Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Mitochondrial Biogenesis through Activation of Nuclear Signaling Proteins

John E. Dominy and Pere Puigserver

Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02215 Correspondence: [email protected]

The dynamics of mitochondrial biogenesis and function is a complex interplayof cellular and molecular processes that ultimately shape bioenergetics capacity. Mitochondrial mass, by itself, represents the net balance between rates of biogenesis and degradation. Mitochondrial biogenesis is dependent on different signaling cascades and transcriptional complexes that promote the formation and assembly of mitochondria—a process that is heavily dependent on timely and coordinated transcriptional control of encoding for mitochondrial pro- teins. In this article, we discuss the major signals and transcriptional complexes, program- ming mitochondrial biogenesis, and bioenergetic activity.This regulatory network represents a new therapeutic window into the treatment of the wide spectrum of mitochondrial and neurodegenerative diseases characterized by dysregulation of mitochondrial dynamics and bioenergetic deficiencies.

itochondria are dense, double membrane- organelle. Mitochondria, for instance, are essen- Menclosed organelles that are present in all tial for the synthesis of pyrimidines and purines, mammalian cells except mature red blood cor- contribute to the synthesis of heme, regulate ni- puscles. They uniquely possess their own ge- trogen balance through the urea cycle, produce nome in a circular DNA molecule. Although ketone bodies, are necessary for sex hormone this genome encodes for only a small fraction production, are involved in the processing of of the total genes needed for mitochondrial or- xenobiotics, play a critical role in redox balance, ganelle assembly and function, it is thus neces- and are key regulators of the apoptotic program sarily complemented by nuclear-encoded genes. in mammals. Given the centrality of mitochon- Mitochondria provide a wide variety of bio- dria to the maintenance of so many pathways in chemical services to the cell. Although it is con- mammalian cells, it is not surprising that there ventionally accepted that energy production exist multiple layers of control that enable a through the aerobic oxidation of carbon sub- cell to coordinate its net mitochondrial activity strates and the generation of ATPis the principal with fuel sources, biosynthetic demands, prolif- function of mitochondria, this narrow view eration rates, and external stimuli. One impor- overlooks the many other impressive biosyn- tant layer in this signaling control is the expres- thetic and regulatory capacities of this versatile sion of mitochondrial nuclear-derived genes

Editors: Douglas C. Wallace and Richard J. Youle Additional Perspectives on Mitochondria available at www.cshperspectives.org Copyright # 2013 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a015008 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008

1 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

J.E. Dominy and P. Puigserver

that initiate the cellular program of mitochon- mitochondrial genome through the direct mod- drial biogenesis. Defects and failure in the in- ulation of transcription factor A mitochondrial tegrity of this signaling system can cause serious (TFAM) and transcription factor B proteins perturbations to cellular metabolism and give (TFBs) expression; TFAM and TFBs are rise to a broad range of tissue specific as well major regulators of mitochondrial DNA tran- as systemic pathologies in humans. The same scription and replication (Gleyzer et al. 2005). signaling system, on the other hand, also opens NRF-1 may play an integral role in coupling many therapeutic opportunities for the correc- mitochondrial replication with nuclear replica- tion of mitochondrial defects. In this article, we tion over the course of the cell cycle through discuss some of the major signaling factors that regulation of E2F transcription factor targets serve to regulate mitochondrial biogenesis and and mitochondrial genes, indicating a link be- their implications for the mitigation of mito- tween cell division and mitochondrial biogen- chondrial disorders. esis (Cam et al. 2004). Studies involving the promoter region of cytochrome c oxidase complex subunit IV REGULATION OF MITOCHONDRIAL (COXIV) revealed the presence of another nu- BIOGENESIS AND FUNCTION THROUGH clear respiratory factor, NRF-2, which was able NUCLEAR-BASED GENE EXPRESSION to regulate the expression of proteins in the Although mitochondria contain in excess of electron transport chain (Carter et al. 1992). 1000 proteins, its tiny circular genome encodes NRF-2 promotes the expression of genes that for only 13 proteins, 22 tRNAs, and two rRNAs; encode for mitochondrial complex IV cyto- the balance of this genetic shortage is made chrome c oxidase (Gugneja et al. 1995). Like up by the nuclear genome (Calvo and Mootha NRF1, NRF2 also controls the expression of 2010). Because the vast majority of mitochon- TFAM and TFBs. The differential regulation drial genes are situated in the nucleus, trans- of NRF1 and NRF2 is not completely under- cription complexes at promoters of these genes stood but phosphorylation of these factors can control their expression. This affords the cell alter their transcriptional activities (Scarpulla the opportunity to coordinately regulate the 2008). In addition, specific coactivators of the expression of both mitochondrial and non- PGC-1 family sculpt NRFs-dependent control mitochondrialgenesetstoaffectaunifiedspecif- of mitochondrial gene expression in response ic cellular response. Below we highlight some to different signaling pathways (Wu et al. 1999). of the major nuclear transcriptional complexes regulating mitochondrial gene expression and Nuclear Hormone Receptors the diverse signaling pathways to which they respond (Fig. 1). Within the superfamily of nuclear hormone receptors, there are several subclasses that con- trol mitochondrial function and biogenesis. Nuclear Respiratory Factors The PPAR group of receptors is one such set. Sequence analysis of the electron transport gene PPARa, which can be activated by long-chain cytochrome c promoter initially identified nu- fatty acids and eicosanoids, can potently pro- clear respiratory factor 1 (NRF-1) as an impor- mote the expression of genes involved in mi- tant regulator of gene expression (Evans and tochondrial b-oxidation (Gulick et al. 1994). Scarpulla 1989). NRF-1 controls the expression The activation of PPARg activation, on the of a significant number of the proteins that other hand, triggers mitochondrial biogenesis make up the five respiratory complexes, as well in white adipose tissue (Wilson-Fritch et al. as proteins integral to mitochondrial import 2004); PPARg activation is also necessary to and heme biosynthesis (Scarpulla 2008). NRF- induce the expression of mitochondrial genes 1 is also able to integrate nuclear control of the involved in the brown fat-mediated thermogen- transcriptional and replicative activity of the ic program (Puigserver et al. 1998). PPARd,in

2 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Programming Mitochondrial Biogenesis

Nuclear-encoded genes

Fusion/ Mitochondrial TCA cycle enzymes Fatty acid β oxidation OXPHOS genes fission factors protein import Tfam/TFBs

PPARs ERRs CREB YY1 THRs NRF1 NRF2

RIP140 PGC-1α/β

JNK mTORC1 Nutrients/ anabolic stimuli PKA SIRT1 ROS

+ CAMK/MAPK AMPK NAD Adrenergic stimuli Muscle contractions/ 2+ Ca other stimuli ?

Mitochondrial gene transcription/ Low adenylate charge DNA replication

SIRT3/ SIRT4/ SIRT5

Deacylation of mitochondrial proteins

Figure 1. An overview of the transcriptional complexes regulating mitochdondrial gene expression and the signaling pathways converging upon them. Illustrated are the major transcriptional complexes/chromatin remodeling factors directly associated with changes in mitochondrial gene expression (see text for additional information).

skeletal muscle, is able to up-regulate mitochon- truncated form of thyroid hormone receptor a drial genes associated with fatty acid oxidation has been reported to be localized to the mito- and specify slow twitch fiber type (Narkar et al. chondrion and may serve to directly activate the 2008; Kleiner et al. 2009). transcription of genes in the mitochondrial ge- Thyroid hormone receptors (THRs) can nome (Casas et al. 1999). also promote mitochondrial biogenesis and tis- Estrogen-related receptors ERR-a, ERR-b, sue-specific function. This includes mitochon- and ERR-g are yet another set of nuclear hor- drial-driven thermogenesisthat occurs in brown mone receptors capable of promoting mito- fat during the adaptation to lower temperatures chondrial biogenesis. These receptors have no (Silva 1995). In some instances, THRs directly known endogenous ligands and are primary drive the transcription of nuclear-encoded genes, expressed in tissues with high oxidative meta- whereas, in others, the effects can occur indi- bolism capacities (Eichner and Giguere 2011). rectly through the thyroid hormone-mediated DNA-binding sites for ERR-a have been up-regulation of NRF-1 (Venditti et al. 2009). A mapped in a large number of nuclear-encoded

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 3 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

