the mitochondrial metallochaperone sco1 is …cell reports article the mitochondrial...

12
Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter CTR1 and Preserve Copper Homeostasis Graphical Abstract Highlights d Liver-specific deletion of the mitochondrial metallochaperone Sco1 is lethal d Sco1-null livers are COX and Cu deficient, like affected tissues of SCO1 patients d Loss of SCO1 causes a Cu deficiency by reducing CTR1 abundance d Sco1 deletion results in enhanced proteasomal degradation of CTR1 Authors Christopher J. Hlynialuk, Binbing Ling, ..., Michael J. Petris, Scot C. Leary Correspondence [email protected] In Brief Hlynialuk et al. demonstrate that SCO1 is integral to a mitochondrial signaling pathway that regulates copper homeostasis by controlling the abundance and plasma membrane localization of CTR1, the high-affinity permease responsible for importing copper into the cell. Accession Numbers GSE58997 Hlynialuk et al., 2015, Cell Reports 10, 933–943 February 17, 2015 ª2015 The Authors http://dx.doi.org/10.1016/j.celrep.2015.01.019

Upload: others

Post on 26-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Article

The Mitochondrial Metallo

chaperone SCO1 IsRequired to Sustain Expression of the High-AffinityCopper Transporter CTR1 and Preserve CopperHomeostasis

Graphical Abstract

Highlights

d Liver-specific deletion of the mitochondrial

metallochaperone Sco1 is lethal

d Sco1-null livers are COX and Cu deficient, like affected

tissues of SCO1 patients

d Loss of SCO1 causes a Cu deficiency by reducing CTR1

abundance

d Sco1 deletion results in enhanced proteasomal degradation

of CTR1

Hlynialuk et al., 2015, Cell Reports 10, 933–943February 17, 2015 ª2015 The Authorshttp://dx.doi.org/10.1016/j.celrep.2015.01.019

Authors

Christopher J. Hlynialuk, Binbing Ling, ...,

Michael J. Petris, Scot C. Leary

[email protected]

In Brief

Hlynialuk et al. demonstrate that SCO1 is

integral to a mitochondrial signaling

pathway that regulates copper

homeostasis by controlling the

abundance and plasma membrane

localization of CTR1, the high-affinity

permease responsible for importing

copper into the cell.

Accession Numbers

GSE58997

Page 2: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Cell Reports

Article

The Mitochondrial Metallochaperone SCO1 Is Requiredto Sustain Expression of the High-Affinity CopperTransporter CTR1 and Preserve Copper HomeostasisChristopher J. Hlynialuk,1,7 Binbing Ling,1,7 Zakery N. Baker,1,7 Paul A. Cobine,2 Lisa D. Yu,1 Aren Boulet,1 Timothy Wai,3

Amzad Hossain,1 Amr M. El Zawily,1,6 Pamela J. McFie,1 Scot J. Stone,1 Francisca Diaz,4 Carlos T. Moraes,4

Deepa Viswanathan,5 Michael J. Petris,5 and Scot C. Leary1,*1Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada2Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA3Institute for Genetics, University of Cologne, 50931 Cologne, Germany4Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA5Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA6Faculty of Science, Damanhour University, Damanhour 22516, Egypt7Co-first author

*Correspondence: [email protected]

http://dx.doi.org/10.1016/j.celrep.2015.01.019

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/3.0/).

SUMMARY

Human SCO1 fulfills essential roles in cytochrome coxidase (COX) assembly and the regulation of copper(Cu) homeostasis, yet it remains unclear why patho-genic mutations in this gene cause such clinicallyheterogeneous forms of disease. Here, we establisha Sco1 mouse model of human disease and showthat ablation of Sco1 expression in the liver is lethalowing to severe COX and Cu deficiencies. We furtherdemonstrate that the Cu deficiency is explained by afunctional connection between SCO1 and CTR1, thehigh-affinity transporter that imports Cu into the cell.CTR1 is rapidly degraded in the absence of SCO1protein, and we show that its levels are restored inSco1�/� mouse embryonic fibroblasts upon inhibi-tion of the proteasome. These data suggest thatmitochondrial signaling through SCO1 provides apost-translational mechanism to regulate CTR1-dependent Cu import into the cell, and they furtherunderpin the importance of mitochondria in cellularCu homeostasis.

INTRODUCTION

Copper (Cu) is an essential trace element that is acquired via the

diet and is required as a co-factor by several mammalian en-

zymes that collectively catalyze biochemical reactions critical

to early development and general physiology (Desai and Kaler,

2008). Because Cu is toxic in its unbound form, conserved

machinery has evolved to tightly regulate cellular Cu uptake

and efflux. Dedicated chaperones further ensure that Cu is safely

trafficked throughout the cell to specific cupro-proteins for their

metallation, or to labile pools for its storage (Kim et al., 2008;

Ce

Festa and Thiele, 2011). Genetic variants or environmental in-

sults that impair these Cu-handling systems negatively affect

innate immunity, impact the progression and treatment of

several types of cancer, and cause severe forms of human dis-

ease (Desai and Kaler, 2008; Turski and Thiele, 2009; Festa

and Thiele, 2012). Thus, there is considerable interest in

advancing our mechanistic understanding of how Cu is handled

at the cellular through to the systemic levels of organization.

