of OX-PHOS defects and particularly complex Idefects in PD patient muscle, blood, brain,and in platelet-derived cybrids; and the in-heritance of PD-like symptoms to variouspathogenic mtDNA mutations (241) includ-ing the common LHON np 11,778 muta-tion (202). Small subsets of PD patients arealso found in Mendelian pedigrees. The genefor one rare autosomal recessive form of PD,which maps to chromosome 1p35-p36, hasbeen cloned and found to code for the PINK1(PTEN-induced kinase) protein, a kinase lo-calized in the mitochondrion (221). A morecommon autosomal recessive locus, the parkingene on chromosome 6q25.2–27, is a E3ubiquitin ligase (228). Genetic inactivation ofparkin in the mouse resulted in nigrostriataldefects associated with a decreased abundanceof several mitochondrial proteins includingthe E1α subunit of pyruvate dehydrogenase,the 24- and 30-kDa subunits of complex Iand the Vb subunit of complex IV and a gen-eral decrease in mitochondrial state III res-piration. The oxidative stress proteins per-oxiredoxin 1, 2, and 6 and lactolyglutathionewere also down-regulated in association witha marked increase in brain lipid peroxidationproducts (171). Hence, it would appear that

the pathophysiology of the parkin mutationis also associated with a mitochondrial defect.Finally, the rare autosomal dominant form ofPD, which maps to chromosome 4q21-q22,is due to mutations in a-synuclein (182). Ex-pression of mutant α-synuclein in various cellsystems has resulted in altered mitochondrialfunction (79, 108, 170). Thus, both the idio-pathic and the inherited forms of PD appear tobe associated with mitochondrial dysfunction.This is consistent with the protective effectsof haplogroups J and Uk for PD (225).

Studies on Huntington disease have impli-cated a mitochondrial dysfunction in its eti-ology as well (172, 195, 241), and mtDNArearrangements are elevated in the basal gan-glia of patients with Huntington disease (76).Thus all of these age-related neurodegenera-tive diseases, as well as aging itself, seem to bedirectly related to the mitochondrial produc-tion of ROS and the resulting accumulation ofmtDNA mutations. These observations implythat the mitochondrial paradigm for neurode-generative diseases appears to be one that canbe broadly applied.

SOMATIC AND GERMLINEmtDNA MUTATIONSIN CANCER

Aging is also the most significant risk factorfor most solid tumors. Moreover, caloric re-striction in rodents markedly reduces cancerrisk (69, 126, 127, 130, 204). Hence, the eti-ology of cancer might also be associated withmitochondrial dysfunction.

Gene defects in certain nDNA-encodedmitochondrial genes have been directly linkedto some hereditary cancers. Mutations in themitochondrial fumarate hydratase have beenassociated with uterine leiomyomas and renalcell carcinomas (112), and mutations in threeof the four nDNA-encoded subunits of com-plex II have been linked to paragangliomas:SDHD (cytb membrane subunit) (15), SDHC(coenzyme Q-binding membrane subunit)(162), and SDHB (iron-sulfur) subunit (11,227). In addition to the paragangliomas seen

386 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 29: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

for mutations in all three subunits, mutationsin the SDHB subunit are also associated withpheochromocytoma and early-onset renal cellcarcinoma. On the other hand, mutations inthe SDHA (FAD-containing subunit) causethe mitochondrial encephalomyopathy Leighsyndrome (19) but not paraganglioma.

The striking difference in clinical effectsof SDHA subunit mutations versus SDHB,C, and D mutations strongly implicates mi-tochondrial ROS production in the etiologyof cancer. SDHA mutations would block theentry of electrons into complex II, thus deplet-ing ATP without increasing ROS production.The result is neuromuscular disease. By con-trast, mutations in subunits SDH B, C, and Dwould impede electron flux through complexII. This would increase ROS production, im-plicating ROS in cancer predisposition. Thisis supported by the mev-1 mutant in the C. ele-gans SDHC subunit, which has been shown toincrease mitochondrial ROS production (84,198).

A role for mitochondrial ROS in canceris supported by the observations that miceheterozygous for the MnSOD knock-out lo-cus have a 100% increase in cancer incidence(226), many types of tumors have reducedMnSOD, transformation of certain tumorswith the MnSOD cDNA can reverse the ma-lignant phenotype, and a cluster of three mu-tations in the MnSOD gene promoter regionthat alter AP-2 binding and promoter effi-ciency is found in a number of tumors (131,252). Moreover, ROS production, in associ-ation with the inactivation of p16ink4a, hasbeen hypothesized to be one of the two mainmechanisms for tumorigenesis; the other isp53 deficiency (10). Hence, it would appearthat mitochondrial ROS production, not ATPdepletion, is the important mitochondrial fac-tor in the etiology of cancer.

Mitochondrial DNA mutations that in-hibit OXPHOS and impede electron flowdown the ETC should increase ROS produc-tion and contribute to cancer. The first clearevidence that mtDNA mutations might playan important role in neoplastic transforma-

tion came with the report of a renal adenocar-cinoma that was heteroplasmic for a 294-npin-frame deletion in the mtDNA ND1 gene.This deletion generated a truncated mRNA,confirming that the mutant mtDNA resultedin functional mitochondrial defect (77). Sub-sequently, a variety of mtDNA-coding re-gion and CR mutations have been reportedin colon cancer cells (4, 66, 181), head andneck tumors (58), astrocytic tumors (95), thy-roid tumors (129, 253), breast tumors (209),and prostate tumors (33, 34, 38, 177).

Mitochondrial ROS could contribute toneoplastic transformation, both as a tu-mor initiator by causing nDNA mutationsin proto-oncogenes and tumor-suppressorgenes and as a tumor promoter through driv-ing cellular proliferation. At low levels, ROShas been found to be an active mitogen,thought to act though interaction with variouskinases (Src kinase, protein kinase C, MAPK,and receptor tyrosine kinases), as well as withdifferent transcription factors (Fos, Jun, NF-κB) (131). Furthermore, the dual functionapurinic/apyrimidinic endonuclease 1 (APE-1) functions not only in the DNA base excisionpathway but also in the redox regulation of thetranscription factors Fos, Jun, NFκB, PAX,HIF-1a, and p53 through the redox modifica-tion of its cysteine 310 (55, 91).

Further evidence of the importanceof mtDNA mutations and mitochondrialROS production in neoplastic transformationcomes from studies of complex IV (COX) mu-tations in prostate cancer. A proteomic surveyof prostate cancer epithelium cells isolated bylaser capture microscopy (LCM) revealed thatthe ratio of nDNA-encoded subunits (COXIV, Vb, and VIc) to mtDNA-encoded subunits(COI and II) was increased in most prostatetumors (72).

This alteration in the ratio of nDNA/mtDNA COX subunits in prostate cancer hasnow been shown to be due, in part, to mu-tations in the mtDNA COI gene (177). Ananalysis of the COI gene sequence from 260prostate cancer specimens, 54 prostate cancer-negative men over age 50, and 898 European

www.annualreviews.org • Mitochondrial & Age-Related Diseases 387

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 30: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

mtDNA sequences revealed that for Ameri-cans of European descent, COI mutations arefound in 11% of the prostate cancer speci-mens, 0% of the cancer-negative prostate con-trols, and 5.5% of the general population,all significant differences. Furthermore, fourprostate cancer COI mutations (T6253C,C6340T, G6261A, A6663G) (Figure 1) werefound in multiple independent patient tu-mors, often on different background mtDNAhaplotypes, and these changed highly con-served amino acids (CI = 69% to 97%)(177). Thus males who inherit COI mis-sense mutations from their mother are at agreatly increased risk for developing prostatecancer.

The average amino acid CI of the COI mu-tants found in prostate cancers was 83% ±25%, whereas that of the COI variants inthe general population was 71% ± 35%.This indicates that somatic mutations affect-ing more functionally conserved amino acidsalso contribute to the etiology of prostatecancer. This assessment was confirmed bythe observation that three of the COI mu-tations in prostate patients were heteroplas-mic: G5949A-G16X (Stop), T6124C-M74T(CI = 95%), C6924T-A341S (CI = 100%)(177). These data demonstrate that mutantCOI genes are prostate cancer oncogenes andthat both germline and somatic mtDNA mu-tations are important factors in this etiologyof prostate cancer.

A more complete assessment of the role ofmtDNA mutations in prostate cancer was ob-tained by sequencing the entire mtDNA fromthe tumorigenic epithelial cells, purified usingLCM, of the tumor harboring the COI chaintermination (G16X). This analysis revealedthree mtDNA variants: the heteroplasmicCOI G16X mutation, a homoplasmic ATP6C8932T (P136G) variant with a CI of 64%,and a homoplasmic cytb A14769G (N8S) vari-ant with a CI of 20.5%. Of these variants,the COI and ATP6 mutations are likely to befunctionally significant. The G16X mutationwas found to be homoplasmic in the cancercells and homoplasmic wild type in the ad-

jacent normal cells, which was confirmed byimmunohistochemical staining for the COIprotein. Therefore, this G16X mutation musthave arisen de novo at the time of neoplastictransformation and been strongly selected forin the cancerous cells (177).

The physiological significance of mtDNAmutations in cancer cells was demonstratedby introducing a known pathogenic mtDNAmutation into the prostate cancer cell linePC3 via transmitochondrial cybrid fusions(26, 240). PC3 cancer cells were cured oftheir resident mtDNAs by treatment withthe mitochondrial poison rhodamine 6G andthen fused with cytoplasts from either ho-moplasmic mutant (T8993G) or homoplas-mic wild-type (T8993T) cell lines (177) de-rived from a patient who was heteroplasmicfor the pathogenic ATP6 np T8993G L156Rmutation (75). This mutation reduces mito-chondrial ADP production (219) but also in-creases ROS production (128). A series of PC3cybrids were generated that were geneticallyidentical except for either a wild-type T or amutant G at np 8993 in the mtDNA.

Injection of these mutant and wild-typePC3 cybrids into nude mice gave a dramaticresult. The PC3 cells harboring the normalT8993T base barely grew at all. By contrast,the PC3 cybrids with the mutant T8993Gbase generated rapidly growing tumors thatkilled the mice (177). Staining the cellularnodules from the T8993T and T8993G cy-brid tumors with dihydroeithidium revealedthat the mutant T8993G tumors were makingmuch more ROS than the wild-type T8993Tnodules (177).

Consistent with the increased ROS pro-duction in the T8993G tumors, MnSOD hasbeen found to be reduced in precancerous in-traepithelial neoplastic lesions and in prostatecancer by immunohistochemical and in situhybridization studies (12, 18). Prostate cancercells also have a 42% increase in cytoplasmicstaining for the redox-sensitive APE-1 (90).Thus these observations support the conclu-sion that increased mitochondrial ROS pro-duction is central to prostate cancer.