J.E. Dominy and P. Puigserver

mitochondrial genes, including those involved several transcription factors involved in the ba- in oxidative phosphorylation, fatty acid oxida- sic functions of the as well as its tion, TCA cycle, and factors regulating mito- rate of biogenesis (Puigserver and Spiegelman chondrial fusion/fission. Although ERRs are 2003; Kelly and Scarpulla 2004). Indeed, PGC- considered orphan nuclear receptors, their ac- 1a and PGC-1b are sufficient to increase to- tivity can certainly be modulated through re- tal mitochondrial mass, reactive oxygen species cruitment of coactivators/corepressor com- scavenging enzymes, oxidative phosphorylation plexes. Indeed, the transcriptional coactivators components, mitochondrial metabolic path- PGC-1a/b are efficacious activators of ERRs ways, protein import complexes, proteins in- and promote the expression of mitochondrial volved in fission and fusion, and the levels of genes (Mootha et al. 2004; Schreiber et al. 2004). mitochondrial sirtuins (Mootha et al. 2003; The transcriptional activity of this transcription Uldry et al. 2006; Cunningham et al. 2007; Ras- factor/activator module is controlled through bach et al. 2010; Handschin and Spiegelman deacetylation by sirtuin 1, which allows for an 2011). This process occurs in part through the important regulatory mechanism that connects ability of PGC-1 proteins to potentiate the func- nutrient/hormonal signaling to the control of tion of the NRFs, ERRs, and YY1. mitochondrial function (Wilson et al. 2010). In skeletal muscle, the increased mitochon- From the standpoint of repression of ERR ac- drial biogenesis caused by transgenic overex- tivity, the RIP140 nuclear hormone corepressor pression of PGC-1a can attenuate the devel- seems to be important (Wilson et al. 2010). opment of mitochondrial disease in mouse models of respiratory complex IV/cytochrome c oxidase deficiencies (Lin et al. 2002; Wenzet al. Other Transcription Factors: CREB and YY1 2008; Viscomi et al. 2011). It has also been CREB, a cAMP activated transcription factor, shown that PGC-1a/b expression improves mi- can promote the expression of several mito- tochondrial respiration capacity in cells from chondrial genes, including the proteins which human patients with a complex III or IV defi- make up complex IV and enzymes involved in ciency (Srivastava et al. 2009). It should also be the b-oxidation pathway (Gopalakrishnan and noted that age-related declines in mitochondri- Scarpulla 1994). Many mitochondrial genes al function are also mitigated by enhanced ac- contain YY1 binding sites within their promot- tivity of PGC-1a. Skeletal muscle specific PGC- er regions and this transcription factor has been 1a transgenic mice are less susceptible to exer- found to work in conjunction with PGC-1a to cise-induced fatigue and are protected against regulate their expression (Cunningham et al. age-related diseases, such as sarcopenia and 2007). Loss of YY1 expression in skeletal muscle metabolic diseases, and as a consequence, these causes intolerance and morphological mice show an increased life span (Wenz et al. signs of mitochondrial myopathy in mice (Blat- 2009). Nevertheless, it has yet to be determined tler et al. 2012b). whether the beneficial health effects of PGC-1a in skeletal muscle are attributable to its promo- tion of mitochondrial function or its effects on Transcriptional Coactivators nonmitochondrial gene expression. Transcriptional coactivators do not bind to DNA but can nonetheless promote gene tran- Sirtuin Proteins scription byenhancing the activityof true DNA- binding transcription factors. Biologically, these Sirtuins are NADþ-dependent protein deacy- proteins can set into motion genome-wide tran- lases that are homologous to yeast Sir2p, which scriptional changes by coactivating many differ- is one of the genes involved in transcriptional ent transcription factors. In mammalian cells, silencing in that organism (Klar et al. 1979). In the PGC-1 family of coactivators (PGC-1a, mammals there are seven sirtuin paralogs. PGC-1b, and PRC) potentiate the activity of SIRT1, SIRT6, and SIRT7 are nuclear proteins,

4 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Programming Mitochondrial Biogenesis

SIRT3, SIRT4, and SIRT5 are imported into of these variables frequently involves the use mitochondria, and SIRT2 is principally cyto- of signaling pathways that ultimately converge plasmic (Finkel et al. 2009; Haigis and Sinclair upon the transcriptional regulators described 2010; Verdin et al. 2010; Guarente 2011). above. In this section, we discuss the key signal- Through their deacylation activities, sirtuins ing modules that control mitochondrial genes modulate the biological activities of awide array and lead to increased mitochondrial mass. of target pathways. From the standpoint of mi- tochondrial biology, SIRT1, SIRT3, SIRT4, and AMP-Activated Protein Kinase (AMPK) SIRT5 have profound effects on the function of this organelle. SIRT1 deacetylates several key AMPK is a phylogenetically ancient cellular transcription factors that result in the up- sensor that is triggered by high cellular energy regulation of numerous genes involved in mito- demands (Hardie 2007). As such, it is not sur- chondrial respiration (Rodgers et al. 2005; Pu- prising that AMPK is strongly connected to mi- rushotham et al. 2009) and may (Price et al. tochondrial bioenergetics. When ATP synthesis 2012) or may not (Park et al. 2012) be necessary is impaired or ATP is being consumed and gen- for the promitochondrial effects of polyphe- erating AMP at a high rate, AMPK becomes ac- nolic stilbenoid compounds like resveratrol. In tive, which, in turn, phosphorylates several en- addition, sirtuins deacetylate numerous meta- zymes involved in the stimulation of catabolic bolic enzymes to govern their specific activity. pathways, such as glucose transport and fatty Changes in SIRT3 activity have been shown to acid oxidation and the inhibition of anabolic be an important determinant in the acetylation pathways, such as glycogen synthesis and lipo- state of mitochondrial in response to nutrient genesis (Kahn et al. 2005). Chronic activation availability (Hirschey et al. 2011; Hebert et al. of AMPK is also able to induce mitochondrial 2013). The acetylation of many mitochondrial biogenesis through the control of nuclear tran- proteins, such as isocitrate dehydrogenase 2 (Yu scription (Fig. 2). Essential to this process are et al. 2012), can alter their catalytic/biological the AMPK-mediated activation of PGC-1a function. As such, loss of SIRT3 activity results and SIRT1. AMPK can directly phosphorylate in profound aberrations in mitochondrial func- and activate PGC-1a (Jager et al. 2007); other tion and exacerbates the metabolic patholo- pathways that activate AMPK can result in a sub- gies associated with chronic nutrient excess. Al- sequent increase in PGC-1a protein expression though no consistent deacylase activities have and mitochondrial biogenesis (Birkenfeld et al. been shown for SIRT4, it has been shown that 2011). AMPK through Nampt-dependent and SIRT4 antagonizes mitochondrial capacity for -independent mechanisms is also able to in- fatty acid oxidation in hepatic and skeletal mus- crease NADþ levels, which promotes SIRT1 ac- cle cells (Nasrin et al. 2010). SIRT5, which has tivity and the deacetylation/activation of PGC- been shown to display desuccinylase, demalo- 1a(Cantoetal.2009). Thus,theAMPK/SIRT1/ nylase, and deacetylase activities, targets carba- PGC1asignalingaxis isthoughttoplayakey role moyl phosphate synthetase 1, and can regulate in the adaptive metabolic programming that oc- mitochondrial urea cycle flux (Nakagawa et al. cursduringcaloricrestrictionandexercise(Can- 2009; Du et al. 2011). to and Auwerx 2009). Nevertheless, it has been alternatively suggested that caloric restriction- mediated activation of AMPK serves to main- SIGNALING MODULES THAT CONTROL tain mitochondrial protein biosynthesis inde- MITOCHONDRIAL BIOGENESIS pendently of transcription (Miller et al. 2012). AND FUNCTION Cells are constantly engaged in both temporal Calcium Signaling and spatial control of mitochondrial biogenesis in response to nutrient availability, hormonal Intracellular calcium dynamics are affected by a cues, and changes in temperature. Integration wide array of signaling pathways and calcium, as

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 5 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

J.E. Dominy and P. Puigserver

Mitochondrial dynamic process Nuclear transcription control

• Mitochondrial unfolded protein response - Zinc finger ZC376.7 (C. elegans) - CHOP/C/EBPβ - PGC-1α/HSF1?

• Mitochondrial fusion/fission - PGC-1α/PGC-1β - ERRα; YY1

• Mitophagy - PARIS/PGC-1α

• Mitochondrial motility - Unknown

• Mitochondrial protein import - PGC-1α/PGC-1β - ERRα; NRF-1; YY1

• Mitochondrial transcription - PGC-1α/PGC-1β - ERRα; NRF-1; YY1

Figure 2. Transcriptional control of processes involved in the regulation of mitochondrial dynamics. Nuclear transcription factors regulate the transcription of specific proteins that are basic components of specific mito- chondrial processes associated with mitochondrial dynamics (see text for further information).