Cellular Cu uptake is mediated by the permease copper trans-

porter 1 (CTR1) (Lee et al., 2002; Puig et al., 2002; Eisses and

Kaplan, 2005), the only high-affinity Cu importer that has been

identified to date. CTR1 cycles between the plasma membrane

(PM) and cytosolic vesicles in many cell types, and regulated

changes in its abundance or localization allow for the rate

of Cu uptake to be rapidly adjusted in response to altered cellular

Cu levels (Petris et al., 2003; Guo et al., 2004). Following import,

Cu is transferred from CTR1 to soluble chaperones for its deliv-

ery to various subcellular compartments. Copper chaperone for

superoxide dismutase 1 (CCS1) delivers and inserts Cu into

SOD1 within the cytosol (Lamb et al., 2001). Cu destined for

the trans-Golgi network and the secretory pathway is delivered

via ATOX1 to ATP7A or ATP7B (Hamza et al., 1999; Wernimont

et al., 2000), homologous P-type ATPases with analogous func-

tions but with limited overlap in their expression patterns across

adult tissues (Lutsenko et al., 2007). ATP7A and ATP7B translo-

cate Cu into the lumen of the trans-Golgi network for its incorpo-

ration into secreted cuproenzymes; however, both proteins also

traffic to the cell periphery to efflux Cu in response to increased

Cu concentrations (Hasan and Lutsenko, 2012). Loss-of-func-

tion mutations in ATP7A cause Menkes disease, a neurodegen-

erative disorder in which defective Cu transport from enterocytes

into the general circulation produces a severe Cu deficiency in

several tissues, particularly the brain (Mercer et al., 1993; Vulpe

et al., 1993). Mutations in ATP7B result in Wilson disease, a con-

dition in which patients suffer from hepatic Cu overload due to an

inability to export Cu into the bile, the main pathway for its

ll Reports 10, 933–943, February 17, 2015 ª2015 The Authors 933

Page 3: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

excretion from the body (Bull et al., 1993; Yamaguchi et al.,

1993). How Cu is delivered to mitochondria, where it is used

to metallate cytochrome c oxidase (COX) and a small fraction

of the total cellular SOD1 pool, is unclear, as known Cu chaper-

ones do not appear to function in this capacity (Cobine et al.,

2006).

Historically, studies have investigated the various pathways

that handle Cu independently of each other; however, evidence

from a number of model systems has elegantly demonstrated

that, under limiting conditions, Cu utilization is prioritized at the

cellular, tissue, and systemic levels of organization (Mendelsohn

et al., 2006; Merchant et al., 2006; Nose et al., 2006; Kim et al.,

2008; White et al., 2009). These findings argue that mechanisms

exist to monitor global Cu status and reallocate labile Cu stores.

How discrete Cu-handling pathways are organized into a func-

tional framework that can be dynamically regulated remains an

open question, and one that is of fundamental importance to

our understanding of both basic biology and human disease.

An active role for mitochondria in this process is now appreci-

ated, and it is clear that Cu loading of COX within the mitochon-

drial intermembrane space is tied to the regulation of cellular Cu

efflux (Leary et al., 2007, 2013b). Cu delivery to COX occurs as

the holoenzyme is assembled and is facilitated by as many as

11 accessory proteins (Leary, 2010). The first of these to be iden-

tified was yeast Cox17 (Glerum et al., 1996a), and SCO1 and

SCO2 subsequently were identified as high copy suppressors

of a COX17 mutant strain (Glerum et al., 1996b). Although this

suggested that both Sco1 and Sco2 act downstream of Cox17

to deliver Cu to COX, only the deletion of yeast SCO1 resulted

in a respiratory phenotype (Glerum et al., 1996b). In contrast,

both SCO1 and SCO2 are essential in humans, with mutations

in either gene resulting in a severe, isolated COX deficiency

and an early onset, fatal clinical outcome. SCO2 mutations are

associated primarily with neonatal encephalocardiomyopathy

(Papadopoulou et al., 1999; Jaksch et al., 2000), and virtually

all of the �30 reported pedigrees carry at least one E140K allele

(DiMauro et al., 2012). Patients with SCO1 mutations are rare,

and only three pedigrees thus far have been described (Valnot

et al., 2000; Stiburek et al., 2009; Leary et al., 2013a). Curiously,

each SCO1 pedigree harbors a unique pathogenic allele whose

expression causes amarkedly different clinical course of disease

that primarily affects heart, liver, or brain function.

Affected tissues in SCO1 and SCO2 patients are both COX

and Cu deficient (Leary et al., 2007; Stiburek et al., 2009). While

the combined deficiency is likely a critical facet of disease pro-

gression, the basis for the tissue-specific manifestations in

SCO patients remains enigmatic, and its elucidation requires

the investigation of robust animal models of the associated dis-

eases. Two Sco2 transgenic mouse models have been charac-

terized thus far, and used in innovative translational research

(Viscomi et al., 2011; Cerutti et al., 2014); however, tissues

from these mice do not exhibit a global Cu deficiency, and the

reduction in COX activity is relativelymild compared toSCO2 pa-

tients (Yang et al., 2010). Here, we establish a conditional Sco1

mouse model and use the Cre-lox system to specifically delete

Sco1 in the liver. We show that this mousemodel exhibits a lethal

phenotype associated with profound hepatic COX and Cu defi-

ciencies, much like the patient from the original SCO1 pedigree

934 Cell Reports 10, 933–943, February 17, 2015 ª2015 The Authors

who succumbed to acute liver failure (Valnot et al., 2000). We

further demonstrate that the Cu deficiency in the Sco1-null liver

is explained by a loss of CTR1, via a mechanism that involves

increased proteasomal turnover of the protein. These findings

reveal that a SCO1-dependent mitochondrial signal regulates

Cu homeostasis by modulating cellular Cu import.

RESULTS

Generation of a Liver-Specific Sco1 Knockout MouseModelBecause the patient from the original SCO1 pedigree presented

with a fatal hepatopathy and the most severe clinical course of

disease of allSCO1 patients (Leary et al., 2013a), we constructed

a liver-specific mousemodel of the disease (Figure S1A; Supple-

mental Experimental Procedures). To do this, we first produced

a Sco1 conditional mouse on a C57BL/6N background

(Sco1loxP/loxP), and crossed it to mice expressing Cre recombi-

nase under the control of the liver-specific albumin promoter

(Alb-Cre, Jackson Laboratory) (Postic et al., 1999). We then

backcrossed Alb-Cretg/wt, Sco1loxP/wt animals to Sco1loxP/loxP

mice to generate liver-specificSco1 knockoutmice (Alb-Cretg/wt,

Sco1loxP/loxP), which we obtained at the expected Mendelian

ratios (c2 analysis, p = 0.72 [n = 150]). Molecular analyses

demonstrated that Cre-mediated deletion ofSco1was restricted

to the liver, and that SCO1 protein levels were unaffected in other

somatic tissues (Figures S1B and S1C). Liver-specific Sco1

knockout mice exhibited normal weight gain for roughly the first

28 days of life, but failed to thrive thereafter, with subsequent

weight loss producing a runted phenotype (Figures 1A and 1B).