388 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 31: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Therefore, mtDNA mutations appear toplay an important causal role in prostate can-cer, and thus may have a similar role in theetiology of other solid tumors. Moreover, therole of mitochondrial defects in the patho-physiology of cancer would appear to be thegeneration of increased ROS, which acts asboth an nDNA mutagen and cellular mitogen.Since mitochondrial mutations that increaseROS would do so by impeding the ETC, theywould also result in the accumulation withinthe cell of unoxidized NADH and pyruvate.The excess NADH and pyruvate would thenbe converted to lactate by lactate dehydroge-nase. This high production of lactate by can-cer cells in the presence of oxygen was firstobserved 70 years ago by Otto Warburg anddescribed as aerobic-glycolysis. Warburg hy-pothesized that mitochondrial defects mightunderlie many forms of cancer (246). Mod-ern molecular genetic analysis might yet provehim right.

Mitochondrial Medicine: AnIntegrated Mitochondrial Model forthe Etiology of DegenerativeDiseases, Aging, and Cancer

All of these observations on the pathophysi-ology of the common degenerative diseases,cancer, and aging reveal that these diverseclinical entities share many common underly-ing biochemical, gene regulation, and geneticfeatures. Moreover, these features consis-tently involve energy metabolism and the mi-tochondrion. Therefore, we can theorize thatall age-related diseases (degenerative diseases,cancer, and aging) share a common underly-ing mitochondrial pathophysiology which issummarized in Figure 6.

Dietary Restriction, DegenerativeDiseases, Longevity, and ClimaticAdaptation

In the energy-utilizing tissues (brain, heart,muscle, kidney, etc.), when calories (reduc-

Mitochondrialmedicine: the newmedical disciplinethat pertains to allclinical problemsthat involve themitochondria

ing equivalents) are plentiful and ATP isbeing rapidly hydrolyzed through physicalactivity, the mitochondrial electrochemicalgradient (�P) is kept depolarized as theATP synthase resynthesizes ATP. As a re-sult, electrons flow continuously throughthe ETC to 1/2O2 to sustain �P, keepingthe NADH/NAD+ ratio low, the ETC ox-idized and mitochondrial ROS productionminimized.

However, if calories are plentiful in tightlycoupled mitochondria and ATP utilization islow (sedentary life style), then �P becomesand remains maximized. This stalls the ETC,resulting in a high NADH/NAD+ ratio, ex-cess electrons in the ETC, and excessive ROSproduction. The increased ROS chronicallydamages the mitochondria, mtDNA, and themtPTP, ultimately resulting in the activa-tion of the mtPTP and cell death (Figure 6)(242).

High carbohydrate diets stimulate insulinsecretion that phosphorylates the FOXOs,removing them from the nucleus. This down-regulates the cellular stress-response path-ways including the mitochondrial and cytoso-lic antioxidant defenses and reduces the tran-scription of PGC-1α, thus down-regulatingmitochondrial OXPHOS. Additionally, thehigh NADH/NAD+ ratio inhibits SIRT1leaving the forkhead transcription factorsacetylated and out of the nucleus. The sup-pression of SIRT1 also permits p53 to be-come acetylated and fully active, increasingthe expression of BAX and the activation ofp66Shc. BAX binds to the mitochondrial outermembrane and activates the mtPTP to ini-tiate apoptosis. The activated mitochondrialcomponent of p66shc increases mitochondrialROS production and stimulates cytochrome crelease and apoptosis. This destructive trendis partially countered by the ROS-mediatedstimulation of the interaction of SIRT1 withthe forkheads, enhancing the deacetylationof forkheads and their import into the nu-cleus to activate the antioxidant and stress re-sponse systems. However, continued caloricoverload counters this protective effect. In

www.annualreviews.org • Mitochondrial & Age-Related Diseases 389

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 32: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

the end, high calorie diets lead to the down-regulation of mitochondrial OXPHOS andantioxidant and stress response defenses, in-creasing mitochondrial ROS production andthe accumulation of oxidative damage to themitochondria and mtDNA, leading to celldeath.

As an individual ages, the chronic ox-idative stress results in the accumulation ofsomatic mtDNA mutations, including re-arrangements and base substitutions in theregulatory elements of the CR and in the

structural genes. Ultimately, the decline ofthe mitochondrial biosynthetic capacity ren-ders the mitochondria unable to adequatelyrepair the accumulating mitochondrial ox-idative damage. This activates the mtPTP,p53, and p66Shc systems, increasing apopto-sis rates and resulting in cell loss, tissue dys-function, and the symptoms of age-relateddiseases.

These processes are ameliorated in indi-viduals harboring partially uncoupled mito-chondria. In loosely coupled mitochondria,

390 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 33: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

inefficient proton pumping and/or exces-sive proton leakage though the ATP syn-thase keeps �P submaximal, irrespective ofthe caloric availability. This keeps electronsflowing through the ETC and generatingheat, diminishes mitochondrial ROS produc-tion, decreases the rate of cellular apop-tosis, and thus increases longevity and in-hibits the progression of the age-relateddiseases.

The Mitochondrial Energetics andROS Biology of Cancer

These same factors also modulate the cel-lular predilection to neoplastic transforma-tion, cancer. Mutations in nDNA or mtDNAOXPHOS genes that impede the flow ofelectrons through the ETC increase mito-chondrial ROS production. The resultingH2O2, which is relatively stable, diffuses outof the mitochondrion, through the cytosol,and into the nucleus. In the nucleus, H2O2

can interact with transition metals and beconverted to •OH, which mutagenizes the

nDNA. The resulting nDNA damage acti-vates the nDNA repair systems, including thepolyADP-ribose polymerase (PARP). The ac-tivated PARP degrades the nuclear NAD+ inthe process of adding poly ADP-ribose chainsto histones and other nuclear proteins (242)(Figure 6).

The degradation of the nuclear NAD+,together with the high NADH/NAD+ ra-tio, inactivates nuclear SIRT1. SIRT1 is a hi-stone deacetylase, and nuclear transcriptionis repressed by the deacetylation of histonesand activated by histone acetylation. There-fore, inhibition of SIRT1 permits histoneacetylation to predominate, turning on thetranscription of normally inactive genes. Inpost-mitotic tissues, histone acetylation per-mits the activation of the genes that regulatecell replication and differentiation, the proto-oncogenes.

The diffusion of H2O2 into the nucleusand its conversion to •OH can then mu-tate the proto-oncogenes, converting theminto functional oncogenes. Moreover, in-creased cytosolic and nuclear H2O2 activates a

←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−Figure 6Model for the proposed role for mitochondrial dysfunction in an energy-utilization tissue cell inmetabolic and degenerative diseases, aging, and cancer. The mitochondrial pathophysiology of theseclinical entities is envisioned to result from the interplay between mitochondrial energy production,ROS generation, and the initiation of apoptosis through activation of the mtPTP. These components ofenergy metabolism are modulated by environmental constraints such as caloric availability and coldstress through the regulation of the FOXO and PGC-1α transcription factors and the SIRT1NAD+-dependent deacetylase. The FOXO transcription factors coordinately regulate mitochondrialenergy metabolism through PGC-1α as well as the antioxidant and stress response genes necessary tocope with the increased oxidative stress of oxidative metabolism. SIRT1 fine-tunes the interrelationshipbetween energy metabolism and apoptosis through the deacetylation of PGC-1α, p53, and the histoneproteins. Caloric overload or inhibition of OXPHOS perturbs the cellular mitochondrial energeticbalance resulting in increased ROS. The increased ROS and decreased mitochondrial energy outputsensitizes the mtPTP, ultimately driving the cell to apoptotic death. The increased ROS also diffusesinto the nucleus as H2O2 where it can mutate and activate proto-oncogenes (initiation) and can interactwith NFκB, APE-1 and various kinases to initiate cell division (promotion) leading to neoplastictransformation (cancer). Figure abbreviations are PARP, poly ADP-ribose polymerase; SIRT1, themammalian homologue to Sir2; FOXO, the mammalian forkhead transcription factor; P, aphosphorylated protein; Ac, an acetylated protein; IL, insulin ligand; ILL, insulin-like ligand; IR, insulinreceptor; ILR, insulin-like growth factor receptor; IRE, insulin response element; PI3K, the PI3 kinase;PI2, the membrane-bound phosphotidyl-inositol diphosphate; PI3, membrane-boundphosphatidyl-inositol triphosphate, AKT 1/2, the AKT kinases; ATPsynβ, ATP synthase β subunit;cytc, cytochrome c.

www.annualreviews.org • Mitochondrial & Age-Related Diseases 391

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 34: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

Evolutionarymedicine: a clinicalperspective thatposits that many ofthe common clinicalproblems of todayare rooted inadaptive geneticprograms thatpermitted ourhuman ancestors tosurvive in theenvironments whichthey confronted inthe past

variety of cellular signal transduction factorsincluding NFκB, APE-1, Fos, Jun, and tyro-sine kinases. This drives the cell into replica-tion. Consequently, mutations in mitochon-drial genes that inhibit electron flow throughthe ETC result in chronically increasedmitochondrial ROS production, which canact as both a tumor initiator (mutation ofproto-oncogenes) and tumor promoter (acti-vation of transcription and replication) (242)(Figure 6).

MITOCHONDRIALADAPTATION ANDEVOLUTIONARY MEDICINE

The Interplay betweenEnvironment and Energetics

These considerations indicate that mitochon-dria lie at the intersection between environ-mental factors such as calories and cold andthe human capacity to energetically cope withthe environmental challenges in different re-gions of the globe. Our ancient ancestors hadto be able to adapt to two classes of envi-ronmental changes: (a) short term changes incalories and climate associated with seasonalvariation and (b) long term changes in the na-ture of calories and average annual tempera-tures defined by the latitude and geographiczone in which they lived.

Aboriginal human populations living inthe colder temperate and arctic zones neededto adjust to acute cold and seasonal varia-tion in calories. Short-term adaptation to coldcan be accomplished by uncoupling mito-chondrial OXPHOS in both BAT and mus-cle by induction of the UCPs. Today, inindustrial societies, most individuals avoidcold stress through central heating. More-over, most ancient populations had to accu-mulate plant carbohydrate calories and storethem as fat during the plant growing sea-son and then to efficiently use the storedfat calories to sustain their cellular ener-getics when the plants were dormant. This

seasonal variation in caloric availability wasmetabolically managed via the insulin sig-naling network that coordinately integratedthe energy-utilization, energy-storage, andenergy-homeostasis tissues, through the hor-monal signals of the energy-sensing tissues,the pancreatic α and β cells. This was ac-complished by cueing the energy-sensing tis-sues to serum glucose concentration that os-cillated based on the availability of plantcarbohydrate calories. When plant calorieswere abundant, blood sugar was high, in-sulin was secreted, mitochondrial OXPHOSand its attendant antioxidant defenses weredown-regulated in the energy-utilization tis-sues, and the excess carbohydrate calorieswere stored as fat in energy-storage tissues.When plant calories declined at the end ofthe growing season, blood sugar levels de-clined, resulting in decreased insulin secre-tion and increased glucagon secretion. Thesehormonal changes up-regulated mitochon-drial OXPHOS and the attendant antioxi-dant defenses in the energy-utilization tissues,mobilized the stored fats in the energy-storage tissues for use as mitochondrial fuel,and activated glucose synthesis in the energy-homeostasis liver to sustain minimal bloodsugar levels.