a second messenger molecule, and constitutes lient examples of the power of cAMP-mediated an important mechanism for initiating changes effects on mitochondrial function is found in mitochondrial gene expression. This phe- in the adaptive nonshivering thermogenic re- nomenon is most evident in skeletal muscle, a sponse to lower temperatures (Cannon and Ne- tissue that displays coordinated changes in cal- dergaard 2004). This heat-generating process cium homeostasis as part of its basic contrac- is highly dependent on induction of uncoupled tile mechanism. In this tissue, calcium-sensitive mitochondrial respiration in adipose depots— protein networks, such as calcium/calmodulin- primarily brown fat tissue—through the up- dependent kinase and p38 mitogen-activated regulation of uncoupling proteins such as kinase, are critical actors in the mobilization UCP1 and the up-regulation of oxidative path- of the transcriptional machinery that promotes ways (Cannon and Nedergaard 2004). Sym- mitochondrial proliferation and fatty acid oxi- pathetic stimulation of brown and “beige” fat dation (Wu et al. 2002; Wright et al. 2007). tissues increases cAMP-PKA tone in these tis- sues and precipitates the initial uncoupling re- sponse. Whereas the specific determination of cAMP Pathway the brown fat cell lineage is transcriptionally Yet another well-conserved second messenger specified through PRDM16 (Seale et al. 2007), system capable of altering mitochondrial func- the mitochondrial thermogenic response is de- tion is the cAMP-PKA pathway. The principal fined by the PGC-1 coactivators, which account transcriptional effector of this pathway is the for approximately 30% of the cAMP response in CREB transcription factor, whose phosphoryla- brown fat cells (Uldry et al. 2006). tion by PKA promotes CREB activity and target gene transcription (Mayr and Montminy 2001). mTOR Pathway PKA activation can also enhance the expression of PGC-1a, through the intermediacy of CREB The mechanistic target of rapamycin (mTOR) (Herzig et al. 2001), as well as directly activate pathway is a logic gate for integrating cell growth the catalytic function of SIRT1 (Gerhart-Hines and cell size with the availability of growth fac- et al. 2011). Biologically, one of the most sa- tors, energy levels, and certain nutrients, such

6 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Programming Mitochondrial Biogenesis

as the branched-chain amino acids. These pro- Calcium Signaling cesses require a net increase in cellular anabolic/ Much like the endoplasmic reticulum, mito- biosynthetic capacities, which will ultimately chondria are quantitatively significant stores of depend on enhanced mitochondrial biogene- calcium in the cell. As such, they can provide a sis and bioenergetics. The effects of mTOR on potential signaling avenue for the regulation of mitochondrial biology occur through several cellular processes—indeed, overloading the mi- mechanisms. One of the mechanisms seems to tochondrial calcium pool is a signal for the in- be independent of transcription through direct duction of apoptosis. At this point, although effects on mitochondrial organelles and stimu- mechanisms of calcium uptake by the mito- lation of respiration (Schieke et al. 2006). In a chondria have been characterized (Baughman second mechanism, mTOR modulates mito- et al. 2011; De Stefani et al. 2011), there is chondrial gene expression through the regula- no clearly delineated pathway by which mi- tion of different transcription factors. In liver, tochondrial calcium can be released to regulate mTOR affects genes of fatty acid oxidation and the calcium sensitive transcription/chroma- ketone body production through its ability to tin factor pathways discussed above. Nonethe- activate NCoR and inactivate PPARa (Sengupta less, PGC-1a increases mitochondrial calcium et al. 2010). In skeletal muscle, mTOR binds release, which establishes a positive feedback to the transcription factor YY1 and recruits loop for the maintenance of a mutual connec- PGC1a, increasing the ability of YY1 to activate tion between mitochondrial and nuclear gene mitochondrial gene expression. mTOR inhibi- expression (Bianchi et al. 2006). tion by using rapamycin, for example, results in a dissociation of PGC1a and decreases mito- chondrial gene expression (Cunningham et al. Reactive Oxygen Species (ROS) 2007; Blattler et al. 2012a). ROS are produced at respiratory chain com- plexes located in the inner mitochondrial mem- Retrograde Signaling from Mitochondria brane. In Drosophila, disruption of respiratory to the Nucleus complex 1 signals to components of the G1 to S cell cycle machinery trough JNK and FoxO The retrograde transmission of regulatory sig- (Owusu-Ansah et al. 2008). In mammalian nals from the mitochondria to the nucleus, cells, a similar retrograde signaling has been un- from a teleological standpoint, makes sense in covered through a systematic analysis of differ- that deficits or excesses in mitochondrial activ- ent mitochondrial dysfunction mutations; this ity would need to be ultimately corrected by pathway increases OXPHOS genes and involves adjustments in nuclear gene expression. This ROS signaling through JNK and the RXRa/ phenomenon has been actively explored in yeast PGC1a pathway (Chae et al. 2013). (Liu and Butow 2006). In mammalian cells, however, the nature of these signals and the mechanisms that they use are less well under- CONTROL OF MITOCHONDRIAL stood. Nevertheless, evidence of this regulatory PROCESSES THAT CONTRIBUTE TO THE phenomenon can be seen in the skeletal muscle BIOGENESIS OF MITOCHONDRIA of patients with mitochondrial diseases, such as Mitochondrial Unfolded Protein Response those with genetic mutations of oxidative respi- and Quality Control ration proteins, which display increased mito- chondrial proliferation—perhaps as an effort to The mitochondria contains an internal protease compensate for impaired mitochondrial activi- system that monitors accumulation of unfolded ty (DiMauro and Hirano 2009; Wallace and Fan or misfolded proteins (encoded by the nucle- 2009). The nature of this communication is un- ar or mitochondrial genomes) in the different known, but calcium and ROS signaling have compartments, including the outer membrane, been connected to this retrograde control. intermembrane space, inner membrane, and

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 7 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

J.E. Dominy and P. Puigserver

matrix. Thus, during mitochondrial biogenesis ring structures that will constrict and pro- or stress, chaperones and quality-control pro- duce fission. DRP1 has adaptor proteins that teases recognize hydrophobic amino acids that either activate fission (MFF) or suppress fission are in the interior structure of normal folded (MIEF) modulating the DRP1 GTPase activity. proteins (Baker and Haynes 2011). In addition FIS1 is the DRP1 receptor located in the out- to this organelle’s internal quality control, there er mitochondrial membrane and also interacts is a retrograde signaling to the nucleus via trans- and sequesters MIEF1. In cells, fusion makeup location of several transcription factors. In Cae- of interconnected mitochondria is essential to norhabditis elegans, mitochondrial chaperones maintain a highly metabolic and energetic state. such as mtHSP70 proteins bind to unfolded In contrast, quiescent or resting cells have frag- mitochondrial proteins; however, if the amount mented mitochondria with small spherical of unfolded proteins exceeds the chaperone ca- shapes with low metabolic activities (Chen and pacity, a protease called ClpXP degrades these Chan 2010). proteins into peptides. Mitochondrial efflux of Thus, it seems plausible that the mecha- these peptides activates a transcription factor, nisms that control mitochondrial biogenesis ZC376.7, which translocates to the nucleus to are tightly associated with the fusion machinery. activate expression of mitochondrial chaperone Along these lines, the transcriptional coactiva- genes (Haynes et al. 2010). In mammalian cells, tors PGC-1a and PGC1-b are positive acti- accumulation of unfolded proteins in the mi- vators of MFN2 (Cartoni et al. 2005; Soriano tochondrial matrix increases CHOP/C/EBPb et al. 2006). Whether there is independent tran- transcriptional activities on promoters encod- scriptional control of the fission machinery, or ing for mitochondrial chaperone genes, includ- if their regulation is simply bundled with the ing chaperonins 10 and 60, mtDNAJ, and ClpP rest of the nuclear-encoded mitochondrial pro- (Zhao et al. 2002). In hepatocyte cells, PGC-1a teins is currently unknown. interacts with HSF1; however, it is unknown whether this pathway controls the mitochon- Mitophagy drial unfolded protein response (Charos et al. 2012). Mitochondrial mass is defined by the balances of mitochondrial formation and degradation. This latter process largely occurs through a par- Mitochondrial Fusion/Fission ticular type of cellular autophagy, termed mi- Mitochondria are dynamic organelles that un- tophagy, that clears defective mitochondria. Mi- dergo constant division or fission and fusion tophagy is a cellular degradation process that (Twig et al. 2008; Westermann 2010; Corrado maintains “healthy” mitochondrial mass (Ash- et al. 2012). The mitochondrial mass undergoes rafi and Schwarz 2013). Elongated and fused mi- different shapes depending on the cellular state. tochondrial are resistant to mitophagy, whereas Fusion of the outer mitochondrial membrane is fragmented mitochondria generated by fission executed by the activity of two dynamin-related are more accessible to mitophagy. The elimina- GTPases, MFN1 and MFN2, that form homo- tion of damaged mitochondria is mediated by or heterodimers. In addition, OPA1—another the activities of PINK1 (PTEN-induced puta- dynamin-related GTPase—controls mitochon- tive protein kinase 1) and the E3 ubiquitin li- drial fusion and has also been involved in cyto- gase, Parkin. PINK1 and Parkin accumulate at chrome release and apoptosis. The mitochon- the mitochondrial surface of damaged organ- drial fission machinery is made up of DRP1, elles through the loss of mitochondrial mem- FIS1, MFF, and MIEF1. DRP1 is a GTPase pro- brane potential (Youle and van der Bliek 2012). tein that is localized in the cytoplasm, but In turn, this promotes separation from the after calcineurin-mediated dephosphorylation functional mitochondrial network followed by is recruited to the mitochondria. Once at the targeting defective mitochondria for degrada- mitochondria DRP1 oligomerizes and forms tion. This latter process requires Parkin-depen-