Liver-specific Sco1 knockout mice had amedian life expectancy

of 70 days (c2 analysis, p < 0.0001) (Figure 1C), which was asso-

ciated with the loss of immunologically detectable SCO1 in the

liver (Figure 1D).

Lethality in Liver-Specific Sco1 Knockout Mice IsAssociated with a Severe Hepatic COX and CuDeficiencyTo further investigate the underlying cause of morbidity andmor-

tality in liver-specific Sco1 knockout mice, we quantified body

and tissue weights and examined gross liver morphology.

Liver-specific Sco1 knockout mice exhibited profound splenic

atrophy, with an initial loss of white pulp and reduced levels of

white blood cells in the circulation, in the absence of any detect-

able changes in the total number or properties of the red blood

cell population (Figures S1D–S1F). While the brain and liver

weights of liver-specific Sco1 knockout mice were unchanged

relative to wild-type littermates, despite the considerable loss

of total body weight (Figure S1D), Sco1-null livers differed mark-

edly with respect to the quality and consistency of their color

(Figure 2A) from roughly postnatal day 37 (P37) onward. Trans-

mission electron microscopy (TEM) analyses revealed that the

spotty nature of Sco1-null livers was attributable to an increase

in lipid droplet accumulation (Figure 2B, asterisks), and thin-layer

chromatography confirmed that there was an age-dependent in-

crease in hepatic cholesterol ester and triacylglycerol content

(Figures 2C and 2D). These findings were further corroborated

by Ingenuity pathway analysis of microarray data, which

Page 4: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Figure 1. Liver-Specific Sco1 Deletion Is

Lethal

(A) Change in body weight (g) over time in liver-

specific Sco1 knockout mice (Sco1liv/liv, n = 38)

and wild-type littermates (Controls, n = 184).

(B) Runted phenotype of a liver-specific Sco1

knockout mouse at P57.

(C) Kaplan-Meier survival curve (Sco1liv/liv, n = 17;

Controls, n = 96).

(D) SCO1, COX IV, and CCS1 abundance in wild-

type (flox) and Sco1-null (liv) livers at P47. Tubulin

served as an internal loading control (see also

Figure S1).

identified genetic reprogramming in Sco1-null livers consistent

with hepatic steatosis and impaired mitosis (Figure S2). TEM

also indicated that marked mitochondrial proliferation had

occurred in Sco1-null livers (Figure 2B, arrows). Consistent

with this observation, the activity of citrate synthase (CS), a

biochemical marker of mitochondrial content, increased signifi-

cantly with time in Sco1-null livers, as did the abundance of

Complexes I, II, and III of oxidative phosphorylation (OXPHOS),

and the steady-state levels of TOM40 and the a subunit of Com-

plex V (Figures 2E and 2F). In contrast, no changes in CS activity

(data not shown) or OXPHOS complex content (Figure 2F) were

observed in the heart, a representative unaffected tissue from

liver-specific Sco1 knockout mice.

In humans, mutations in SCO1 or SCO2 cause tissue-specific

diseases that likely reflect a combined COX and Cu deficiency

in affected tissues (Leary et al., 2007, 2013a). To determine

whether Sco1-null livers recapitulate these clinical hallmarks

of the human diseases, we serially measured the activity of

COX in Sco1-null and wild-type livers at P18, P27, and P57.

Sco1-null livers exhibited a progressive loss of COX activity

and holoenzyme content (Figures 2E and 2F). The total cellular

activity of SOD1, another Cu-dependent enzyme, was also

significantly lower in Sco1-null livers (Figure 2E, inset), and

was accompanied by a concordant increase in the activity (Fig-

ure 2E, inset) and content (Figure 2F) of the matrix-localized,

manganese-dependent enzyme SOD2. Consistent with the

reduced activity of these two Cu-dependent enzymes, Sco1-

null livers were Cu deficient as early as P18, with the severity

of the Cu deficiency worsening with time (Figures 3A and 3B).

The hepatic Cu deficiency preceded changes in tissue zinc

(Zn) and iron (Fe) content (Figure 3B), and H&E and Perls stain-

ing revealed Fe accumulation in the absence of necrosis in the

Cell Reports 10, 933–943,

Kupffer cells of the liver (Figures 3C and

3D) and the reticuloendothelial cells of

the spleen (Figure S1E) in liver-specific

Sco1 knockout mice at P57. Surprisingly,

the severe global Cu deficiency in Sco1-

null livers did not significantly affect the

total hepatic mitochondrial Cu pool (Fig-

ures S3A and S3B) or the Cu levels of

other peripheral organs (Figures 3A and

S3C), apart from a modest reduction in

renal Cu content. These data collectively

demonstrate that SCO1 fulfills essential

functions in COX assembly and the regulation of Cu homeosta-

sis in the liver.