This energetic system worked well for ourancestors who lived in an environment of pe-riodic carbohydrate surplus and deficiency.However, in today’s developed societies, tech-nology provides us with unlimited dietarycalories including carbohydrates throughoutthe year. As a result, our energy signalingsystem remains continuously in the high car-bohydrate state. Consequently, mitochondrialOXPHOS and its attendant antioxidant de-fenses are chronically down-regulated. At thesame time, our sedentary life style meansthat we do not turnover ATP through sus-tained physical activity. Therefore, the ex-cess of caloric reducing equivalents in ourdiet leads to fat accumulation and obesity andkeeps the mitochondrial electron carriers ofour down-regulated ETC fully reduced. This

392 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 35: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

results in chronically increased mitochondrialROS production, which damages the mito-chondria and mtDNAs. The reduced mito-chondrial energy output and the increasedoxidative stress sensitize the mtPTP to activa-tion, driving post-mitotic cells into prematureapoptosis. The resulting loss of post-mitoticcells results in the tissue-specific symptomsassociated with aging and the age-relateddiseases.

While the insulin and cAMP signal trans-duction systems permitted our ancestors torapidly adapt their metabolism to suddenchanges in environmental temperature andcarbohydrate calorie availability, these short-term oscillations were insufficient to copewith the more general environmental dif-ferences experienced by individuals living inthe tropics versus the temperate zone ver-sus the arctic. These different climatic zonesrequired adaptation to differences in aver-age ambient temperature, amount of sunlight,and nature of available calories. In the trop-ics the predominate source of calories wasplant carbohydrates, whereas in the arctic thecaloric availability shifted more toward animalprotein and fat.

Humans adapted to these more generalregional differences through the fixation offunctional mutations in their mtDNAs thatchanged the coupling efficiency of OXPHOS,thus shifting the energetic balance from pre-dominantly ATP production in the tropics toincreased heat production in the arctic. Func-tional mtDNA mutations undoubtedly per-mitted adaptation to many additional envi-ronmental factors as yet undiscovered. Whatis clear is that the random occurrence offunctional mtDNA variants shifted the en-ergetic balance in certain individuals suffi-ciently that they could move into and sur-vive in more northern environments fromwhich their warm-adapted predecessors wereexcluded. However, these adaptive mtDNAmutations required trade-offs. In the trop-ics, tightly coupled mitochondria maximizedthe efficiency of physical work and mini-

mized heat production. Mutations that par-tially uncoupled OXPHOS decreased workefficiency, but provided essential heat for sur-viving chronic cold. The uncoupling muta-tions also partially relieved the constraints ofthe inner membrane electrochemical gradient(�P) on electron flow through the ETC, per-mitting the ETC to run continuously. Thiskept the electron carriers oxidized and min-imized ROS production. Hence, in industri-alized societies where calories are unlimitedand exercise is minimized, individuals withuncoupled mitochondria (e.g., European hap-logroup J) are partially protected from theincreased oxidative stress associated with ex-cessive caloric intake. Therefore, these indi-viduals are on average less prone to degen-erative diseases and have increased longevity.Still, these individuals require more calories tosustain their tissue energetics. Hence, if calo-ries become limiting again, they will be thefirst to starve.

CONCLUSION

Evolutionary medicine posits that the geneticvariants that permitted our ancestors to adaptto diverse environments (niches) are havinga profound influence on our predispositionto common diseases today. The informationprovided in this review now places this con-cept on a solid experimental foundation andshows that the major environmental chal-lenges that confronted our hunter-gather an-cestors were bioenergetics for which the mi-tochondria adaptations were pivotal. Thus,the adaptive mtDNA mutations of our ances-tors, when combined with the environmentalcontrol and unlimited calories of our modernenvironment, are increasingly resulting inindividual energetic imbalances. It is theseimbalances between ancient mitochondrialgenetics and modern caloric intake that,I believe, are driving the modern epi-demics of obesity, diabetes, neurodegen-erative disease, cardiovascular disease, andcancer.

www.annualreviews.org • Mitochondrial & Age-Related Diseases 393

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 36: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

ACKNOWLEDGMENTS

The author would like to thank Ms. M.T. Lott for her assistance with this manuscript. Thiswork has been supported by NIH grants NS21328, AG13154, NS41650, AG24373, TW01366,HL64017, and an Ellison Senior Scholar Award.

LITERATURE CITED

1. Accili D, Arden KC. 2004. FoxOs at the crossroads of cellular metabolism, differentiation,and transformation. Cell 117:421–26

2. Adams JU. 2005. Autophagy and longevity. How keeping house may keep one young.Scientist 19:22–24

3. Adams V, Griffin L, Towbin J, Gelb B, Worley K, McCabe ER. 1991. Porin interac-tion with hexokinase and glycerol kinase: metabolic microcompartmentation at the outermitochondrial membrane. Biochem. Med. Metab. Biol. 45:271–91

4. Alonso A, Martin P, Albarran C, Aquilera B, Garcia O, et al. 1997. Detection of somaticmutations in the mitochondrial DNA control region of colorectal and gastric tumors byheteroduplex and single-strand conformation analysis. Electrophoresis 18:682–85

5. Altshuler D, Hirschhorn JN, Klannemark M, Lindgren CM, Vohl MC, et al. 2000. Thecommon PPARgamma Pro12Ala polymorphism is associated with decreased risk of type2 diabetes. Nat. Genet. 26:76–80

6. Alzheimer’s Association. 2005. Alzheimer’s Disease Statistics. http://www.alz.org/Resources/FactSheets/FSAlzheimerStats.pdf

7. Anandatheerthavarada HK, Biswas G, Robin MA, Avadhani NG. 2003. Mitochondrialtargeting and a novel transmembrane arrest of Alzheimer’s amyloid precursor proteinimpairs mitochondrial function in neuronal cells. J. Biol. Chem. 161:41–54

8. Andreu AL, Bruno C, Dunne TC, Tanji K, Shanske S, et al. 1999. A nonsense muta-tion (G15059A) in the cytochrome b gene in a patient with exercise intolerance andmyoglobinuria. Ann. Neurol. 45:127–30

9. Ankarcrona M, Hultenby K. 2002. Presenilin-1 is located in rat mitochondria. Biochem.Biophys. Res. Commun. 295:766–70

10. Arbiser JL. 2004. Molecular regulation of angiogenesis and tumorigenesis by signal trans-duction pathways: evidence of predictable and reproducible patterns of synergy in diverseneoplasms. Semin. Cancer Biol. 14:81–91

11. Astuti D, Latif F, Dallol A, Dahia PL, Douglas F, et al. 2001. Gene mutations in the suc-cinate dehydrogenase subunit SDHB cause susceptibility to familial pheochromocytomaand to familial paraganglioma. Am. J. Hum. Genet. 69:49–54

12. Baker AM, Oberley LW, Cohen MB. 1997. Expression of antioxidant enzymes in humanprostatic adenocarcinoma. Prostate 32:229–33

13. Ballinger SW, Shoffner JM, Gebhart S, Koontz DA, Wallace DC. 1994. Mitochondrialdiabetes revisited. Nat. Genet. 7:458–59

14. Ballinger SW, Shoffner JM, Hedaya EV, Trounce I, Polak MA, et al. 1992. Maternallytransmitted diabetes and deafness associated with a 10.4 kb mitochondrial DNA deletion.Nat. Genet. 1:11–15

15. Baysal BE, Ferrell RE, Willett-Brozick JE, Lawrence EC, Myssiorek D, et al. 2000.Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma.Science 287:848–51

16. Bluher M, Kahn BB, Kahn CR. 2003. Extended longevity in mice lacking the insulinreceptor in adipose tissue. Science 299:572–74

394 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 37: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

17. Bosetti F, Brizzi F, Barogi S, Mancuso M, Siciliano G, et al. 2002. Cytochrome c oxidaseand mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain frompatients with Alzheimer’s disease. Neurobiol. Aging 23:371–76

18. Bostwick DG, Alexander EE, Singh R, Shan A, Qian J, et al. 2000. Antioxidant enzymeexpression and reactive oxygen species damage in prostatic intraepithelial neoplasia andcancer. Cancer 89:123–34

19. Bourgeron T, Rustin P, Chretien D, Birch-Machin M, Bourgeois M, et al. 1995. Mutationof a nuclear succinate dehydrogenase gene results in mitochondrial respiratory chaindeficiency. Nat. Genet. 11:144–49

20. Brown MD, Starikovskaya YB, Derbeneva O, Hosseini S, Allen JC, et al. 2002. The roleof mtDNA background in disease expression: a new primary LHON mutation associatedwith Western Eurasian haplogroup J. Hum. Genet. 110:130–38

21. Brown MD, Sun F, Wallace DC. 1997. Clustering of Caucasian Leber hereditary opticneuropathy patients containing the 11778 or 14484 mutations on an mtDNA lineage.Am. J. Hum. Genet. 60:381–87

22. Brown MD, Trounce IA, Jun AS, Allen JC, Wallace DC. 2000. Functional analysis of lym-phoblast and cybrid mitochondria containing the 3460, 11778, or 14484 Leber’s Hered-itary Optic Neuropathy mtDNA mutation. J. Biol. Chem. 275:39831–36

23. Brown MD, Zhadanov S, Allen JC, Hosseini S, Newman NJ, et al. 2001. Novel mtDNAmutations and oxidative phosphorylation dysfunction in Russion LHON families. Hum.Genet. 109:33–39

24. Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, et al. 2004. Stress-dependentregulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303:2011–15

25. Bruno C, Martinuzzi A, Tang Y, Andreu AL, Pallotti F, et al. 1999. A stop-codon mutationin the human mtDNA cytochrome c oxidase I gene disrupts the functional structure ofcomplex IV. Am. J. Hum. Genet. 65:611–20

26. Bunn CL, Wallace DC, Eisenstadt JM. 1974. Cytoplasmic inheritance of chlormapheni-col resistance in mouse tissue culture cells. Proc. Natl. Acad. Sci. USA 71:1681–85

27. Canevari L, Clark JB, Bates TE. 1999. Beta-amyloid fragment 25–35 selectively decreasescomplex IV activity in isolated mitochondria. FEBS Lett. 457:131–34