8 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Programming Mitochondrial Biogenesis

dent ubiquitination of mitochondrial proteins, tochondrial membrane proteins Miro1 and but the critical Parkin substrates necessary for Miro2, involving an adaptor protein termed mitophagy are unknown. MFN1 and MFN2 Milton. This is a calcium-dependent process: are ubiquitinated by Parkin, although they High concentrations of calcium halt mitochon- are not required for mitophagy. Mitochondria dria through the EF hands of Miro, anchor- are selectively removed upon treatment with ing mitochondria to sites of high ATP re- chemical uncouplers. In this process, opening quirements. It is unclear whether calcium is the of the mitochondrial permeability transition only regulator of this movement or there are pore (PTP) also triggers selective mitophagy. additional sites of control including establish- In addition, starvation signals such as glucagon ment of sensing gradients or transcriptional/ can also induce mitophagy but require opening translational regulation (Stowers et al. 2002; of the PTP. Similar to mitochondrial fusion/ Wang et al. 2011). fission, little is known about the transcriptional control of mitophagy. Interestingly, mutations Mitochondrial Protein Import that occur in Parkinson’s disease and inactivate the E3 ligase activity of Parkin, lead to the ac- Biogenesis of functional and competent mito- cumulation of Parkin substrates (Narendra et al. chondria requires the import and assembly of 2012). Among them is PARIS, a zinc finger co- proteins synthesized in the cytoplasm. The mi- repressor, which can suppress the transcription- tochondria contain more than 1000 proteins al coactivator PGC1a (Shin et al. 2011). Con- that are mostly imported from the cytoplasm sistent with this, PGC-1a responsive genes are through the translocase of the outer mitochon- underexpressed in microdissected dopaminer- drial membrane (TOM complex) (Endo et al. gic neurons of Parkinson’s disease mouse mod- 2011; Gebert et al. 2011). els, suggesting that this pathway could be caus- Once the mitochondrial protein has passed ative in this disease (Beal 2009; Zheng et al. the TOM channel, it is sorted by interactions 2010). Based on these data, it is possible to spec- with different machineries located in the in- ulate that during mitophagy Parkin activation termembrane space and inner membrane. Pro- can signal to the nucleus through Paris degra- teins that contain a presequence target signaling dation, resulting in an increase in PGC-1a and to the matrix are imported by the translocase will subsequently promote mitochondrial bio- of the inner membrane (TIM23) complex that genesis. is associated with the presequence translocase- associated motor (PAM). The TIM23 complex is also involved in laterally releasing proteins Mitochondrial Motility to the inner mitochondrial membrane. Several Depending on the cell type, an increase in mi- complexes in the intermembrane space par- tochondrial mass needs to be coordinated with ticipate in sorting mitochondrial proteins. For a precise intracellular localization of mitochon- example, the MIA (mitochondrial intermem- drial organelles to meet local energetic cellular brane assembly) complex promotes oxidative demands. Mitochondrial motility is influenced folding of intermembrane proteins. The Tim9– by the organelle’s size, which is in turn con- Tim10 chaperone complex transfers hydropho- trolled by fusion/fission processes. Failure in bic proteins to the mitochondrial outer mem- these processes prevents efficient intracellular brane through the SAM complex (e.g., b-barrel movement of mitochondria, as is most evident proteins) or the mitochondrial inner membrane in neurons, which criticallydepend on this path- through the TIM22 carrier complex (e.g., in- way to localize mitochondria close to synaptic ner membrane carrier proteins). Additional im- sites (Chen and Chan 2009). port pathways include the oxidase assembly Mitochondrial transport is based on micro- (OXA) machinery that transfers mitochondri- tubule dynamics. Kinesin-1 type motors are al proteins from the matrix ribosomes to the recruited to mitochondria through outer mi- inner membrane, or the mitochondrial import

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 9 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

J.E. Dominy and P. Puigserver

1 (Mim1) that imports a-helical outer mem- control mitochondrial transcription (Rebelo brane proteins (Endo and Yamano 2009; et al. 2011). How the transcription of mito- Schmidt et al. 2010). chondrial-encoded genes is precisely controlled The dynamics of this complex mitochondri- is not completely understood. Several of the al protein import process will depend not only proteins that control mitochondrial DNA tran- on the number of existing import complexes scription have posttranslational modifications, but also on the amounts of mitochondrial pro- including phosphorylation and acetylation. For tein translated in the cytoplasm. Little is known example, TFAM is specifically phosphorylated about the protein stability or activities of these (Lu et al. 2013) and acetylated (Dinardo et al. mitochondrial proteins, for example, control by 2003), suggesting that its activity could be mod- ubiquitination or other posttranslational mod- ified through cycles of acetylation/deacetyla- ifications (phosphorylation, acetylation, etc.) tion in response to energetic signals. Another that control the import processes. However, an level of control of this mitochondrial transcrip- important regulatory control is the coordinated tional machinery is by inducing amounts of its transcription of genes encoding for these pro- components. Along these lines elements essen- teinsthat will determine the rate of synthesis and tial to mitochondrial transcription, such as cellular concentrations that will promote and TFAM, TFB2M, MTERF,and POLRMT,are reg- contribute to mitochondrial mass. In mamma- ulated through the transcriptional coactivators lian cells, this transcriptional control involves PGC-1a and PGC-1b through their interaction the PGC-1 pathway and its interactions with with NRFs, ERRs, and YY1 (Mootha et al. 2004; transcription factors including NRFs and YY1 Blattler et al. 2012b; Scarpulla et al. 2012). that are bound to promoters of mitochondrial import genes (Blattler et al. 2012b; Scarpulla et al. 2012). Whether there is selectivity or spe- THERAPEUTIC POTENTIAL OF SIGNALING PATHWAYS FOR THE ATTENUATION OF cificity among the different import pathways MITOCHONDRIAL-RELATED described here is currently unknown. PATHOLOGIES Mitochondrial dysfunction, whether it is at- Mitochondrial Transcription tributable to deleterious mutations in nuclear Balanced and efficient mitochondrial biogen- or mtDNA genes (DiMauro and Hirano 2009; esis requires the activation of mitochondrial Wallace and Fan 2009) or to secondary systemic transcription (Peralta et al. 2012). The mam- changes caused by factors such as aging/senes- malian mitochondrial DNA is a circular dou- cence, significantly impairscellfunction andcan ble-stranded genome (16.5 kb) that encodes result in a spectrum of pathologies. In certain 37 genes. Transcription of the mitochondrial circumstances, such as aging, the mechanisms DNA (mtDNA) is tightly coordinated with nu- for why or how mitochondrial function and clear genes (particularly involved in OXPHOS) dynamics become abnormal are unknown (Pe- to achieve competent bieoenergetic mitochon- tersen et al. 2003; Reznicket al. 2007). Regardless drial organelles. The mtDNA contains two of the mechanism, however, data gleaned from strands: the heavy (H) and the light (L) strands. signaling studies suggest that certain elements The H strand contains most of the genes. In of these pathways can be leveraged to either pre- addition, there are two noncoding regions des- vent or attenuate a wide array of mitochondrial ignated as the D-loop that harbors the promoter dysfunctions. of both strands and the origin of replication. Mitochondrial transcription is bidirectional Genetically Based Mitochondrial Disorders and starts in the D-loop region through the assembly of a complex composed by TFAM, Despite the fact that significant advances have TFB2M, and the RNA polymerase POLRMT. been made in understanding the causative mo- MTERF proteins also bind to promoters and lecular mechanisms underlying many genetical-

10 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Programming Mitochondrial Biogenesis