SCO1 Depletion Causes a Severe Hepatic Cu Deficiencyby Attenuating CTR1 ExpressionSco1 deletion in the liver resulted in a marked, age-dependent

reduction in SCO1 abundance (Figure 4A, arrow), with the

upper band of the immunoreactive doublet being the mature

form of the protein, which was barely detectable in crude mito-

chondrial extracts at P18 (Figure S4). While the temporal loss

of SCO1 in Sco1-null livers accounts for the progressive

decrease in COX activity and content (Figures 2E and 2F), the

resultant hepatic Cu deficiency could be explained by enhanced

Cu export or impaired Cu uptake. To begin to distinguish be-

tween these two possibilities, we analyzed the abundance of

the Cu exporters ATP7A and ATP7B in wild-type and Sco1-null

livers at P18, P27, P37, and P57. The steady-state levels of

ATP7B were unchanged in Sco1-null livers over time (Figure 4A),

and biliary Cu content was comparable to that of wild-type litter-

mates (flox, 1.63 ng/ml Cu [pooled sample, n = 7]; liv, 0.92 ±

0.14 ng/ml Cu [n = 5, mean ± SEM]); however, the volume of

bile that was produced by Sco1-null livers increased markedly

starting at P37 (data not shown). Increases in ATP7A abundance

were only evident from P47 onward (Figure S5A), and coincided

with elevated serum content of Cu and Fe (Figure S5B). Consis-

tent with these observations, circulating levels of the Fe carrier

protein ferritin and the enzymatically active form of the Cu-

dependent ferroxidase ceruloplasmin, which both depend on

the secretory pathway for their maturation, also increased (Fig-

ures S5C and S5D). Although these data collectively suggest

that Cu transport into the secretory pathway and biliary Cu

excretion became more active with time in the Sco1-null liver,

February 17, 2015 ª2015 The Authors 935

Page 5: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Figure 2. Liver-Specific Sco1Deletion Is Associatedwith an Isolated COXDeficiency, Loss of SOD1Activity, Mitochondrial Proliferation, and

Accumulation of Intracellular Lipid Droplets

(A and B) (A) Gross morphology and (B) TEM images of a wild-type (flox) and Sco1-null (liv) liver at P57. For TEM images, arrows indicate representative

mitochondria, while the asterisks in the Sco1-null liver denote lipid droplets (3,3403 magnification; scale bar, 2 mm).

(C) Chromatographic analysis of cholesterol ester (CE), triglyceride (TG), fatty acid, and cholesterol content of representative wild-type (flox) and Sco1-null (liv)

livers at P27, P37, and P47.

(D) Quantitation of CE and TG content in Sco1-null livers (liv) and age-matched wild-type (flox) littermates (n = 6 [mean ± SEM]).

(E) COX, CS, and total cellular SOD activities in Sco1-null livers (liv) at P18, P27, and P57 expressed as a percentage of age-matched, wild-type littermates

(liv, n = 4–5; Controls, n = 5–7). (Inset) Percentage contribution of SOD1 and SOD2 to the total SOD activity (mean ± SEM) in Sco1-null (liv, white bars) and wild-

type livers (Controls, black bars) at P57 (liv, n = 4; Controls, n = 5).

(F) SDS-PAGE analysis of the steady-state levels of SOD2, ATPase a, and TOM40, and BN-PAGE analysis of the abundance of OXPHOS Complexes I–III and

COX in livers at P18 and in livers and hearts at P57 (liv, liver-specific Sco1 knockout mice; flox, wild-type littermates) (see also Table S1 and Figure S2).

these changes do not explain the onset of the Cu deficiency,

which occurred well before P37 (Figure 3B).

To evaluate whether the Cu deficiency in Sco1-null livers is

caused by impaired Cu uptake, we measured the abundance

of CTR1 in wild-type and Sco1-null livers at P18, P27, P37,

and P57. Unlike ATP7A and ATP7B, we observed a striking,

936 Cell Reports 10, 933–943, February 17, 2015 ª2015 The Authors

age-dependent reduction in the steady-state levels of the

mature, glycosylated form of CTR1 (Figure 4A, arrow). Indeed,

CTR1 abundance in Sco1-null livers was less than half that of

wild-type littermates at P27 (Figures S6A and S6B). Genome-

wide RNA expression profiling (Table S1) and semiquantitative

RT-PCR (Figure S6C) demonstrated that ablation of Sco1

Page 6: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Figure 3. Liver-Specific Sco1 Deletion

Causes a Severe Hepatic Cu Deficiency

without Markedly Perturbing Cu Homeosta-

sis in Peripheral Tissues

(A) Cu, Fe, and Zn content of tissues from liver-

specificSco1 knockoutmice (liv) at P47 expressed

as a percentage of wild-type littermates (Controls).

Li, liver; K, kidney; Sp, spleen; H, heart; and Br,

brain (liv, n = 5; Controls, n = 8 [mean ± SEM]).

(B) Cu, Fe, and Zn content of Sco1-null livers from

P18 to P57 expressed as a percentage of

age-matched, wild-type littermates (Controls) (liv,

n = 4–5; Controls, n = 4–11 [mean ± SEM]).

(C and D) (C) H&E and (D) Perls staining in wild-

type (flox) and Sco1-null (liv) livers at P57 (scale

bar, 50 mm) (see also Figures S3 and S5).

expression did not grossly affect Ctr1 mRNA levels, suggesting

that SCO1 may be required to regulate the abundance of

CTR1 post-transcriptionally. To determine whether this effect

is specific to SCO1, or a general phenomenon associated with

an isolated COX deficiency, we analyzed null livers from two

additional mouse models that exhibit a COX defect due to dele-

tion of either the heme A:farnesyltransferase gene Cox10 or

Lrpprc, which encodes a factor required for the stability and

handling of mature mitochondrial mRNAs (Sasarman et al.,

2010). The steady-state levels of SCO1weremarkedly increased

in both Lrpprc- and Cox10-null livers (Figures 4B, 4C, and S4C,

arrow); however, the abundance of the mature, glycosylated

form of CTR1 was only affected in Cox10-null livers, where it

was virtually undetectable (Figure 4C, arrow). In contrast, the

abundance of Cu exporter ATP7A was significantly higher in

Lrpprc-null livers compared with age-matched, wild-type litter-

mates (Figure 4B). Since Cox10- and Lrpprc-null livers exhibited

an equivalent COX deficiency (Figure 4D), we conclude that the

observed changes in CTR1 abundance in liver-specific Sco1

knockout mice cannot be explained by a general decrease in

COX activity, but may be a consequence of perturbed SCO1

function. Consistent with this idea, CTR1 levels were reduced

upon stable knockdown of SCO1 to immunologically undetect-

able levels in SCO1 patient fibroblasts (Figure 4E), and its

expression was attenuated but still supported in rho0 cells, which

harbor SCO1 but lack mitochondrial DNA and, therefore, cannot

assemble the COX holoenzyme (Figure 4F). These data collec-

tively suggest that mitochondrial signaling, through a subset of

COX assembly factors that includes SCO1, provides a post-

translational mechanism for the regulation of CTR1 abundance.