28. Cann RL, Stoneking M, Wilson AC. 1987. Mitochondrial DNA and human evolution.Nature 325:31–36

29. Cardoso SM, Proenca MT, Santos S, Santana I, Oliveira CR. 2004. Cytochrome c oxidaseis decreased in Alzheimer’s disease platelets. Neurobiol. Aging 25:105–10

30. Castellani R, Hirai K, Aliev G, Drew KL, Nunomura A, et al. 2002. Role of mitochondrialdysfunction in Alzheimer’s disease. J. Neurosci. Res. 70:357–60

31. Cent. Dis. Control Prev. 2005. Prostate Cancer: The Public Health Perspective, 2004/2005Fact Sheet. http://www.cdc.gov/cancer/prostate/prospdf/about2004.pdf

32. Chagnon P, Gee M, Filion M, Robitaille Y, Belouchi M, Gauvreau D. 1999. Phylogeneticanalysis of the mitochondrial genome indicates significant differences between patientswith Alzheimer disease and controls in a French-Canadian founder population. Am. J.Med. Genet. 85:20–30

33. Chen JZ, Gokden N, Greene GF, Mukunyadzi P, Kadlubar FF. 2002. Extensive somaticmitochondrial mutations in primary prostate cancer using laser capture microdissection.Cancer Res. 62:6470–74

34. Chinnery PF, Samuels DC, Elson J, Turnbull DM. 2002. Accumulation of mitochon-drial DNA mutations in ageing, cancer, and mitochondrial disease: Is there a commonmechanism? Lancet 360:1323–25

www.annualreviews.org • Mitochondrial & Age-Related Diseases 395

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 38: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

35. Clancy DJ, Gems D, Hafen E, Leevers SJ, Partridge L. 2002. Dietary restriction inlong-lived dwarf flies. Science 296:319

36. Clancy DJ, Gems D, Harshman LG, Oldham S, Stocker H, et al. 2001. Extension oflife-span by loss of CHICO, a Drosophila insulin receptor substrate protein. Science292:104–6

37. Comi GP, Bordoni A, Salani S, Franceschina L, Sciacco M, et al. 1998. Cytochromec oxidase subunit I microdeletion in a patient with motor neuron disease. Ann. Neurol.43:110–16

38. Copeland WC, Wachsman JT, Johnson FM, Penta JS. 2002. Mitochondrial DNA alter-ations in cancer. Cancer Invest. 20:557–69

39. Corral-Debrinski M, Horton T, Lott MT, Shoffner JM, Beal MF, Wallace DC. 1992.Mitochondrial DNA deletions in human brain: regional variability and increase withadvanced age. Nat. Genet. 2:324–29

40. Corral-Debrinski M, Horton T, Lott MT, Shoffner JM, McKee AC, et al. 1994. Markedchanges in mitochondrial DNA deletion levels in Alzheimer brains. Genomics 23:471–76

41. Corral-Debrinski M, Shoffner JM, Lott MT, Wallace DC. 1992. Association of mito-chondrial DNA damage with aging and coronary atherosclerotic heart disease. Mutat.Res. 275:169–80

42. Corral-Debrinski M, Stepien G, Shoffner JM, Lott MT, Kanter K, Wallace DC. 1991.Hypoxemia is associated with mitochondrial DNA damage and gene induction. Implica-tions for cardiac disease. JAMA 266:1812–16

43. Cortopassi GA, Arnheim N. 1990. Detection of a specific mitochondrial DNA deletionin tissues of older humans. Nucleic Acids Res. 18:6927–33

44. Coskun PE, Ruiz-Pesini EE, Wallace DC. 2003. Control region mtDNA variants:longevity, climatic adaptation and a forensic conundrum. Proc. Natl. Acad. Sci. USA 100:2174–76

45. Cottrell DA, Borthwick GM, Johnson MA, Ince PG, Turnbull DM. 2002. The role ofcytochrome c oxidase deficient hippocampal neurones in Alzheimer’s disease. Neuropathol.Appl. Neurobiol. 28:390–96

46. Crouch PJ, Blake R, Duce JA, Ciccotosto GD, Li QX, et al. 2005. Copper-dependentinhibition of human cytochrome c oxidase by a dimeric conformer of amyloid-beta1-42.J. Neurosci. 25:672–79

47. Czubryt MP, McAnally J, Fishman GI, Olson EN. 2003. Regulation of peroxisomeproliferator-activated receptor gamma coactivator 1 alpha (PGC-1 alpha) and mi-tochondrial function by MEF2 and HDAC5. Proc. Natl. Acad. Sci. USA 100:1711–16

48. Daitoku H, Yamagata K, Matsuzaki H, Hatta M, Fukamizu A. 2003. Regulation of PGC-1 promoter activity by protein kinase B and the forkhead transcription factor FKHR.Diabetes 52:642–49

49. Das DK, George A, Liu XK, Rao PS. 1989. Detection of hydroxyl radical in the mitochon-dria of ischemic-reperfused myocardium by trapping with salicylate. Biochem. Biophys. Res.Commun. 165:1004–9

50. De Benedictis G, Rose G, Carrieri G, De Luca M, Falcone E, et al. 1999. MitochondrialDNA inherited variants are associated with successful aging and longevity in humans.FASEB J. 13:1532–36

51. DiMauro S, Schon EA. 2003. Mitochondrial respiratory-chain diseases. N. Engl. J. Med.348:2656–68

396 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 39: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

52. Dumoulin R, Sagnol I, Ferlin T, Bozon D, Stepien G, Mousson B. 1996. A novelgly290asp mitochondrial cytochrome b mutation linked to a complex III deficiency inprogressive exercise intolerance. Mol. Cell. Probes 10:389–91

53. Ek J, Andersen G, Urhammer SA, Gaede PH, Drivsholm T, et al. 2001. Mutation analy-sis of peroxisome proliferator-activated receptor-gamma coactivator-1 (PGC-1) and rela-tionships of identified amino acid polymorphisms to Type II diabetes mellitus. Diabetologia44:2220–26

54. Esposito LA, Melov S, Panov A, Cottrell BA, Wallace DC. 1999. Mitochondrial diseasein mouse results in increased oxidative stress. Proc. Natl. Acad. Sci. USA 96:4820–25

55. Evans AR, Limp-Foster M, Kelley MR. 2000. Going APE over ref-1. Mutat. Res. 461:83–108

56. Feng J, Bussiere F, Hekimi S. 2001. Mitochondrial electron transport is a key determinantof life span in Caenorhabditis elegans. Dev. Cell 1:633–44

57. Flanagan SW, Anderson RD, Ross MA, Oberley LW. 2002. Overexpression of manganesesuperoxide dismutase attenuates neuronal death in human cells expressing mutant (G37R)Cu/Zn-superoxide dismutase. J. Neurochem. 81:170–77

58. Fliss MS, Usadel H, Caballero OL, Wu L, Buta MR, et al. 2000. Facile detection ofmitochondrial DNA mutations in tumors and bodily fluids. Science 287:2017–19

59. Furuyama T, Kitayama K, Yamashita H, Mori N. 2003. Forkhead transcription factorFOXO1 (FKHR)-dependent induction of PDK4 gene expression in skeletal muscle dur-ing energy deprivation. Biochem. J. 375:365–71

60. Gelb BD, Adams V, Jones SN, Griffin LD, MacGregor GR, McCabe ER. 1992. Targetingof hexokinase 1 to liver and hepatoma mitochondria. Proc. Natl. Acad. Sci. USA 89:202–6

61. Giannakou ME, Partridge L. 2004. The interaction between FOXO and SIRT1: tippingthe balance towards survival. Trends Cell Biol. 14:408–12

62. Gibson GE, Huang HM. 2002. Oxidative processes in the brain and non-neuronal tissuesas biomarkers of Alzheimer’s disease. Front. Biosci. 7:d1007–d15

63. Giles RE, Blanc H, Cann HM, Wallace DC. 1980. Maternal inheritance of human mi-tochondrial DNA. Proc. Natl. Acad. Sci. USA 77:6715–19

64. Goto Y, Nonaka I, Horai S. 1990. A mutation in the tRNALeu(UUR) gene associated withthe MELAS subgroup of mitochondrial encephalomyopathies. Nature 348:651–53

65. Guarente L, Kenyon C. 2000. Genetic pathways that regulate ageing in model organisms.Nature 408:255–62

66. Habano W, Sugai T, Yoshida T, Nakamura S. 1999. Mitochondrial gene mutation, butnot large-scale deletion, is a feature of colorectal carcinomas with mitochondrial mi-crosatellite instability. Int. J. Cancer 83:625–29

67. Hamilton ML, Van Remmen H, Drake JA, Yang H, Guo ZM, et al. 2001. Does oxidativedamage to DNA increase with age? Proc. Natl. Acad. Sci. USA 98:10469–74

68. Hansson CA, Frykman S, Farmery MR, Tjernberg LO, Nilsberth C, et al. 2004. Nicas-trin, presenilin, APH-1 and PEN-2 form active gamma-secretase complexes in mito-chondria. J. Biol. Chem. 279:51654–60

69. Harrison DE, Archer JR. 1987. Genetic differences in effects of food restriction on agingin mice. J. Nutr. 117:376–82

70. Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. 2003. Alzheimer disease in theUS population: prevalence estimates using the 2000 census. Arch. Neurol. 60:1119–22

71. Hekimi S, Burgess J, Bussiere F, Meng Y, Benard C. 2001. Genetics of lifespan in C. ele-gans: molecular diversity, physiological complexity, mechanistic simplicity. Trends Genet.17:712–18

www.annualreviews.org • Mitochondrial & Age-Related Diseases 397

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 40: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

72. Herrmann PC, Gillespie JW, Charboneau L, Bichsel VE, Paweletz CP, et al. 2003.Mitochondrial proteome: altered cytochrome c oxidase subunit levels in prostate cancer.Proteomics 3:1801–10

73. Hogan P, Dall T, Nikolov P. 2003. Economic costs of diabetes in the US in 2002. DiabetesCare 26:917–32

74. Holt IJ, Harding AE, Morgan-Hughes JA. 1988. Deletions of muscle mitochondrial DNAin patients with mitochondrial myopathies. Nature 331:717–19

75. Holt IJ, Harding AE, Petty RK, Morgan-Hughes JA. 1990. A new mitochondrial diseaseassociated with mitochondrial DNA heteroplasmy. Am. J. Hum. Genet. 46:428–33

76. Horton TM, Graham BH, Corral-Debrinski M, Shoffner JM, Kaufman AE, et al.1995. Marked increase in mitochondrial DNA deletion levels in the cerebral cortex ofHuntington’s disease patients. Neurology 45:1879–83