ly based human mitochondrial disorders, there cleoside (AICAR) as one of the most efficacious is a frightening paucity of demonstrable treat- molecules (Golubitzky et al. 2011). In several ments (Hirano et al. 2012). In theory, however, mouse models of cytochrome c oxidase defi- interventions that boost the net mitochondrial ciency, AICAR treatment was also shown to activity in these patients to restore energy pro- lead to a partial rescue of COXIV activity and duction and antioxidant capacities to some some measure of improvement in motor perfor- minimum threshold should be sufficient to at- mance (Viscomi et al. 2011). tenuate afflicted organ dysfunction and the sub- The third most therapeutically attractive sequent pathologies. As such, there are several targets for mitochondrial diseases are the sir- druggable signaling avenues to pursue. tuin family of proteins. Within this family, The first is the PGC-1a signaling axis. As SIRT1 and SIRT3 offer the strongest connection mentioned earlier, transgenic overexpression of with improvements in mitochondrial health. PGC-1a has been shown to significantly blunt Small molecule activation of SIRT1 has been the pathological effects of cytochrome c oxidase shown in vivo to improve mitochondrial respi- (Viscomi et al. 2011), COXIV deficiency in mice ratory capacity in mice on a high-fat diet (Mi- (Lin et al. 2002; Wenz et al. 2008), and COXIII/ nor et al. 2011). In healthy human volunteers, IV deficiency in human patient-derived cell a next-generation SIRT1 activator (SRT2104) lines. Thus, small molecule therapies that po- has been shown to decrease serum lipid levels tentiate PGC-1a function or the function of and increase the recovery of adenosine diphos- one of its cognate transcription factors could phate and phosphocreatine after exercise, which yield tangible benefits. To date, efforts to specif- is suggestive of an increase in mitochondrial ically exploit this pathway have been largely lim- oxidative phosphorylation (Libri et al. 2012). ited to the testing of PPAR panagonists such Although there are no human in vivo studies as the fibrate family of compounds, which are of the effects of SIRT1 activation on mitochon- already in clinical use as antihyperlipidemic drial diseases, in vitro studies with the putative drugs. Studies with bezofibrate suggest that it SIRT1 activator resveratrol haveshownimprove- is of moderate use in humans suffering from ments in mitochondrial fatty acid oxidation mutations in carnitine palmitoyltransferase II in fibroblasts derived from patients with carni- deficiency (Bonnefont et al. 2009). Data on be- tine palmitoyltransferase II or very long-chain zofibrate’s ability to correct the phenotypes as- acyl-CoA dehydrogenase deficiencies (Bastin sociated with mutations in respiratory complex et al. 2011). In the case of SIRT3, up-regulation proteins, in contrast, have been confined largely of activity has been shown to improve mito- to mouse models and have yielded very mixed chondrial homeostasis in mice (Brown et al. results (Wenz et al. 2008; Viscomi et al. 2011; 2013), although no data are available in humans. Yatsuga and Suomalainen 2012). The major Ideally, therapies that simultaneously increase limitation of fibrates is that they target only a SIRT1 and SIRT3 activities, such as supplemen- single cognate transcription factor of PGC-1a, tation with the precursor nicotinamide riboside namely, PPARa, which is involved in up-regu- to increase the levels of their NADþ cosubstrate lation of the mitochondrial proteome. Small (Canto et al. 2012), would be the most desirable molecules that specifically up-regulate PGC- for promoting mitochondrial function. 1a expression or promote its inherent biologi- cal activity, such as through deacetylation, are no doubt needed for a full effect. Prevention of Mitochondrial Dysregulation Associated with Aging and Metabolic The second signaling axis isthat mediated by Disorders AMPK activation. High throughput screenings for compounds that improve the growth and Many of the therapeutic strategies described for ATP levels of fibroblasts from humans with a the amelioration of genetically based mitochon- complex I deficiency revealed the AMPK activa- drial disorders are applicable to the decline in tor 5-aminoimidazole-4-carboxamide ribonu- mitochondrial function that accompanies age-

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 11 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

J.E. Dominy and P. Puigserver

related senescence or certain metabolic disease trol mitochondrial function at the level of nu- states such as Type-2 diabetes. Other therapeu- clear transcription and discerning how these tic options are available to the nongenetic mi- pathways are targeted through extrinsic signals tochondrial disorders, because of their general- to ultimately govern mitochondrial activities. ly reduced phenotypic severity. These options Collectively, these signaling pathways can effec- include dietary manipulation and physical ex- tively boost the cell’s mitochondrial mass, im- ercise. Restriction of caloric intake can result in prove the mitochondrial bioenergetics profile, profound alterations in mitochondrial physiol- and protect it against oxidative stress. As such, ogy, including increased proliferation, enhanced they represent a very fertile ground for the de- bioenergetic efficiency, reduced membrane po- velopment of therapies for the rectification of tential, and reduced production of oxidative diseases characterized by mitochondrial dys- species in mice (Lopez-Lluch et al. 2006) and function. in humans (Civitarese et al. 2007). These mito- chondrial changes are thought to be a major contributor to the ability of caloric restriction ACKNOWLEDGMENTS to prevent the development of diseases linked Mitochondrial-related research in the Puig- to aging. Indeed, calorie restriction extends life server laboratory is supported from the Dana- span in different organisms but the molecu- Farber Cancer Institute funds and grants from lar mechanisms are not completely understood the NIH/NIDDK, American Diabetes Associa- (Sohal and Weindruch 1996; Spindler 2001). tion, American Heart Association, and Muscle Proposed mechanisms for the longevity effects Dystrophy Association. include changes in the signaling pathways asso- ciated with nutrient availability and insulin lev- els that converge to regulate mTOR (Kaeberlein REFERENCES et al. 2005; Zoncu et al. 2011) and FoxOs (Salih Ashrafi G, Schwarz TL. 2013. The pathways of mitophagy for and Brunet 2008) protein activities. In terms of quality control and clearance of mitochondria. Cell Death mitochondrial function, however, other regula- Differ 20: 31–42. tory pathways appear to be connected with the Baker BM, Haynes CM. 2011. Mitochondrial protein quality control during biogenesis and aging. Trends Biochem Sci effects of caloric restriction, namely, the AMPK, 36: 254–261. sirtuins, and PGC1a regulatory network. Bastin J, Lopes-Costa A, Djouadi F. 2011. Exposure to res- Exercise is an extremely powerful inducer of veratrol triggers pharmacological correction of fatty acid mitochondrial activity and proliferation—par- utilization in human fatty acid oxidation-deficient fibro- blasts. Hum Mol Genet 20: 2048–2057. ticularly in skeletal muscle (Yan et al. 2011). Baughman JM, Perocchi F,Girgis HS, Plovanich M, Belcher- Coincident with this change, exercise is also Timme CA, Sancak Y,Bao XR, Strittmatter L, Goldberger able to strongly protect against many types of O, Bogorad RL, et al. 2011. Integrative genomics identi- fies MCU as an essential component of the mitochondri- age-related metabolic disorders. Less clear, how- al calcium uniporter. Nature 476: 341–345. ever, is whether exercise can mimic the longevi- Beal MF. 2009. Therapeutic approaches to mitochondrial ty effects of caloric restriction. An important dysfunction in Parkinson’s disease. Parkinsonism Relat point, however, is whether the beneficial health Disord 15 (Suppl 3): S189–S194. effects of exercise are to an increase in the mass Bianchi K, Vandecasteele G, Carli C, Romagnoli A, Szabad- kai G, Rizzuto R. 2006. Regulation of Ca2þ signalling and of mitochondrial and/or the myriad other cel- Ca2þ-mediated cell death by the transcriptional coacti- lular and systemic changes that occur with ex- vator PGC-1a. Cell Death Differ 13: 586–596. ercise. Birkenfeld AL, Lee HY, Guebre-Egziabher F, Alves TC, Jurczak MJ, Jornayvaz FR, Zhang D, Hsiao JJ, Martin- Montalvo A, Fischer-Rosinsky A, et al. 2011. Deletion of the mammalian INDY homolog mimics aspects of die- CONCLUDING REMARKS tary restriction and protects against adiposity and insulin Researchers across multiple disciplines have resistance in mice. Cell Metab 14: 184–195. Blattler SM, Cunningham JT, Verdeguer F, Chim H, Haas made considerable progress by uncovering W, Liu H, Romanino K, Ruegg MA, Gygi S, Shi Y, many important signaling pathways that con- et al. 2012a. Yin Yang 1 deficiency in skeletal muscle

12 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Programming Mitochondrial Biogenesis