Lack of SCO1 Protein Reduces the Abundance of CTR1at the PM and Promotes Its Turnover by the ProteasomePrevious studies have demonstrated that CTR1 abundance is

inversely proportional to cellular Cu status, and that increases

Cell Reports 10, 933–943,

in cellular Cu levels promote the endocy-

tosis of CTR1 from the PM and facilitate

its subsequent degradation (Petris et al.,

2003; Guo et al., 2004). While the cellular

origin and nature of these regulatory in-

puts have yet to be fully defined, our

data implicate mitochondria in this signaling process. To further

investigate how SCO1 contributes to the regulation of CTR1

function, we harvested individual wild-type and Sco1-null livers

at P22, P27, P37, and P47, and used each one for multiple ana-

lyses to ensure the internal consistency of the data. Similar to our

previous findings, Sco1-null livers exhibited an age-dependent

decrease in the abundance of the mature, glycosylated form of

CTR1 (Figure 5A, arrow). While reductions in CTR1 abundance

were accompanied by a concomitant decrease in total liver Cu

content (Figure 5B), the small but significant Cu deficiency we

observed at P18 (Figure 3B) and P22 (Figure 5B) occurred in

the absence of obvious changes in CTR1 levels (Figures 4A

and 5A). We therefore conducted immunofluorescence analyses

to test the hypothesis that the onset of the Cu deficiency inSco1-

null livers is caused by reduced localization of the total CTR1

pool at the PM. We found that, although CTR1 was localized to

the cytosol and PM in both wild-type and Sco1-null livers at

P22 (Figures 5C and S7), there was indeed a modest reduction

in the extent of its coincident localization with Na+/K+ ATPase,

a PM marker protein (overlay correlation coefficients [OCC] =

89.2% ± 4.7% of wild-type, Figure 5B). The intensity of CTR1

staining at the PM in the Sco1-null liver had further declined by

P27, and the overall immunoreactivity was barely detectable at

P37 and P47 (Figure 5C), consistent with the marked reduction

in total CTR1 abundance (Figure 5A). Taken together, these

data suggest that mitochondrial signaling through SCO1 sup-

ports CTR1 localization at the PM and, therefore, Cu import

into the cell.

To further explore the relationship between SCO1 and CTR1,

we interrogated CTR1 in Sco1 knockout mouse embryonic fibro-

blasts (MEFs) (Figures 6A and 6B). Four independent clonal

Sco1�/� MEF lines (KO-2 through KO-5) were all COX and Cu

deficient, and had reduced steady-state levels of CTR1 relative

to control (flox) MEFs (Figures 6C and 6D). Stable overexpres-

sion of a Sco1 cDNA in Sco1�/� MEFs rescued CTR1 levels

February 17, 2015 ª2015 The Authors 937

Page 7: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Figure 4. Lack of SCO1 Protein Leads to a

Diminution in CTR1 Abundance

(A) ATP7A, ATP7B, CTR1, SCO1, and CCS1 abun-

dance in age-matched, wild-type (flox) and Sco1-null

(liv) livers from P18 to P57. Tubulin served as an in-

ternal loading control.

(B and C) ATP7A, CTR1, SCO1, and SCO2 abun-

dance in representative Lrpprc-null (P70); Cox10-null

(P47–P67); and age-matched, wild-type (flox) livers.

Extracts from P47 Sco1-null (liv) and wild-type (flox)

livers served as internal controls, while calnexin or

GAPDH served as loading controls.

(D) Cu content, COX, and SOD1 enzyme activities

(mean ± SEM) in Sco1-, Cox10- (n = 8), and Lrpprc-

(n = 3) null livers expressed as a percentage of that in

age-matched, wild-type littermates (Controls, Cox10

[n = 6]; Lrpprc [n = 3]). Data for Sco1-null livers are

from Figures 2E and 3B, and are shown here for

comparative purposes.

(E) SCO1 and CTR1 in control (C1) and SCO1-2

patient fibroblasts (Leary et al., 2013a) alone (�) or

stably overexpressing an shRNA to abolish SCO1

expression (Leary et al., 2007). Tubulin served as a

loading control.

(F) CTR1, SCO1, COX II, and porin abundance in a

rho+ (143B) and rho0 human osteosarcoma cell line.

Tubulin served as an internal loading control. For

(A–C), the left-hand-side arrow denotes the mature,

mitochondrial form of the SCO1 protein, and the

double asterisk highlights a second immunoreactive

band. For all except (D), the arrow and asterisk on the

right-hand side indicate the mature, glycosylated

CTR1 and the cleaved form of the protein, respec-

tively (see also Figures S4 and S6).

938 Cell Reports 10, 933–943, February 17, 2015 ª2015 The Authors

Page 8: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Figure 5. SCO1 Is Required to Maintain

CTR1 Abundance and Its Localization at

the PM in the Liver

(A) ATP7A, CTR1, and SCO1 abundance in age-

matched, wild-type (flox), and Sco1-null (liv) livers

from P22 to P47. The left-hand-side arrow denotes

the mature, mitochondrial form of the SCO1 pro-

tein, and the double asterisk highlights a second

immunoreactive band. The right-hand-side arrow

denotes mature, glycosylated CTR1, while the

asterisk indicates the cleaved form of the protein.