77. Horton TM, Petros JA, Heddi A, Shoffner J, Kaufman AE, et al. 1996. Novel mitochon-drial DNA deletion found in a renal cell carcinoma. Genes Chromosomes Cancer 15:95–101

78. Houten SM, Auwerx J. 2004. PGC-1alpha: turbocharging mitochondria. Cell 119:5–779. Hsu LJ, Sagara Y, Arroyo A, Rockenstein E, Sisk A, et al. 2000. Alpha-synuclein promotes

mitochondrial deficit and oxidative stress. Am. J. Pathol. 157:401–1080. Huang X, Atwood CS, Hartshorn MA, Multhaup G, Goldstein LE, et al. 1999. The A

beta peptide of Alzheimer’s disease directly produces hydrogen peroxide through metalion reduction. Biochemistry 38:7609–16

81. Hutchin T, Cortopassi G. 1995. A mitochondrial DNA clone is associated with increasedrisk for Alzheimer disease. Proc. Natl. Acad. Sci. USA 92:6892–95

82. Imai S, Armstrong CM, Kaeberlein M, Guarente L. 2000. Transcriptional silencing andlongevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403:795–800

83. Ingman M, Kaessmann H, Paabo S, Gyllensten U. 2000. Mitochondrial genome variationand the origin of modern humans. Nature 408:708–13

84. Ishii N, Fujii M, Hartman PS, Tsuda M, Yasuda K, et al. 1998. A mutation in succinatedehydrogenase cytochrome b causes oxidative stress and ageing in nematodes. Nature 394:694–97

85. Jaarsma D, Rognoni F, van Duijn W, Verspaget HW, Haasdijk ED, Holstege JC.2001. CuZn superoxide dismutase (SOD1) accumulates in vacuolated mitochondria intransgenic mice expressing amyotrophic lateral sclerosis-linked SOD1 mutations. ActaNeuropathol. 102:293–305

86. Johnson MJ, Wallace DC, Ferris SD, Rattazzi MC, Cavalli-Sforza LL. 1983. Radiation ofhuman mitochondria DNA types analyzed by restriction endonuclease cleavage patterns.J. Mol. Evol. 19:255–71

87. Jonassen T, Larsen PL, Clarke CF. 2001. A dietary source of coenzyme Q is essential forgrowth of long-lived Caenorhabditis elegans clk-1 mutants. Proc. Natl. Acad. Sci. USA 98:421–26

88. Jun AS, Brown MD, Wallace DC. 1994. A mitochondrial DNA mutation at np 14459 ofthe ND6 gene associated with maternally inherited Leber’s hereditary optic neuropathyand dystonia. Proc. Natl. Acad. Sci. USA 91:6206–10

89. Kamei Y, Ohizumi H, Fujitani Y, Nemoto T, Tanaka T, et al. 2003. PPARgamma coac-tivator 1beta/ERR ligand 1 is an ERR protein ligand, whose expression induces a high-energy expenditure and antagonizes obesity. Proc. Natl. Acad. Sci. USA 100:12378–83

90. Kelley MR, Cheng L, Foster R, Tritt R, Jiang J, et al. 2001. Elevated and altered expressionof the multifunctional DNA base excision repair and redox enzyme Ape1/ref-1 in prostatecancer. Clin. Cancer Res. 7:824–30

398 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 41: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

91. Kelley MR, Parsons SH. 2001. Redox regulation of the DNA repair function of thehuman AP endonuclease Ape1/ref-1. Antioxid. Redox Signal. 3:671–83

92. Kelly DP, Scarpulla RC. 2004. Transcriptional regulatory circuits controlling mitochon-drial biogenesis and function. Genes Dev. 18:357–68

93. Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G. 1997. Daf-2, an insulin receptor-likegene that regulates longevity and diapause in Caenorhabditis elegans. Science 277:942–46

94. King MP, Attardi G. 1989. Human cells lacking mtDNA: repopulation with exogenousmitochondria by complementation. Science 246:500–3

95. Kirches E, Michael M, Woy C, Schneider T, Warich-Kirches M, et al. 1999. Loss ofheteroplasmy in the displacement loop of brain mitochondrial DNA in astrocytic tumors.Genes Chromosomes Cancer 26:80–83

96. Kish SJ, Mastrogiacomo F, Guttman M, Furukawa Y, Taanman JW, et al. 1999. De-creased brain protein levels of cytochrome oxidase subunits in Alzheimer’s disease and inhereditary spinocerebellar ataxia disorders: a nonspecific change? J. Neurochem. 72:700–7

97. Kokoszka JE, Coskun P, Esposito L, Wallace DC. 2001. Increased mitochondrial oxida-tive stress in the Sod2 (+/−) mouse results in the age-related decline of mitochondrialfunction culminating in increased apoptosis. Proc. Natl. Acad. Sci. USA 98:2278–83

98. Kokoszka JE, Waymire KG, Levy SE, Sligh JE, Cai J, et al. 2004. The ADP/ATP translo-cator is not essential for the mitochondrial permeability transition pore. Nature 427:461–65

99. Kontush A. 2001. Amyloid-beta: an antioxidant that becomes a pro-oxidant and criticallycontributes to Alzheimer’s disease. Free Radic. Biol. Med. 31:1120–31

100. Kontush A, Atwood CS. 2004. Amyloid-beta: phylogenesis of a chameleon. Brain Res.Brain Res. Rev. 46:118–20

101. Kontush A, Berndt C, Weber W, Akopyan V, Arlt S, et al. 2001. Amyloid-beta is an an-tioxidant for lipoproteins in cerebrospinal fluid and plasma. Free Radic. Biol. Med. 30:119–28

102. Koster JC, Marshall BA, Ensor N, Corbett JA, Nichols CG. 2000. Targeted overactivityof beta cell K(ATP) channels induces profound neonatal diabetes. Cell 100:645–54

103. Koutnikova H, Campuzano V, Foury F, Dolle P, Cazzalini O, Koenig M. 1997. Studiesof human, mouse and yeast homologues indicate a mitochondrial function for frataxin.Nat. Genet. 16:345–51

104. Langley E, Pearson M, Faretta M, Bauer UM, Frye RA, et al. 2002. Human SIR2 deacety-lates p53 and antagonizes PML/p53-induced cellular senescence. EMBO J. 21:2383–96

105. Lee CH, Olson P, Evans RM. 2003. Minireview: lipid metabolism, metabolic diseases,and peroxisome proliferator-activated receptors. Endocrinology 144:2201–7

106. Lee CK, Klopp RG, Weindruch R, Prolla TA. 1999. Gene expression profile of agingand its retardation by caloric restriction. Science 285:1390–93

107. Lee CK, Weindruch R, Prolla TA. 2000. Gene-expression profile of the ageing brain inmice. Nat. Genet. 25:294–97

108. Lee HJ, Shin SY, Choi C, Lee YH, Lee SJ. 2002. Formation and removal of alpha-synuclein aggregates in cells exposed to mitochondrial inhibitors. J. Biol. Chem. 277:5411–17

109. Lee SS, Kennedy S, Tolonen AC, Ruvkun G. 2003. DAF-16 target genes that control C.elegans life-span and metabolism. Science 300:644–4

110. Lee SS, Lee RY, Fraser AG, Kamath RS, Ahringer J, Ruvkun G. 2003. A systematicRNAi screen identifies a critical role for mitochondria in C. elegans longevity. Nat. Genet.33:40–48

www.annualreviews.org • Mitochondrial & Age-Related Diseases 399

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 42: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

111. Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM, Kelly DP. 2000. Peroxisomeproliferator-activated receptor gamma coactivator-1 promotes cardiac mitochondrial bio-genesis. J. Clin. Invest. 106:847–56

112. Lehtonen R, Kiuru M, Vanharanta S, Sjoberg J, Aaltonen LM, et al. 2004. Biallelicinactivation of fumarate hydratase (FH) occurs in nonsyndromic uterine leiomyomas butis rare in other tumors. Am. J. Pathol. 164:17–22

113. Leissring MA, Farris W, Wu X, Christodoulou DC, Haigis MC, et al. 2004. Alternativetranslation initiation generates a novel isoform of insulin-degrading enzyme targeted tomitochondria. Biochem. J. 383:439–46

114. Lemieux J, Lakowski B, Webb A, Meng Y, Ubach A, et al. 2001. Regulation of physio-logical rates in Caenorhabditis elegans by a tRNA-modifying enzyme in the mitochondria.Genetics 159:147–57

115. Li Y, Huang TT, Carlson EJ, Melov S, Ursell PC, et al. 1995. Dilated cardiomyopathyand neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat.Genet. 11:376–81

116. Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, et al. 1999. Induction of au-tophagy and inhibition of tumorigenesis by beclin 1. Nature 402:672–76

117. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, et al. 2002. Transcriptional co-activator PGC-1alpha drives the formation of slow-twitch muscle fibres. Nature 418:797–801

118. Lin J, Wu PH, Tarr PT, Lindenberg KS, St-Pierre J, et al. 2004. Defects in adaptive en-ergy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell 119:121–35

119. Lin SJ, Ford E, Haigis M, Liszt G, Guarente L. 2004. Calorie restriction extends yeastlife span by lowering the level of NADH. Genes Dev. 18:12–16

120. Lin SJ, Guarente L. 2003. Nicotinamide adenine dinucleotide, a metabolic regulator oftranscription, longevity and disease. Curr. Opin. Cell Biol. 15:241–46

121. Louet JF, Hayhurst G, Gonzalez FJ, Girard J, Decaux JF. 2002. The coactivator PGC-1 isinvolved in the regulation of the liver carnitine palmitoyltransferase I gene expression bycAMP in combination with HNF4 alpha and cAMP-response element-binding protein(CREB). J. Biol. Chem. 277:37991–8000

122. Lund J, Tedesco P, Duke K, Wang J, Kim SK, Johnson TE. 2002. Transcriptional profileof aging in C. elegans. Curr. Biol. 12:1566–73

123. Luo J, Nikolaev AY, Imai S, Chen D, Su F, et al. 2001. Negative control of p53 bySir2alpha promotes cell survival under stress. Cell 107:137–48

124. Lustbader JW, Cirilli M, Lin C, Xu HW, Takuma K, et al. 2004. ABAD directly linksAbeta to mitochondrial toxicity in Alzheimer’s disease. Science 304:448–52

125. Malaisse-Lagae F, Malaisse WJ. 1988. Hexose metabolism in pancreatic islets: regulationof mitochondrial hexokinase binding. Biochem. Med. Metab. Biol. 39:80–89

126. Masoro EJ. 1993. Dietary restriction and aging. J. Am. Geriatr. Soc. 41:994–99127. Masoro EJ, McCarter RJ, Katz MS, McMahan CA. 1992. Dietary restriction alters char-

acteristics of glucose fuel use. J. Gerontol. 47:B202–8. Erratum. 1993. J Gerontol. 48(2):B73128. Mattiazzi M, Vijayvergiya C, Gajewski CD, DeVivo DC, Lenaz G, et al. 2004. The

mtDNA T8993G (NARP) mutation results in an impairment of oxidative phosphoryla-tion that can be improved by antioxidants. Hum. Mol. Genet. 13:869–79