protects against rapamycin-induced diabetic-like symp- Chen H, Chan DC. 2009. Mitochondrial dynamics—fusion, toms through activation of insulin/IGF signaling. Cell fission, movement, and mitophagy—in neurodegenera- Metab 15: 505–517. tive diseases. Hum Mol Genet 18: R169–R176. Blattler SM, Verdeguer F, Liesa M, Cunningham JT, Vogel Chen H, Chan DC. 2010. Physiological functions of mito- RO, Chim H, Liu H, Romanino K, Shirihai OS, VazquezF, chondrial fusion. Ann NY Acad Sci 1201: 21–25. et al. 2012b. Defective mitochondrial morphology and Civitarese AE, Carling S, Heilbronn LK, Hulver MH, Uk- bioenergetic function in mice lacking the transcription ropcova B, Deutsch WA, Smith SR, Ravussin E. 2007. factor Yin Yang 1 in skeletal muscle. Mol Cell Biol 32: Calorie restriction increases muscle mitochondrial bio- 3333–3346. genesis in healthy humans. PLoS Med 4: e76. Bonnefont JP,Bastin J, Behin A, Djouadi F.2009. Bezafibrate Corrado M, Scorrano L, Campello S. 2012. Mitochondrial for an inborn mitochondrial b-oxidation defect. N Engl dynamics in cancer and neurodegenerative and neuro- JMed360: 838–840. inflammatory diseases. Int J Cell Biol 2012: 729290. Brown K, Xie S, Qiu X, Mohrin M, Shin J, Liu Y, Zhang D, Cunningham JT, Rodgers JT, Arlow DH, Vazquez F, Mootha Scadden DT, Chen D. 2013. SIRT3 reverses aging-associ- VK, Puigserver P. 2007. mTOR controls mitochondrial ated degeneration. Cell Rep 3: 319–327. oxidative function through a YY1-PGC-1a transcrip- Calvo SE, Mootha VK. 2010. The mitochondrial proteome tional complex. Nature 450: 736–740. and human disease. Annu Rev Genomics Hum Genet 11: De Stefani D, Raffaello A, TeardoE, Szabo I, Rizzuto R. 2011. 25–44. A forty-kilodalton protein of the inner membrane is the Cam H, Balciunaite E, Blais A, Spektor A, Scarpulla RC, mitochondrial calcium uniporter. Nature 476: 336–340. Young R, Kluger Y, Dynlacht BD. 2004. A common set of DiMauro S, Hirano M. 2009. Pathogenesis and treatment of gene regulatory networks links metabolism and growth mitochondrial disorders. Adv Exp Med Biol 652: 139– inhibition. Mol Cell 16: 399–411. 170. Cannon B, Nedergaard J. 2004. Brown adipose tissue: Func- Dinardo MM, Musicco C, Fracasso F, Milella F, Gadaleta tion and physiological significance. Physiol Rev 84: 277– MN, Gadaleta G, Cantatore P.2003. Acetylation and level 359. of mitochondrial transcription factor A in several organs Canto C, Auwerx J. 2009. PGC-1a, SIRT1 and AMPK, an of young and old rats. Biochem Biophys Res Commun 301: energy sensing network that controls energy expenditure. 187–191. Curr Opin Lipidol 20: 98–105. Du J, Zhou Y,Su X, Yu JJ, Khan S, Jiang H, Kim J, WooJ, Kim Canto C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L, JH, Choi BH, et al. 2011. Sirt5 is a NAD-dependent pro- Milne JC, Elliott PJ, Puigserver P,Auwerx J. 2009. AMPK tein lysine demalonylase and desuccinylase. Science 334: regulates energy expenditure by modulating NADþ me- 806–809. tabolism and SIRT1 activity. Nature 458: 1056–1060. Eichner LJ, Giguere V. 2011. Estrogen related receptors Canto C, Houtkooper RH, Pirinen E, Youn DY, Oosterveer (ERRs): A new dawn in transcriptional control of mito- MH, Cen Y, Fernandez-Marcos PH, Yamamoto H, An- chondrial gene networks. Mitochondrion 11: 544–552. dreux PA, Cettour-Rose P,et al. 2012. The NADþ precur- Endo T, Yamano K. 2009. Multiple pathways for mitochon- sor nicotinamide riboside enhances oxidative metabo- drial protein traffic. Biol Chem 390: 723–730. lism and protects against high-fat diet-induced obesity. Endo T, Yamano K, Kawano S. 2011. Structural insight into Cell Metab 15: 838–847. the mitochondrial protein import system. Biochim Bio- Carter RS, Bhat NK, Basu A, Avadhani NG. 1992. The basal phys Acta 1808: 955–970. promoter elements of murine cytochrome c oxidase sub- Evans MJ, Scarpulla RC. 1989. Interaction of nuclear factors unit IV gene consist of tandemly duplicated ets motifs with multiple sites in the somatic cytochrome c promot- that bind to GABP-related transcription factors. J Biol er. Characterization of upstream NRF-1, ATF, and intron Chem 267: 23418–23426. Sp1 recognition sequences. J Biol Chem 264: 14361– Cartoni R, Leger B, Hock MB, Praz M, Crettenand A, Pich S, 14368. Ziltener JL, Luthi F, Deriaz O, Zorzano A, et al. 2005. Finkel T,Deng CX, Mostoslavsky R. 2009. Recent progress in Mitofusins 1/2 and ERRa expression are increased in the biology and physiology of sirtuins. Nature 460: 587– human skeletal muscle after physical exercise. J Physiol 591. 567: 349–358. Gebert N, Ryan MT, Pfanner N, Wiedemann N, Stojanovski Casas F, Rochard P,Rodier A, Cassar-Malek I, Marchal-Vic- D. 2011. Mitochondrial protein import machineries and torion S, Wiesner RJ, Cabello G, Wrutniak C. 1999. A lipids: A functional connection. Biochim Biophys Acta variant form of the nuclear triiodothyronine receptor 1808: 1002–1011. c-ErbAa1 plays a direct role in regulation of mitochon- Gerhart-Hines Z, Dominy JE Jr, Blattler SM, Jedrychowski drial RNA synthesis. Mol Cell Biol 19: 7913–7924. MP, Banks AS, Lim JH, Chim H, Gygi SP, Puigserver P. Chae S, Ahn BY,Byun K, Cho YM, Yu MH, Lee B, Hwang D, 2011. The cAMP/PKA pathway rapidly activates SIRT1 Park KS. 2013. A systems approach for decoding mito- to promote fatty acid oxidation independently of changes chondrial retrograde signaling pathways. Science Signal in NADþ. Mol Cell 44: 851–863. 6: rs4. Gleyzer N, Vercauteren K, Scarpulla RC. 2005. Control of Charos AE, Reed BD, Raha D, Szekely AM, Weissman mitochondrial transcription specificity factors (TFB1M SM, Snyder M. 2012. A highly integrated and complex and TFB2M) by nuclear respiratory factors (NRF-1 and PPARGC1A transcription factor binding network in NRF-2) and PGC-1 family coactivators. Mol Cell Biol 25: HepG2 cells. Genome Res 22: 1668–1679. 1354–1366.

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 13 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