All samples were immunoblotted on the same

membrane, and tubulin served as an internal

loading control.

(B) Cu content and correlation coefficient (mean ±

SEM) of CTR1 and Na+/K+ ATPase costaining

(OCC, n = 7 for each time point) in Sco1-null livers

expressed as a percentage of age-matched, wild-

type livers (Controls).

(C) Individual and overlay images of the localization

of CTR1 and Na+/K+ATPase in wild-type (Controls)

and Sco1-null (Sco1liv/liv) livers (scale bar, 10 mm)

(see also Figure S7).

and the combined COX and Cu deficiency (Figures 6D and 6E),

demonstrating a SCO1-specific effect. CTR1 levels were also

increased by stable overexpression of a Ctr1 cDNA in

Sco1+/+MEFs; however, no change in its abundance was

observed in Sco1�/� MEFs, suggesting that CTR1 is actively

degraded by the proteasome in the absence of SCO1 expression

(Figure 6E). Consistent with this idea, incubation of Sco1�/�, butnot Sco1+/+, MEFs with the proteasomal inhibitors MG132 (Fig-

ure 7) or lactacystin (data not shown) resulted in a significant

increase in CTR1 abundance. These data further argue that dele-

tion of Sco1 affects a mitochondrial signaling pathway that reg-

ulates high-affinity Cu uptake into the cell by CTR1.

DISCUSSION

Several lines of evidence presented herein directly implicate

SCO1 in the post-translational regulation of the high-affinity Cu

importer CTR1. First, liver-specific deletion of Sco1 caused a

severe Cu deficiency associated with a lack of themature, glyco-

sylated form of CTR1. Second, reductions in SCO1 abundance

attenuated the PM localization of CTR1, and, ultimately, led to

a profound reduction in CTR1 protein levels, without affecting

its steady-state mRNA levels. Third, CTR1 protein levels were

Cell Reports 10, 933–943,

restored in Sco1�/� MEFs by inhibiting

the proteasome or overexpressing a

Sco1 cDNA. Finally, rho0 cells could not

assemble COX yet still expressed SCO1

and CTR1, while SCO1 patient fibroblasts

that lacked immunologically detectable

SCO1 but harbored residual COX activity

(�12% of control; Leary et al., 2013a)

had reduced levels of CTR1. These data

further argue that the observed effect on

CTR1 is largely attributable to SCO1 and

independent of COX activity, and are

consistent with our ability to dissociate the Cu and COX defi-

ciency phenotypes in patient cells (Leary et al., 2007). When

combined with our previous work, which supports the idea that

SCO1 function also impinges on the trafficking and Cu efflux ac-

tivity of ATP7A (Leary et al., 2007, 2013b), these investigations

emphasize that a SCO1-mediated mitochondrial signal is at

the center of a coordinated cellular response to limit Cu

availability.

Like affected tissues in SCO1 and SCO2 patients (Leary et al.,

2007; Stiburek et al., 2009), Sco1-null mouse livers exhibit a

combined COX and Cu deficiency. Ultrastructural, biochemical,

and molecular analyses all strongly suggest that Sco1-null livers

attempt to compensate for the severe, isolated COX deficiency

by increasing mitochondrial content, a classic response to

impaired organelle function observed in many mitochondrial

disorders (DiMauro et al., 2012). Lipid homeostasis is also signif-

icantly perturbed in Sco1-null livers, and there is an age-depen-

dent increase in lipid droplet accumulation. It is, therefore,

tempting to speculate that hepatic steatosis is responsible for

the death of liver-specific Sco1 knockout mice. However, liver-

specific Cox10 deletion also results in profound steatosis,

yet hepatic function eventually normalizes and these mice

remain viable (Diaz et al., 2008). Mice similarly recover following

February 17, 2015 ª2015 The Authors 939

Page 9: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Figure 6. Sco1�/� MEFs Are COX and Cu Deficient Owing to an

Inability to Express Adequate Levels of CTR1

(A and B) (A) PCR genotyping and (B) abundance of SCO1 and COX IV in Sco1-

floxedMEFs cocultured in the presence or absence of Cre recombinase. SD70

served as an internal loading control.

(C) SCO1, CTR1, and ATP7A abundance in parental (flox) and Sco1�/� MEF

clones (KO-2 through KO-4). SD70 served as an internal loading control.

(D) COX activity and total Cu content of 4 distinct Sco1�/� MEF clones alone

(�) or overexpressing a wild-type Sco1 or Ctr1 cDNA, expressed as a per-

centage of the parental MEF lines (flox) (mean ± SEM). ND, not determined.

(E) SCO1, COX I, COX IV, CTR1, and ATP7A abundance in parental (flox) and

Sco1�/�MEF clones (KO-5) alone (�) or overexpressing SCO1orCTR1. Tubulin

served as an internal loading control. For (C and E), the arrow denotes mature,

glycosylated CTR1, while the asterisk indicates the cleaved form of the protein.

liver-specific deletion of Ctr1 (Kim et al., 2009), and there is no

evidence of death in liver-specific Lrpprc knockout mice up to

3 months of age (E. Shoubridge, personal communication).

This suggests that the high frequency of mortality in liver-specific

Sco1 knockout mice also reflects the fact that Sco1-null livers

exhibit the most severe COX and Cu deficiencies of all of these

mouse models.

Perhaps milder models of liver failure are able to repopulate

the failing liver by engaging the proliferation of a subpopulation

of hepatocytes with low or no Cre recombinase activity. Sco1-

null livers appear to be particularly deficient in this regard,

because residual levels of the mature form of the SCO1 protein

are barely detectable at P18, and pathway analysis of our

microarray data indicates that Sco1 deletion significantly im-

pairs the mitotic potential of hepatocytes. It is also possible

that impaired function of other organ systems contributes to

940 Cell Reports 10, 933–943, February 17, 2015 ª2015 The Authors

the mortality of liver-specific Sco1 knockout mice. Fe levels

are ultimately elevated in the kidney and the spleen of liver-

specific Sco1 knockout mice, and the spleen is severely atro-

phied. Hematological and histological analyses indicate that

there is an initial and selective loss of white pulp; however, sub-

sequent Fe accumulation in the reticuloendothelial cells of the

spleen and in the Kupffer cells of the Sco1-null liver are both

consistent with the eventual loss of red pulp function, with

the latter being an established compensatory response aimed

at removing senescent red blood cells from the circulation.