129. Maximo V, Soares P, Lima J, Cameselle-Teijeiro J, Sobrinho-Simoes M. 2002. Mitochon-drial DNA somatic mutations (point mutations and large deletions) and mitochondrialDNA variants in human thyroid pathology: a study with emphasis on Hurthle cell tumors.Am. J. Pathol. 160:1857–65

400 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 43: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

130. McCarter RJ, Palmer J. 1992. Energy metabolism and aging: a lifelong study of Fischer344 rats. Am. J. Physiol. 263:E448–52

131. McCord JM. 2000. The evolution of free radicals and oxidative stress. Am. J. Med. Genet.108:652–59

132. McDavid K, Lee J, Fulton J, Tonita J, Thompson T. 2004. Prostate cancer incidence andmortality rates and trends in the United States and Canada. Public Health Rep. 119:174–86

133. McMahon FJ, Chen YS, Patel S, Kokoszka J, Brown MD, et al. 2000. MitochondrialDNA sequence diversity in bipolar affective disorder. Am. J. Psychiatr. 157:1058–64

134. Mecocci P, MacGarvey U, Beal MF. 1994. Oxidative damage to mitochondrial DNA isincreased in Alzheimer’s disease. Ann. Neurol. 36:747–51

135. Mecocci P, MacGarvey U, Kaufman AE, Koontz D, Shoffner JM, et al. 1993. Oxidativedamage to mitochondrial DNA shows marked age-dependent increases in human brain.Ann. Neurol. 34:609–16

136. Melendez A, Talloczy Z, Seaman M, Eskelinen EL, Hall DH, Levine B. 2003. Autophagygenes are essential for dauer development and life-span extension in C. elegans. Science301:1387–91

137. Melov S, Coskun P, Patel M, Tunistra R, Cottrell B, et al. 1999. Mitochondrial diseasein superoxide dismutase 2 mutant mice. Proc. Natl. Acad. Sci. USA 96:846–51

138. Melov S, Doctrow SR, Schneider JA, Haberson J, Patel M, et al. 2001. Lifespan extensionand rescue of spongiform encephalopathy in superoxide dismutase 2 nullizygous micetreated with superoxide dismutase-catalase mimetics. J. Neurosci. 21:8348–53

139. Melov S, Hinerfeld D, Esposito L, Wallace DC. 1997. Multi-organ characterization ofmitochondrial genomic rearrangements in ad libitum and caloric restricted mice showstriking somatic mitochondrial DNA rearrangements with age. Nucleic Acids Res. 25:974–82

140. Melov S, Ravenscroft J, Malik S, Gill MS, Walker DW, et al. 2000. Extension of life-spanwith superoxide dismutase/catalase mimetics. Science 289:1567–69

141. Melov S, Schneider JA, Coskun PE, Bennett DA, Wallace DC. 1999. MitochondrialDNA rearrangements in aging human brain and in situ PCR of mtDNA. Neurobiol. Aging20:565–71

142. Melov S, Schneider JA, Day BJ, Hinerfeld D, Coskun P, et al. 1998. A novel neurologicalphenotype in mice lacking mitochondrial manganese superoxide dismutase. Nat. Genet.18:159–63

143. Melov S, Shoffner JM, Kaufman A, Wallace DC. 1995. Marked increase in the number andvariety of mitochondrial DNA rearrangements in aging human skeletal muscle. NucleicAcids Res. 23:4122–26. Erratum. 1995. Nucleic Acids Res. 23(23):4938

144. Merriwether DA, Clark AG, Ballinger SW, Schurr TG, Soodyall H, et al. 1991. Thestructure of human mitochondrial DNA variation. J. Mol. Evol. 33:543–55

145. Michikawa Y, Mazzucchelli F, Bresolin N, Scarlato G, Attardi G. 1999. Aging-dependentlarge accumulation of point mutations in the human mtDNA control region for replica-tion. Science 286:774–79

146. Migliaccio E, Giorgio M, Mele S, Pelicci G, Reboldi P, et al. 1999. The p66shc adaptorprotein controls oxidative stress response and life span in mammals. Nature 402:309–13

147. Mishmar D, Ruiz-Pesini E, Brandon M, Wallace DC. 2004. Mitochondrial DNA-likesequences in the nucleus (NUMTs): insights into our African origins and the mechanismof foreign DNA integration. Hum. Mutat. 23:125–33

148. Mishmar D, Ruiz-Pesini EE, Golik P, Macaulay V, Clark AG, et al. 2003. Natural selec-tion shaped regional mtDNA variation in humans. Proc. Natl. Acad. Sci. USA 100:171–76

www.annualreviews.org • Mitochondrial & Age-Related Diseases 401

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 44: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

149. MITOMAP. 2005. A Human Mitochondrial Genome Database. http://www.mitomap.org150. Miyadera H, Amino H, Hiraishi A, Taka H, Murayama K, et al. 2001. Altered quinone

biosynthesis in the long-lived clk-1 mutants of Caenorhabditis elegans. J. Biol. Chem. 276:7713–16

151. Miyata M, Smith JD. 1996. Apolipoprotein E allele-specific antioxidant activity and ef-fects on cytotoxicity by oxidative insults and beta-amyloid peptides. Nat. Genet. 14:55–61

152. Mootha VK, Handschin C, Arlow D, Xie X, St Pierre J, et al. 2004. Erralpha and Gabpa/bspecify PGC-1alpha-dependent oxidative phosphorylation gene expression that is alteredin diabetic muscle. Proc. Natl. Acad. Sci. USA 101:6570–75

153. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, et al. 2003. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately down-regulated in human diabetes. Nat. Genet. 34:267–73

154. Moraes CT, DiMauro S, Zeviani M, Lombes A, Shanske S, et al. 1989. MitochondrialDNA deletions in progressive external ophthalmoplegia and Kearns-Sayre syndrome. N.Engl. J. Med. 320:1293–99

155. Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, et al. 2004. Mammalian SIRT1represses forkhead transcription factors. Cell 116:551–63

156. Muller YL, Bogardus C, Pedersen O, Baier L. 2003. A Gly482Ser missense mutationin the peroxisome proliferator-activated receptor gamma coactivator-1 is associated withaltered lipid oxidation and early insulin secretion in Pima Indians. Diabetes 52:895–98

157. Murdock DG, Christacos NC, Wallace DC. 2000. The age-related accumulation of amitochondrial DNA control region mutation in muscle, but not brain, detected by asensitive PNA-directed PCR clamping based method. Nucleic Acids Res. 28:4350–55

158. Mutisya EM, Bowling AC, Beal MF. 1994. Cortical cytochrome oxidase activity is reducedin Alzheimer’s disease. J. Neurochem. 63:2179–84

159. Nagy Z, Esiri MM, LeGris M, Matthews PM. 1999. Mitochondrial enzyme expression inthe hippocampus in relation to Alzheimer-type pathology. Acta Neuropathol. 97:346–54

160. Natl. Parkinson Found. 2004. About Parkinson Disease. http://www.parkinson.org/site/pp.asp?c= 9dJFJLPwB&b=71125

161. Nemoto S, Fergusson MM, Finkel T. 2005. SIRT1 functionally interacts with themetabolic regulator and transcriptional coactivator PGC-1α. J. Biol. Chem. 280:16456–60

162. Niemann S, Muller U. 2000. Mutations in SDHC cause autosomal dominant paragan-glioma, type 3. Nat. Genet. 26:268–70

163. Niemi AK, Hervonen A, Hurme M, Karhunen PJ, Jylha M, Majamaa K. 2003. Mito-chondrial DNA polymorphisms associated with longevity in a Finnish population. Hum.Genet. 112:29–33

164. Obeso JA, Olanow CW, Nutt JG. 2000. Levodopa motor complications in Parkinson’sdisease. Trends Neurosci. 23:S2–7

165. Ogg S, Paradis S, Gottlieb S, Patterson GI, Lee L, et al. 1997. The Fork head transcriptionfactor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature389:994–99

166. Okado-Matsumoto A, Fridovich I. 2001. Subcellular distribution of superoxide dismu-tases (SOD) in rat liver: Cu,Zn-SOD in mitochondria. J. Biol. Chem. 276:38388–93

167. Okado-Matsumoto A, Fridovich I. 2002. Amyotrophic lateral sclerosis: a proposed mech-anism. Proc. Natl. Acad. Sci. USA 99:9010–14

168. Orr WC, Sohal RS. 1994. Extension of life-span by overexpression of superoxide dismu-tase and catalase in Drosophila melanogaster. Science 263:1128–30

402 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 45: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

169. Orsini F, Migliaccio E, Moroni M, Contursi C, Raker VA, et al. 2004. The life spandeterminant p66Shc localizes to mitochondria where it associates with mitochondrialheat shock protein 70 and regulates trans-membrane potential. J. Biol. Chem. 279:25689–95

170. Orth M, Tabrizi SJ, Schapira AH, Cooper JM. 2003. Alpha-synuclein expression inHEK293 cells enhances the mitochondrial sensitivity to rotenone. Neurosci. Lett. 351:29–32

171. Palacino JJ, Sagi D, Goldberg MS, Krauss S, Motz C, et al. 2004. Mitochondrial dys-function and oxidative damage in parkin-deficient mice. J. Biol. Chem. 279:18614–22

172. Panov AV, Gutekunst CA, Leavitt BR, Hayden MR, Burke JR, et al. 2002. Early mito-chondrial calcium defects in Huntington’s disease are a direct effect of polyglutamines.Nat. Neurosci. 5:731–36

173. Parkes TL, Elia AJ, Dickinson D, Hilliker AJ, Phillips JP, Boulianne GL. 1998. Extensionof Drosophila lifespan by overexpression of human SOD1 in motorneurons. Nat. Genet.19:171–74

174. Patrick L. 2002. Nonalcoholic fatty liver disease: relationship to insulin sensitivity andoxidative stress. Treatment approaches using vitamin E, magnesium, and betaine. Alt.Med. Rev. 7:276–91

175. Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, et al. 2003. Coordinated reduction ofgenes of oxidative metabolism in humans with insulin resistance and diabetes: potentialrole of PGC1 and NRF1. Proc. Natl. Acad. Sci. USA 100:8466–71

176. Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI. 2004. Impaired mitochondrialactivity in the insulin-resistant offspring of patients with type 2 diabetes. N. Engl. J. Med.350:664–71

177. Petros JA, Baumann AK, Ruiz-Pesini E, Amin MB, Sun CQ, et al. 2005. mtDNA muta-tions increase tumorigenicity in prostate cancer. Proc. Natl. Acad. Sci. USA 102:719–24