J.E. Dominy and P. Puigserver

Golubitzky A, Dan P, Weissman S, Link G, Wikstrom JD, clues to modern understanding of metabolism. Cell Saada A. 2011. Screening for active small molecules in Metab 1: 15–25. mitochondrial complex I deficient patient’s fibroblasts, Kelly DP, Scarpulla RC. 2004. Transcriptional regulatory reveals AICAR as the most beneficial compound. PloS circuits controlling mitochondrial biogenesis and func- ONE 6: e26883. tion. Genes Dev 18: 357–368. Gopalakrishnan L, Scarpulla RC. 1994. Differential regula- Klar AJ, Fogel S, Macleod K. 1979. MAR1—A regulator of tion of respiratory chain subunits by a CREB-dependent the HMa and HMa loci in Saccharomyces cerevisiae. Ge- signal transduction pathway. Role of cyclic AMP in cyto- netics 93: 37–50. chrome c and COXIV gene expression. J Biol Chem 269: Kleiner S, Nguyen-Tran V, Bare O, Huang X, Spiegelman B, 105–113. Wu Z. 2009. PPARd agonism activates fatty acid oxida- Guarente L. 2011. Franklin H. Epstein Lecture: Sirtuins, tion via PGC-1a but does not increase mitochondrial aging, and medicine. N Engl J Med 364: 2235–2244. gene expression and function. J Biol Chem 284: 18624– Gugneja S, Virbasius JV, Scarpulla RC. 1995. Four structur- 18633. ally distinct, non-DNA-binding subunits of human nu- Libri V, Brown AP, Gambarota G, Haddad J, Shields GS, clear respiratory factor 2 share a conserved transcription- Dawes H, Pinato DJ, Hoffman E, Elliot PJ, Vlasuk GP, al activation domain. Mol Cell Biol 15: 102–111. et al. 2012. A pilot randomized, placebo controlled, dou- Gulick T, Cresci S, Caira T, Moore DD, Kelly DP. 1994. The ble blind phase I trial of the novel SIRT1 activator peroxisome proliferator-activated receptor regulates mi- SRT2104 in elderly volunteers. PloS ONE 7: e51395. tochondrial fatty acid oxidative enzyme gene expression. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, Boss O, Michael LF, Proc Natl Acad Sci 91: 11012–11016. Puigserver P, Isotani E, Olson EN, et al. 2002. Transcrip- Haigis MC, Sinclair DA. 2010. Mammalian sirtuins: Biolog- tional co-activator PGC-1 a drives the formation of slow- ical insights and disease relevance. Annu Rev Pathol 5: twitch muscle fibres. Nature 418: 797–801. 253–295. Liu Z, Butow RA. 2006. Mitochondrial retrograde signaling. Handschin C, Spiegelman BM. 2011. PGC-1 coactivators Annu Rev Genet 40: 159–185. and the regulation of skeletal muscle fiber-type determi- Lopez-Lluch G, Hunt N, Jones B, Zhu M, Jamieson H, nation. Cell Metab 13: 351; author reply 352. Hilmer S, Cascajo MV, Allard J, Ingram DK, Navas P, et al. 2006. Calorie restriction induces mitochondrial Hardie DG. 2007. AMP-activated/SNF1 protein kinases: biogenesis and bioenergetic efficiency. Proc Natl Acad Conserved guardians of cellular energy. Nat Rev Mol Sci 103: 1768–1773. Cell Biol 8: 774–785. Lu B, Lee J, Nie X, Li M, Morozov YI, Venkatesh S, Bogen- Haynes CM, Yang Y,Blais SP,Neubert TA, Ron D. 2010. The hagen DF, Temiakov D, Suzuki CK. 2013. Phosphoryla- matrix peptide exporter HAF-1 signals a mitochondrial tion of human TFAM in mitochondria impairs DNA UPR by activating the transcription factor ZC376.7 in binding and promotes degradation by the AAAþ Lon C. elegans. Mol Cell 37: 529–540. protease. Mol Cell 49: 121–132. Hebert AS, Dittenhafer-Reed KE, Yu W, Bailey DJ, Selen ES, Mayr B, Montminy M. 2001. Transcriptional regulation by Boersma MD, Carson JJ, Tonelli M, Balloon AJ, Higbee the phosphorylation-dependent factor CREB. Nat Rev AJ, et al. 2013. Calorie restriction and SIRT3 trigger glob- Mol Cell Biol 2: 599–609. al reprogramming of the mitochondrial protein acety- lome. Mol Cell 49: 186–199. Miller BF, Robinson MM, Bruss MD, Hellerstein M, Ham- ilton KL. 2012. A comprehensive assessment of mito- Herzig S, Long F, Jhala US, Hedrick S, Quinn F, Bauer A, chondrial protein synthesis and cellular proliferation Rudolph D, Schutz G, Yoon C, Puigserver P, et al. 2001. with age and caloric restriction. Aging Cell 11: 150–161. CREB regulates hepatic gluconeogenesis through the co- Minor RK, Baur JA, Gomes AP, Ward TM, Csiszar A, activator PGC-1. Nature 413: 179–183. Mercken EM, Abdelmohsen K, Shin YK, Canto C, Schei- Hirano M, Garone C, Quinzii CM. 2012. CoQ10 deficiencies bye-Knudsen M, et al. 2011. SRT1720 improves survival and MNGIE: Two treatable mitochondrial disorders. Bi- and healthspan of obese mice. Scientific Rep 1: 70. ochim Biophys Acta 1820: 625–631. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Hirschey MD, Shimazu T, Jing E, Grueter CA, Collins AM, Sihag S, Lehar J, Puigserver P,Carlsson E, Ridderstrale M, Aouizerat B, Stancakova A, Goetzman E, Lam MM, Laurila E, et al. 2003. PGC-1a-responsive genes involved Schwer B, et al. 2011. SIRT3 deficiency and mitochon- in oxidative phosphorylation are coordinately downregu- drial protein hyperacetylation accelerate the development lated in human diabetes. Nat Genet 34: 267–273. of the metabolic syndrome. Mol Cell 44: 177–190. Mootha VK, Handschin C, Arlow D, Xie X, St Pierre J, Sihag Jager S, Handschin C, St-Pierre J, Spiegelman BM. 2007. S, Yang W, Altshuler D, Puigserver P, Patterson N, et al. AMP-activated protein kinase (AMPK) action in skeletal 2004. Erra and Gabpa/b specify PGC-1a-dependent ox- muscle via direct phosphorylation of PGC-1a. Proc Natl idative phosphorylation gene expression that is altered in Acad Sci 104: 12017–12022. diabetic muscle. Proc Natl Acad Sci 101: 6570–6575. Kaeberlein M, Powers RW III, Steffen KK, Westman EA, Hu Nakagawa T,Lomb DJ, Haigis MC, Guarente L. 2009. SIRT5 D, Dang N, Kerr EO, Kirkland KT, Fields S, Kennedy BK. Deacetylates carbamoyl phosphate synthetase 1 and reg- 2005. Regulation of yeast replicative life span by TOR and ulates the urea cycle. Cell 137: 560–570. Sch9 in response to nutrients. Science 310: 1193–1196. Narendra D, Walker JE, Youle R. 2012. Mitochondrial qual- Kahn BB, Alquier T, Carling D, Hardie DG. 2005. AMP- ity control mediated by PINK1 and Parkin: Links to par- activated protein kinase: Ancient energy gauge provides kinsonism. Cold Spring Harb Perspect Biol 4: a011338.

14 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Programming Mitochondrial Biogenesis

Narkar VA,Downes M, Yu RT, Embler E, Wang YX, Banayo Scarpulla RC, Vega RB, Kelly DP. 2012. Transcriptional in- E, Mihaylova MM, Nelson MC, Zou Y, Juguilon H, et al. tegration of mitochondrial biogenesis. Trends Endocrinol 2008. AMPK and PPARd agonists are exercise mimetics. Metab: 23: 459–466. Cell 134: 405–415. Schieke SM, Phillips D, McCoy JP Jr, Aponte AM, Shen RF, Nasrin N, Wu X, Fortier E, Feng Y,Bare OC, Chen S, Ren X, Balaban RS, Finkel T. 2006. The mammalian target of Wu Z, Streeper RS, Bordone L. 2010. SIRT4 regulates rapamycin (mTOR) pathway regulates mitochondrial fatty acid oxidation and mitochondrial gene expression oxygen consumption and oxidative capacity. J Biol in liver and muscle cells. J Biol Chem 285: 31995–32002. Chem 281: 27643–27652. Owusu-Ansah E, Yavari A, Mandal S, Banerjee U. 2008. Schmidt O, Pfanner N, Meisinger C. 2010. Mitochondrial Distinct mitochondrial retrograde signals control the protein import: From proteomics to functional mecha- G1-S cell cycle checkpoint. Nat Genet 40: 356–361. nisms. Nat Rev Mol Cell Biol 11: 655–667. Park SJ, Ahmad F,Philp A, Baar K, Williams T,Luo H, Ke H, Schreiber SN, Emter R, Hock MB, Knutti D, Cardenas J, Rehmann H, TaussigR, Brown AL, et al. 2012. Resveratrol Podvinec M, Oakeley EJ, Kralli A. 2004. The estrogen- ameliorates aging-related metabolic phenotypes by in- related receptor a (ERRa) functions in PPARg coactiva- hibiting cAMP phosphodiesterases. Cell 148: 421–433. tor 1a (PGC-1a)-induced mitochondrial biogenesis. Peralta S, Wang X, Morales CT. 2012. Mitochondrial tran- Proc Natl Acad Sci 101: 6472–6477. scription: Lessons from mouse models. Biochim Biophys Seale P, Kajimura S, Yang W, Chin S, Rohas LM, Uldry M, Acta 1819: 961–969. Tavernier G, Langin D, Spiegelman BM. 2007. Transcrip- Petersen KF, Befroy D, Dufour S, Dziura J, Ariyan C, Roth- tional control of brown fat determination by PRDM16. man DL, DiPietro L, Cline GW, Shulman GI. 2003. Mi- Cell Metab 6: 38–54. tochondrial dysfunction in the elderly: Possible role in Sengupta S, Peterson TR, Laplante M, Oh S, Sabatini DM. insulin resistance. Science 300: 1140–1142. 2010. mTORC1 controls fasting-induced ketogenesis Price NL, Gomes AP,Ling AJ, Duarte FV,Martin-Montalvo and its modulation by ageing. Nature 468: 1100–1104. A, North BJ, Agarwal B, Ye L, Ramadori G, Teodoro JS, Shin JH, Ko HS, Kang H, Lee Y, Lee YI, Pletinkova O, Tro- et al. 2012. SIRT1 is required for AMPK activation and conso JC, Dawson VL, Dawson TM. 2011. PARIS the beneficial effects of resveratrol on mitochondrial (ZNF746) repression of PGC-1a contributes to neuro- function. Cell Metab 15: 675–690. degeneration in Parkinson’s disease. Cell 144: 689–702. Puigserver P, Spiegelman BM. 2003. Peroxisome prolifera- Silva JE. 1995. Thyroid hormone control of thermogenesis tor-activated receptor-g coactivator 1a (PGC-1a): Tran- and energy balance. Thyroid 5: 481–492. scriptional coactivator and metabolic regulator. Endocr Rev 24: 78–90. Sohal RS, Weindruch R. 1996. Oxidative stress, caloric re- striction, and aging. Science 273: 59–63. Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spie- gelman BM. 1998. A cold-inducible coactivator of nucle- Soriano FX, Liesa M, Bach D, Chan DC, Palacin M, Zorzano ar receptors linked to adaptive thermogenesis. Cell 92: A. 2006. Evidence for a mitochondrial regulatory path- 829–839. way defined by peroxisome proliferator-activated recep- a a Purushotham A, Schug TT, Xu Q, Surapureddi S, Guo X, Li tor-gamma coactivator-1 , estrogen-related receptor- , X. 2009. Hepatocyte-specific deletion of SIRT1 alters fat- and mitofusin 2. Diabetes 55: 1783–1791. ty acid metabolism and results in hepatic steatosis and Spindler SR. 2001. Calorie restriction enhances the expres- inflammation. Cell Metab 9: 327–338. sion of key metabolic enzymes associated with protein Rasbach KA, Gupta RK, Ruas JL, Wu J, Naseri E, Estall JL, renewal during aging. Ann NY Acad Sci 928: 296–304. Spiegelman BM. 2010. PGC-1a regulates a HIF2a-de- Srivastava S, Diaz F, Iommarini L, Aure K, Lombes A, Mor- pendent switch in skeletal muscle fiber types. Proc Natl aes CT. 2009. PGC-1a/b induced expression partially Acad Sci 107: 21866–21871. compensates for respiratory chain defects in cells from Rebelo AP, Dillon LM, Moraes CT. 2011. Mitochondrial patients with mitochondrial disorders. Hum Mol Genet DNA transcription regulation and nucleoid organiza- 18: 1805–1812. tion. J Inherit Metabol Dis 34: 941–951. Stowers RS, Megeath LJ, Gorska-Andrzejak J, Meinertzha- Reznick RM, Zong H, Li J, Morino K, Moore IK, Yu HJ, Liu gen IA, Schwarz TL. 2002. Axonal transport of mitochon- ZX, Dong J, Mustard KJ, Hawley SA, et al. 2007. Aging- dria to synapses depends on milton, a novel Drosophila associated reductions in AMP-activated protein kinase protein. Neuron 36: 1063–1077. activity and mitochondrial biogenesis. Cell Metab 5: Twig G, Hyde B, Shirihai OS. 2008. Mitochondrial fusion, 151–156. fission and autophagy as a quality control axis: The bio- Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, energetic view. Biochim Biophys Acta 1777: 1092–1097. Puigserver P. 2005. Nutrient control of glucose homeo- Uldry M, Yang W,St-Pierre J, Lin J, Seale P,Spiegelman BM. stasis through a complex of PGC-1a and SIRT1. Nature 2006. Complementary action of the PGC-1 coactivators 434: 113–118. in mitochondrial biogenesis and brown fat differentia- Salih DA, Brunet A. 2008. FoxO transcription factors in the tion. Cell Metab 3: 333–341. maintenance of cellular homeostasis during aging. Curr Venditti P,Bari A, Di Stefano L, Cardone A, Della Ragione F, Opin Cell Biol 20: 126–136. D’Esposito M, Di Meo S. 2009. Involvement of PGC-1, Scarpulla RC. 2008. Transcriptional paradigms in mamma- NRF-1, and NRF-2 in metabolic response by rat liver to lian mitochondrial biogenesis and function. Physiol Rev hormonal and environmental signals. Mol Cell Endocri- 88: 611–638. nol 305: 22–29.