While there are intriguing parallels between our mouse model

and patients with nonalcoholic fatty liver disease, who also pre-

sent with hepatic Cu deficiencies (Aigner et al., 2010), these

patients typically exhibit splenomegaly and not hyposplenism.

Additional studies will be required to tease apart how hepatic

ablation of Sco1 expression impairs the ability of the liver to

communicate with the spleen, thereby impinging upon its

function.

Our data strongly suggest that the Cu deficiency in Sco1-null

livers is driven by changes in the localization and, ultimately,

abundance of CTR1. The mild but significant Cu deficiency in

Sco1-null livers at P18 and P22 occurs in the absence of a

detectable change in CTR1 abundance, but is accompanied

by a modest attenuation in the PM localization of the protein.

The Cu deficiency in Sco1-null livers is markedly worse at P27,

and coincides with a clear reduction in the PM localization and

abundance of CTR1, which is further exacerbated with age. Re-

sidual COX activity is less than 10% of wild-type and barely de-

clines thereafter, arguing that Sco1-null liver mitochondria

already lack SCO1 protein at P27. The progressive reduction in

CTR1 abundance in Sco1-null livers likely reflects its enhanced

proteasomal turnover, because hepatic Ctr1 mRNA levels are

normal and treatment of Sco1�/� MEFs with the proteasomal in-

hibitor MG132 restored CTR1 abundance to wild-type levels.

These findings collectively suggest that SCO1 contributes to

the generation of a mitochondrial signal that is required to pro-

mote the PM localization of CTR1 and/or inhibit its turnover by

the proteasome. Because endocytosis of CTR1 from the PM

and its subsequent degradation are appropriate responses to

cellular Cu overload and reflect an attempt to normalize Cu levels

(Petris et al., 2003), or at least limit further Cu uptake, we favor

the idea that the lack of SCO1mis-signals a state of Cu overload

in the Sco1-null liver. Consistent with this idea, the abundance of

the known biomarker of Cu deficiency CCS1 is lower inSco1-null

livers than it is in wild-type livers, even though it is well known

that CCS1 levels are increased in a Cu-deficient state (Car-

uano-Yzermans et al., 2006). To the best of our knowledge,

this represents the first example of a stimulus other than direct

manipulation of Cu status that regulates CTR1 function. Whether

the SCO1-dependent mitochondrial redox signal that regulates

the trafficking and Cu efflux activity of ATP7A in human fibro-

blasts (Leary et al., 2007, 2013b) also functions to modulate

the activity of CTR1 is not yet known. However, it seems unlikely

that CTR2, which physically interacts with the mature, glycosy-

lated form of CTR1 and facilitates its proteolytic processing (Ohr-

vik et al., 2013), plays a role in this pathway in the liver because

the cleaved CTR1 product does not accumulate in Ctr2- (Ohrvik

et al., 2013), Cox10-, and Sco1-null livers.

Page 10: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Figure 7. Proteasome Inhibition Restores CTR1 Levels in Sco1�/�

MEFs

(A) CTR1 and ATP7A abundance in parental (flox) and Sco1�/� MEF clones

(KO-5) grown in basal media or in media containing MG132. Tubulin served as

an internal loading control. The arrow denotes mature, glycosylated CTR1,

while the asterisk indicates the cleaved form of the protein.

(B) Individual and overlay images of the localization of CTR1 and wheat germ

agglutinin (WGA) in parental (flox) and Sco1�/� MEF clones (KO-5) grown

in basal media or in media containing the MG132 (scale bar, 20 mm). For

(A and B), cells were grown for 6 hr in media containing vehicle (i.e., DMSO) or

40 mM MG132.

Even though there was a clear connection between SCO1 and

CTR1 that, when perturbed, drove the onset and early progres-

sion of the Cu deficiency in Sco1-null livers, hepatic Cu levels

never dropped below �20% of wild-type. Biochemical analyses

indicated that very little of this residual Cu was contained within

COX or SOD1. Yet, like affected cell types from SCO patients

(Dodani et al., 2011), Sco1-null livers preserved the total size of

their mitochondrial Cu pool even in the face of a severe cellular

Cu deficiency. SCO2 abundance is significantly increased in

Sco1-, Cox10-, and Lrpprc-null livers, while livers of transgenic

mice expressing a SCO2 variant equivalent to the pathogenic

E140K allele in SCO2 patients exhibit a mild reduction in mito-

chondrial Cu content in the absence of a change in cellular Cu

Ce

status (Yang et al., 2010). Thus, an attractive postulate is that

SCO2 acts alone or in concert with SCO1 tomodulate the activity

of a transporter that is essential for Cu import (Vest et al., 2013) or

export across the inner mitochondrial membrane. Our data sug-

gest that, although the total mitochondrial Cu pool remains intact

in Sco1-null livers, Cu is preferentially routed to the secretory

pathway once CTR1 levels are barely detectable and the Cu

deficiency has stabilized. Serum levels of Cu and of the secreted

Cu-loaded ferroxidase ceruloplasmin increased considerably

beginning at P37, and the latter observation is unlikely to be

explained by reduced protein turnover because the half-life of

holo-ceruloplasmin in mouse serum is estimated to be roughly

1 day (Gitlin and Janeway, 1960). The increased rate of holo-

ceruloplasmin maturation also was accompanied by a marked

increase in biliary volume in Sco1-null livers relative to wild-

type littermates (�10–203), but we did not observe a concordant

increase in the Cu content of feces (data not shown). At present,

it is unclear if lower affinity Cu uptake systems allow the Sco1-

null liver to maintain a fixed residual Cu content in the absence

of immunologically detectable CTR1.