178. Picard F, Guarente L. 2005. Molecular links between aging and adipose tissue. Int. J.Obes. Relat. Metab. Disord. 29(Suppl. 1):S36–39

179. Picard F, Kurtev M, Chung N, Topark-Ngarm A, Senawong T, et al. 2004. Sirt1 promotesfat mobilization in white adipocytes by repressing PPAR-gamma. Nature 429:771–76

180. Pletcher SD, Macdonald SJ, Marguerie R, Certa U, Stearns SC, et al. 2002. Genome-wide transcript profiles in aging and calorically restricted Drosophila melanogaster. Curr.Biol. 12:712–23

181. Polyak K, Li Y, Zhu H, Lengauer C, Willson JK, et al. 1998. Somatic mutations of themitochondrial genome in human colorectal tumours. Nat. Genet. 20:291–93

182. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, et al. 1997. Mutation in thealpha-synuclein gene identified in families with Parkinson’s disease. Science 276:2045–47

183. Pratico D. 2002. Alzheimer’s disease and oxygen radicals: new insights. Biochem.Pharmacol. 63:563–67

184. Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM. 1998. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92:829–39

185. Ramassamy C, Averill D, Beffert U, Theroux L, Lussier-Cacan S, et al. 2000. Oxida-tive insults are associated with apolipoprotein E genotype in Alzheimer’s disease brain.Neurobiol. Dis. 7:23–37

186. Ramassamy C, Krzywkowski P, Averill D, Lussier-Cacan S, Theroux L, et al. 2001. Impactof apoE deficiency on oxidative insults and antioxidant levels in the brain. Brain Res. Mol.Brain Res. 86:76–83

www.annualreviews.org • Mitochondrial & Age-Related Diseases 403

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 46: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

187. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. 2005. Nutrientcontrol of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature434:113–18

188. Rogina B, Reenan RA, Nilsen SP, Helfand SL. 2000. Extended life-span conferred bycotransporter gene mutations in Drosophila. Science 290:2137–40

189. Rose G, Passarino G, Carrieri G, Altomare K, Greco V, et al. 2001. Paradoxes in longevity:sequence analysis of mtDNA haplogroup J in centenarians. Eur. J. Hum. Genet. 9:701–7

190. Roses AD, Einstein G, Gilbert J, Goedert M, Han SH, et al. 1996. Morphological, bio-chemical, and genetic support for an apolipoprotein E effect on microtubular metabolism.Ann. NY Acad. Sci. 777:146–57

191. Ross OA, McCormack R, Curran MD, Duguid RA, Barnett YA, et al. 2001. MitochondrialDNA polymorphism: its role in longevity of the Irish population. Exp. Gerontol. 36:1161–78

192. Rotig A, de Lonlay P, Chretien D, Foury F, Koenig M, et al. 1997. Aconitase and mi-tochondrial iron-sulphur protein deficiency in Friedreich ataxia. Nat. Genet. 17:215–17

193. Ruiz-Pesini E, Diez C, Lapena AC, Perez-Martos A, Montoya J, et al. 1998. Correlationof sperm motility with mitochondrial enzymatic activities. Clin. Chem. 44:1616–20

194. Ruiz-Pesini E, Mishmar D, Brandon M, Procaccio V, Wallace DC. 2004. Effects ofpurifying and adaptive selection on regional variation in human mtDNA. Science 303:223–26

195. Sawa A, Wiegand GW, Cooper J, Margolis RL, Sharp AH, et al. 1999. Increased apoptosisof Huntington disease lymphoblasts associated with repeat length-dependent mitochon-drial depolarization. Nat. Med. 5:1194–98

196. Scarpulla RC. 2002. Nuclear activators and coactivators in mammalian mitochondrialbiogenesis. Biochim. Biophys. Acta. 1576:1–14

197. Schriner SE, Linford NJ, Martin GM, Treuting P, Ogburn CE, et al. 2005. Extension ofmurine life span by overexpression of catalase targeted to mitochondria. Science 308:1909–11

198. Senoo-Matsuda N, Yasuda K, Tsuda M, Ohkubo T, Yoshimura S, et al. 2001. A defectin the cytochrome b large subunit in complex II causes both superoxide anion overpro-duction and abnormal energy metabolism in Caenorhabditis elegans. J. Biol. Chem. 276:41553–58

199. Shoffner JM, Brown MD, Torroni A, Lott MT, Cabell MR, et al. 1993. MitochondrialDNA variants observed in Alzheimer disease and Parkinson disease patients. Genomics17:171–84

200. Shoffner JM, Lott MT, Lezza AM, Seibel P, Ballinger SW, Wallace DC. 1990. Myoclonicepilepsy and ragged-red fiber disease (MERRF) is associated with a mitochondrial DNAtRNALys mutation. Cell 61:931–37

201. Silva JP, Kohler M, Graff C, Oldfors A, Magnuson MA, et al. 2000. Impaired insulinsecretion and beta-cell loss in tissue-specific knockout mice with mitochondrial diabetes.Nat. Genet. 26:336–40

202. Simon DK, Pulst SM, Sutton JP, Browne SE, Beal MF, Johns DR. 1999. Familial mul-tisystem degeneration with parkinsonism associated with the 11778 mitochondrial DNAmutation. Neurology 53:1787–93

203. Smith MA, Casadesus G, Joseph JA, Perry G. 2002. Amyloid-beta and tau serve an-tioxidant functions in the aging and Alzheimer brain. Free Radic. Biol. Med. 33:1194–99

404 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 47: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

204. Sohal RS, Ku HH, Agarwal S, Forster MJ, Lal H. 1994. Oxidative damage, mitochon-drial oxidant generation and antioxidant defenses during aging and in response to foodrestriction in the mouse. Mech. Ageing Dev. 74:121–33

205. Solano A, Roig M, Vives-Bauza C, Hernandez-Pena J, Garcia-Arumi E, et al. 2003.Bilateral striatal necrosis associated with a novel mutation in the mitochondrial ND6gene. Ann. Neurol. 54:527–30

206. Soong NW, Hinton DR, Cortopassi G, Arnheim N. 1992. Mosaicism for a specificsomatic mitochondrial DNA mutation in adult human brain. Nat. Genet. 2:318–23

207. Sturtz LA, Diekert K, Jensen LT, Lill R, Culotta VC. 2001. A fraction of yeast Cu,Zn-superoxide dismutase and its metallochaperone, CCS, localize to the intermembranespace of mitochondria. A physiological role for SOD1 in guarding against mitochondrialoxidative damage. J. Biol. Chem. 276:38084–89

208. Sun J, Folk D, Bradley TJ, Tower J. 2002. Induced overexpression of mitochondrialMn-superoxide dismutase extends the life span of adult Drosophila melanogaster. Genetics161:661–72

209. Tan DJ, Bai RK, Wong LJ. 2002. Comprehensive scanning of somatic mitochondrialDNA mutations in breast cancer. Cancer Res. 62:972–76

210. Tanaka M, Gong J, Zhang J, Yamada Y, Borgeld HJ, Yagi K. 2000. Mitochondrial genotypeassociated with longevity and its inhibitory effect on mutagenesis. Mech. Ageing Dev.116:65–76

211. Tanaka M, Gong JS, Zhang J, Yoneda M, Yagi K. 1998. Mitochondrial genotype associatedwith longevity. Lancet 351:185–86

212. Tandon A, Rogaeva E, Mullan M, St George-Hyslop PH. 2000. Molecular genetics ofAlzheimer’s disease: the role of beta-amyloid and the presenilins. Curr. Opin. Neurol.13:377–84

213. Tatuch Y, Christodoulou J, Feigenbaum A, Clarke JTR, Wherret J, et al. 1992. Hetero-plasmic mtDNA mutation (T-G) at 8993 can cause Leigh disease when the percentageof abnormal mtDNA is high. Am. J. Hum. Genet. 50:852–58

214. Tissenbaum HA, Guarente L. 2001. Increased dosage of a sir-2 gene extends lifespan inCaenorhabditis elegans. Nature 410:227–30

215. Tissenbaum HA, Guarente L. 2002. Model organisms as a guide to mammalian aging.Dev. Cell 2:9–19

216. Trifunovic A, Wredenberg A, Falkenberg M, Spelbrink JN, Rovio AT, et al. 2004. Pre-mature ageing in mice expressing defective mitochondrial DNA polymerase. Nature429:417–23

217. Trimmer PA, Keeney PM, Borland MK, Simon FA, Almeida J, et al. 2004. Mitochondrialabnormalities in cybrid cell models of sporadic Alzheimer’s disease worsen with passagein culture. Neurobiol. Dis. 15:29–39

218. Trinei M, Giorgio M, Cicalese A, Barozzi S, Ventura A, et al. 2002. A p53-p66Shc sig-nalling pathway controls intracellular redox status, levels of oxidation-damaged DNA andoxidative stress-induced apoptosis. Oncogene 21:3872–78

219. Trounce I, Neill S, Wallace DC. 1994. Cytoplasmic transfer of the mtDNA nt 8993 TG(ATP6) point mutation associated with Leigh syndrome into mtDNA-less cells demon-strates cosegregation with a decrease in state III respiration and ADP/O ratio. Proc. Natl.Acad. Sci. USA 91:8334–38

220. Trounce IA, Kim YL, Jun AS, Wallace DC. 1996. Assessment of mitochondrial oxidativephosphorylation in patient muscle biopsies, lymphoblasts, and transmitochondrial celllines. Methods Enzymol. 264:484–509

www.annualreviews.org • Mitochondrial & Age-Related Diseases 405

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 48: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

221. Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, et al. 2004. Hered-itary early-onset Parkinson’s disease caused by mutations in PINK1. Science 304:1158–60

222. Valla J, Berndt JD, Gonzalez-Lima F. 2001. Energy hypometabolism in posterior cin-gulate cortex of Alzheimer’s patients: superficial laminar cytochrome oxidase associatedwith disease duration. J. Neurosci. 21:4923–30

223. van den Ouweland JM, Lemkes HH, Trembath RC, Ross R, Velho G, et al. 1994. Mater-nally inherited diabetes and deafness is a distinct subtype of diabetes and associates witha single point mutation in the mitochondrial tRNALeu(UUR) gene. Diabetes 43:746–51

224. van der Walt JM, Dementieva YA, Martin ER, Scott WK, Nicodemus KK, et al. 2004.Analysis of European mitochondrial haplogroups with Alzheimer disease risk. Neurosci.Lett. 365:28–32

225. van der Walt JM, Nicodemus KK, Martin ER, Scott WK, Nance MA, et al. 2003. Mito-chondrial polymorphisms significantly reduce the risk of Parkinson disease. Am. J. Hum.Genet. 72:804–11