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 15 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

J.E. Dominy and P. Puigserver

Verdin E, Hirschey MD, Finley LW,Haigis MC. 2010. Sirtuin drial biogenesis by a pathway leading to p38 mitogen- regulation of mitochondria: Energy production, apopto- activated protein kinase activation. J Biol Chem 282: sis, and signaling. Trends Biochem Sci 35: 669–675. 18793–18799. Viscomi C, Bottani E, Civiletto G, Cerutti R, Moggio M, Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Fagiolari G, Schon EA, Lamperti C, Zeviani M. 2011. In Mootha V, Troy A, Cinti S, Lowell B, Scarpulla RC, et al. vivo correction of COX deficiency by activation of the 1999. Mechanisms controlling mitochondrial biogene- AMPK/PGC-1a axis. Cell Metab 14: 80–90. sis and respiration through the thermogenic coactivator Wallace DC, Fan W. 2009. The pathophysiology of mito- PGC-1. Cell 98: 115–124. chondrial disease as modeled in the mouse. Genes Dev Wu H, Kanatous SB, Thurmond FA, Gallardo T, Isotani E, 23: 1714–1736. Bassel-Duby R, Williams RS. 2002. Regulation of mito- Wang X, Winter D, Ashrafi G, Schlehe J, WongYL, Selkoe D, chondrial biogenesis in skeletal muscle by CaMK. Science Rice S, Steen J, LaVoie MJ, Schwarz TL. 2011. PINK1 and 296: 349–352. Parkin target Miro for phosphorylation and degradation Yan Z, Okutsu M, Akhtar YN, Lira VA.2011. Regulation of to arrest mitochondrial motility. Cell 147: 893–906. exercise-induced fiber type transformation, mitochon- Wenz T, Diaz F, Spiegelman BM, Moraes CT. 2008. Activa- drial biogenesis, and angiogenesis in skeletal muscle. J tion of the PPAR/PGC-1a pathway prevents a bioener- Appl Physiol 110: 264–274. getic deficit and effectively improves a mitochondrial Yatsuga S, Suomalainen A. 2012. Effect of bezafibrate treat- myopathy phenotype. Cell Metab 8: 249–256. ment on late-onset mitochondrial myopathy in mice. WenzT,Rossi SG, Rotundo RL, Spiegelman BM, Moraes CT. Hum Mol Genet 21: 526–535. 2009. Increased muscle PGC-1a expression protects from Youle RJ, van der Bliek AM. 2012. Mitochondrial fission, sarcopenia and metabolic disease during aging. Proc Natl fusion, and stress. Science 337: 1062–1065. Acad Sci 106: 20405–20410. Yu W, Dittenhafer-Reed KE, Denu JM. 2012. SIRT3 protein Westermann B. 2010. Mitochondrial fusion and fission in deacetylates isocitrate dehydrogenase 2 (IDH2) and reg- cell life and death. Nat Rev Mol Cell Biol 11: 872–884. ulates mitochondrial redox status. J Biol Chem 287: Wilson BJ, Tremblay AM, Deblois G, Sylvain-Drolet G, Gi- 14078–14086. guere V. 2010. An acetylation switch modulates the tran- Zhao Q, Wang J, Levichkin IV, Stasinopoulos S, Ryan MT, scriptional activity of estrogen-related receptor a. Mol Hoogenraad NJ. 2002. A mitochondrial specific stress Endocrinol 24: 1349–1358. response in mammalian cells. EMBO J 21: 4411–4419. Wilson-Fritch L, Nicoloro S, Chouinard M, Lazar MA, Chui Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt PC, Leszyk J, Straubhaar J, Czech MP, Corvera S. 2004. ML, Eklund AC, Zhang-James Y, Kim PD, Hauser MA, Mitochondrial remodeling in adipose tissue associated et al. 2010. PGC-1a, a potential therapeutic target for with obesity and treatment with rosiglitazone. J Clin In- early intervention in Parkinson’s disease. Sci Transl Med vest 114: 1281–1289. 2: 52ra73. Wright DC, Geiger PC, Han DH, Jones TE, Holloszy JO. Zoncu R, Efeyan A, Sabatini DM. 2011. mTOR: From 2007. Calcium induces increases in peroxisome prolifer- growth signal integration to cancer, diabetes and ageing. ator-activated receptor g coactivator-1a and mitochon- Nat Rev Mol Cell Biol 12: 21–35.

16 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a015008 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Mitochondrial Biogenesis through Activation of Nuclear Signaling Proteins

John E. Dominy and Pere Puigserver

Cold Spring Harb Perspect Biol 2013; doi: 10.1101/cshperspect.a015008

Subject Collection Mitochondria

Altered Sulfide (H2S) Metabolism in Ethylmalonic Where Killers Meet−−Permeabilization of the Outer Encephalopathy Mitochondrial Membrane during Apoptosis Valeria Tiranti and Massimo Zeviani Tom Bender and Jean-Claude Martinou Mitochondrial DNA Genetics and the Mitochondrial Biogenesis through Activation of Heteroplasmy Conundrum in Evolution and Nuclear Signaling Proteins Disease John E. Dominy and Pere Puigserver Douglas C. Wallace and Dimitra Chalkia The Role of Mitochondria in Cellular Iron−Sulfur Mitochondrial Trafficking in Neurons Protein Biogenesis: Mechanisms, Connected Thomas L. Schwarz Processes, and Diseases Oliver Stehling and Roland Lill Mechanisms of Mitochondrial Fission and Fusion Mitochondrial Dysfunction and Defective Alexander M. van der Bliek, Qinfang Shen and Autophagy in the Pathogenesis of Collagen VI Sumihiro Kawajiri Muscular Dystrophies Paolo Bernardi and Paolo Bonaldo The Mitochondrial Nucleoid: Integrating Clinical and Molecular Features of POLG-Related Mitochondrial DNA into Cellular Homeostasis Mitochondrial Disease Robert Gilkerson, Liliana Bravo, Iraselia Garcia, et Jeffrey D. Stumpf, Russell P. Saneto and William C. al. Copeland Relevance of Mitochondrial Genetics and Mitochondrial Metabolism, Sirtuins, and Aging Metabolism in Cancer Development Michael N. Sack and Toren Finkel Giuseppe Gasparre, Anna Maria Porcelli, Giorgio Lenaz, et al. Mitochondrial Quality Control Mediated by PINK1 Mechanisms of Protein Sorting in Mitochondria and Parkin: Links to Parkinsonism Diana Stojanovski, Maria Bohnert, Nikolaus Derek Narendra, John E. Walker and Richard Youle Pfanner, et al.

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2013 Cold Spring Harbor Laboratory Press; all rights reserved Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Mitochondrial Evolution Michael W. Gray

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2013 Cold Spring Harbor Laboratory Press; all rights reserved