226. Van Remmen H, Ikeno Y, Hamilton M, Pahlavani M, Wolf N, et al. 2003. Life-longreduction in MnSOD activity results in increased DNA damage and higher incidence ofcancer but does not accelerate aging. Physiol. Genomics 16:29–37

227. Vanharanta S, Buchta M, McWhinney SR, Virta SK, Peczkowska M, et al. 2004. Early-onset renal cell carcinoma as a novel extraparaganglial component of SDHB-associatedheritable paraganglioma. Am. J. Hum. Genet. 74:153–59

228. Vaughan JR, Davis MB, Wood NW. 2001. Genetics of Parkinsonism: a review. Ann.Hum. Genet 65:111–26

229. Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, et al. 2001. hSIR2(SIRT1) functionsas an NAD-dependent p53 deacetylase. Cell 107:149–59

230. Vega RB, Huss JM, Kelly DP. 2000. The coactivator PGC-1 cooperates with peroxisomeproliferator-activated receptor alpha in transcriptional control of nuclear genes encodingmitochondrial fatty acid oxidation enzymes. Mol. Cell. Biol. 20:1868–76

231. Velho G, Froguel P. 1998. Genetic, metabolic and clinical characteristics of maturityonset diabetes of the young. Eur. J. Endocrinol. 138:233–39

232. Verwer RW, Jansen KA, Sluiter AA, Pool CW, Kamphorst W, Swaab DF. 2000. De-creased hippocampal metabolic activity in Alzheimer patients is not reflected in the im-munoreactivity of cytochrome oxidase subunits. Exp. Neurol. 163:440–51

233. Wallace DC. 1982. Structure and evolution of organelle genomes. Microbiol. Rev. 46:208–40

234. Wallace DC. 1992. Diseases of the mitochondrial DNA. Annu. Rev. Biochem. 61:1175–212

235. Wallace DC. 1992. Mitochondrial genetics: a paradigm for aging and degenerative dis-eases? Science 256:628–32

236. Wallace DC. 1997. Mitochondrial DNA mutations and bioenergetic defects in aging anddegenerative diseases. In The Molecular and Genetic Basis of Neurological Disease, ed. RNRosenberg, SB Prusiner, S DiMauro, RL Barchi, pp. 237–69. Boston: Butterworth-Heinemann

237. Wallace DC. 1999. Mitochondrial diseases in man and mouse. Science 283:1482–88238. Wallace DC. 2001. Mouse models for mitochondrial disease. Am. J. Med. Genet. 106:71–

93239. Wallace DC, Brown MD, Lott MT. 1999. Mitochondrial DNA variation in human evo-

lution and disease. Gene 238:211–30

406 Wallace

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 49: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

ANRV260-GE39-17 ARI 15 October 2005 13:1

240. Wallace DC, Bunn CL, Eisenstadt JM. 1975. Cytoplasmic transfer of chloramphenicolresistance in human tissue culture cells. J. Cell Biol. 67:174–88

241. Wallace DC, Lott MT. 2002. Mitochondrial genes in degenerative diseases, cancer andaging. In Emery and Rimoin’s Principles and Practice of Medical Genetics, ed. DL Rimoin, JMConnor, RE Pyeritz, BR Korf, pp. 299–409. London: Churchill Livingstone

242. Wallace DC, Lott MT, Brown MD, Kerstann K. 2001. Mitochondria and neuro-ophthalmological diseases. In The Metabolic and Molecular Basis of Inherited Disease, ed.CR Scriver, AL Beaudet, WS Sly, D Valle, pp. 2425–512. New York: McGraw-Hill

243. Wallace DC, Singh G, Lott MT, Hodge JA, Schurr TG, et al. 1988. Mitochondrial DNAmutation associated with Leber’s hereditary optic neuropathy. Science 242:1427–30

244. Wallace DC, Zheng X, Lott MT, Shoffner JM, Hodge JA, et al. 1988. Familial mito-chondrial encephalomyopathy (MERRF): genetic, pathophysiological, and biochemicalcharacterization of a mitochondrial DNA disease. Cell 55:601–10

245. Wang Y, Michikawa Y, Mallidis C, Bai Y, Woodhouse L, et al. 2001. Muscle-specificmutations accumulate with aging in critical human mtDNA control sites for replication.Proc. Natl. Acad. Sci. USA 98:4022–27

246. Warburg O. 1931. The Metabolism of Tumors. New York: R.R. Smith247. Warita H, Hayashi T, Murakami T, Manabe Y, Abe K. 2001. Oxidative damage to mi-

tochondrial DNA in spinal motoneurons of transgenic ALS mice. Brain Res. Mol. BrainRes. 89:147–52

248. Wilson FH, Hariri A, Farhi A, Zhao H, Petersen KF, et al. 2004. A cluster of metabolicdefects caused by mutation in a mitochondrial tRNA. Science 306:1190–94

249. Wilson RB, Roof DM. 1997. Respiratory deficiency due to loss of mitochondrial DNAin yeast lacking the frataxin homologue. Nat. Genet. 16:352–57

250. Wollheim CB. 2000. Beta-cell mitochondria in the regulation of insulin secretion: a newculprit in type II diabetes. Diabetologia 43:265–77

251. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, et al. 1999. Mechanisms con-trolling mitochondrial biogenesis and respiration through the thermogenic coactivatorPGC-1. Cell 98:115–24

252. Xu Y, Krishnan A, Wan XS, Majima H, Yeh CC, et al. 1999. Mutations in the promoterreveal a cause for the reduced expression of the human manganese superoxide dismutasegene in cancer cells. Oncogene 18:93–102

253. Yeh JJ, Lunetta KL, van Orsouw NJ, Moore FD Jr, Mutter GL, et al. 2000. Somaticmitochondrial DNA (mtDNA) mutations in papillary thyroid carcinomas and differentialmtDNA sequence variants in cases with thyroid tumours. Oncogene 19:2060–66

254. Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, et al. 2001. Control of hepaticgluconeogenesis through the transcriptional coactivator PGC-1. Nature 413:131–38

255. Yoon JC, Xu G, Deeney JT, Yang SN, Rhee J, et al. 2003. Suppression of beta cell energymetabolism and insulin release by PGC-1alpha. Dev. Cell 5:73–83

256. Zhang J, Asin-Cayuela J, Fish J, Michikawa Y, Bonafe M, et al. 2003. Strikingly higher fre-quency in centenarians and twins of mtDNA mutation causing remodeling of replicationorigin in leukocytes. Proc. Natl. Acad. Sci. USA 100:1116–21

257. Zimmet P, Alberti KG, Shaw J. 2001. Global and societal implications of the diabetesepidemic. Nature 414:782–87

www.annualreviews.org • Mitochondrial & Age-Related Diseases 407

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 50: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

Contents ARI 20 October 2005 19:18

Annual Review ofGenetics

Volume 39, 2005Contents

John Maynard SmithRichard E. Michod � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 1

The Genetics of Hearing and Balance in ZebrafishTeresa Nicolson � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 9

Immunoglobulin Gene DiversificationNancy Maizels � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �23

Complexity in Regulation of Tryptophan Biosynthesis inBacillus subtilisPaul Gollnick, Paul Babitzke, Alfred Antson, and Charles Yanofsky � � � � � � � � � � � � � � � � � � � � � � �47

Cell-Cycle Control of Gene Expression in Budding and Fission YeastJürg Bahler � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �69

Comparative Developmental Genetics and the Evolution of ArthropodBody PlansDavid R. Angelini and Thomas C. Kaufman � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �95

Concerted and Birth-and-Death Evolution of Multigene FamiliesMasatoshi Nei and Alejandro P. Rooney � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 121

Drosophila as a Model for Human Neurodegenerative DiseaseJulide Bilen and Nancy M. Bonini � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 153

Molecular Mechanisms of Germline Stem Cell RegulationMarco D. Wong, Zhigang Jin, and Ting Xie � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 173

Molecular Signatures of Natural SelectionRasmus Nielsen � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 197

T-Box Genes in Vertebrate DevelopmentL.A. Naiche, Zachary Harrelson, Robert G. Kelly, and Virginia E. Papaioannou � � � � � � 219

Connecting Mammalian Genome with Phenome by ENU MouseMutagenesis: Gene Combinations Specifying the Immune SystemPeter Papathanasiou and Christopher C. Goodnow � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 241

Evolutionary Genetics of Reproductive Behavior in Drosophila:Connecting the DotsPatrick M. O’Grady and Therese Anne Markow � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 263

v

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.

Page 51: A MITOCHONDRIAL PARADIGM OF METABOLIC AND …people.musc.edu/~rosenzsa/Spyropoulos/MitoMetabonAging.pdfA Mitochondrial Paradigm of Metabolic and Degenerative Diseases, Aging, and Cancer:

Contents ARI 20 October 2005 19:18

Sex Determination in the Teleost Medaka, Oryzias latipesMasura Matsuda � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 293

Orthologs, Paralogs, and Evolutionary GenomicsEugene V. Koonin � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 309

The Moss Physcomitrella patensDavid Cove � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 339

A Mitochondrial Paradigm of Metabolic and Degenerative Diseases,Aging, and Cancer: A Dawn for Evolutionary MedicineDouglas C. Wallace � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 359

Switches in Bacteriophage Lambda DevelopmentAmos B. Oppenheim, Oren Kobiler, Joel Stavans, Donald L. Court,

and Sankar Adhya � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 409

Nonhomologous End Joining in YeastJames M. Daley, Phillip L. Palmbos, Dongliang Wu, and Thomas E. Wilson � � � � � � � � � � 431

Plasmid Segregation MechanismsGitte Ebersbach and Kenn Gerdes � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 453

Use of the Zebrafish System to Study Primitive and DefinitiveHematopoiesisJill L.O. de Jong and Leonard I. Zon � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 481

Mitochondrial Morphology and Dynamics in Yeast and MulticellularEukaryotesKoji Okamoto and Janet M. Shaw � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 503

RNA-Guided DNA Deletion in Tetrahymena: An RNAi-BasedMechanism for Programmed Genome RearrangementsMeng-Chao Yao and Ju-Lan Chao � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 537

Molecular Genetics of Axis Formation in ZebrafishAlexander F. Schier and William S. Talbot � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 561

Chromatin Remodeling in Dosage CompensationJohn C. Lucchesi, William G. Kelly, and Barbara Panning � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 615

INDEXES

Subject Index � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 653

ERRATA

An online log of corrections to Annual Review of Geneticschapters may be found at http://genet.annualreviews.org/errata.shtml

vi Contents

Ann

u. R

ev. G

enet

. 200

5.39

:359

-407

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Med

ical

Uni

vers

ity o

f So

uth

Car

olin

a -

LIB

RA

RIE

S &

LE

AR

NIN

G R

ES

CT

R o

n 08

/25/

10. F

or p

erso

nal u

se o

nly.