In summary, we have developed the first animal model of hu-

man disease associated with mutations in SCO1, and used it to

uncover a novel connection between SCO1 function and the

post-translational regulation of the high-affinity Cu importer

CTR1. Our findings expand upon the known role of the mito-

chondrion in the regulation of cellular Cu homeostasis (Leary

et al., 2007, 2013b), and suggest that SCO1 is an essential

component of a mitochondrial signaling pathway that modulates

the activity of both the cellular Cu uptake and efflux machinery.

Why the abundance of this machinery is influenced by the dele-

tion of other COX assembly factors, like Cox10 and Lrpprc, and

whether this involves significant changes in the redox state and

therefore functionality of SCO1 warrant further attention. Future

studies will further explore the functional coupling of SCO1 and

CTR1, and investigate the significance of this coupling to the

regulation of Cu homeostasis in other tissues.

EXPERIMENTAL PROCEDURES

Transgenic Animal Generation and Husbandry

A detailed description of how Sco1 transgenic mice were generated, main-

tained, and manipulated is provided in the Supplemental Experimental Proce-

dures. All animal-related experiments were approved by the Animal Ethics

Review Board at the University of Saskatchewan (AUP #20100091).

Immunoblot Analyses

Mitochondria were isolated frommouse tissues as previously described (Leary

et al., 2013b). For denaturing SDS-PAGE,mitochondrial, whole-tissue, and cell

extracts were prepared on ice in extraction buffer (Potting et al., 2013) supple-

mentedwith a complete protease inhibitor cocktail (Roche) and 0.5mMPMSF,

clarified by centrifugation at 16,600 3 g for 10 min at 4�C, and equal amounts

of protein (10–25 mg/lane) were electrophoresed on 12, 15, or 4%–20% Tris-

HCl gels. For native PAGE, tissue extracts were prepared and equal amounts

of protein (10 mg/lane) were electrophoresed by blue native PAGE on a 5%–

13% gradient gel, as described elsewhere (McKenzie et al., 2007). Equal vol-

umes of serum (2 ml/lane) were electrophoresed at 4�C on 4%–20% Tris-HCl

gels under native conditions to detect the Cu-loaded and apo forms of cerulo-

plasmin (Sato andGitlin, 1991), and under denaturing conditions to detect total

ceruloplasmin content. All gels were transferred to nitrocellulose under semi-

dry conditions and blotted with the appropriate antibodies (see Supplemental

Experimental Procedures). Following incubation with secondary antibody,

ll Reports 10, 933–943, February 17, 2015 ª2015 The Authors 941

Page 11: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

immunoreactive proteins were detected by luminol-enhanced chemilumines-

cence (Pierce). The abundance of the mature, glycosylated form of CTR1

was quantified densitometrically for multiple exposures within the linear range

of the film using ImageQuant software (Leary et al., 2009).

Imaging Analyses

Tissue preparation for various applications is described in detail in the Supple-

mental Experimental Procedures. For immunofluorescence analyses, six

micrometer fixed liver sections mounted on slides were incubated in Tris-buff-

ered saline (TBS [pH 7.4], 25 mM Tris-HCl, 137 mM NaCl, and 2.7 mM KCl)

containing 1% BSA, 0.2% fat free milk, 5% v/v goat serum, and 0.2% Triton

X-100. Slides were washed with PBS, and incubated overnight at 4�C in block-

ing buffer containing polyclonal CTR1 (1:100; Jack Kaplan, University of

Chicago) and monoclonal Na+/K+ ATPase (1:500; Millipore) antibodies. Slides

were washed with PBS, and incubated in blocking buffer containing Alexa

Fluor 488 goat anti-rabbit (1:1,000, Life Technologies) and Alexa Fluor 568

goat anti-mouse (1:500, Life Technologies) secondary antibodies. Slides

were once again washed with PBS, mounted with Prolong Gold Antifade

Mountant containing DAPI (Life Technologies), and dried for 10–15 min.

Stained slides were visualized by confocal laser-scanning microscopy (LSM

700, AxioObserver, Carl Zeiss). Green, red, and DAPI fluorescence was de-

tected using a laser Diodes tuned to 488, 555, and 405 nm, respectively. Fluo-

rescence images were collected simultaneously and OCC were calculated

using a PC running ZEN 2012 software (version 8.0), while Fiji (Schindelin

et al., 2012) or ImageJ software was used to split the channels.

Miscellaneous

Protein concentration and the enzymatic activities of COX, CS, SOD1, and

SOD2 were measured as previously described (Leary et al., 2002, 2004). Sta-

tistical analyses were conducted using Prism (GraphPad), and are detailed

wherever appropriate in the text. Methods related to cell culture experiments

and elemental, lipid, serum, microarray, RT-PCR, and genotyping analyses

are detailed in the Supplemental Experimental Procedures.

ACCESSION NUMBERS

The GEO accession number for the microarray data reported here is

GSE58997.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Supplemental Experimental Procedures,

seven figures, and one table and can be found with this article online at

http://dx.doi.org/10.1016/j.celrep.2015.01.019.

AUTHOR CONTRIBUTIONS

S.C.L. generated the floxed Sco1mouse. C.J.H., B.L., A.H., and Z.N.B. devel-

oped the liver-specific Sco1 knockout mouse model. S.C.L., P.A.C., T.W., and

M.J.P. designed the experiments. F.D. and C.T.M. provided Cox10 floxed

and knockout livers. P.J.M. and S.J.S. conducted the lipid analyses. D.V.

and M.J.P. did the initial IF experiments. C.J.H., B.L., Z.N.B., P.A.C., L.D.Y.,

A.B., T.W., A.H., A.M.E., and S.C.L. executed all other experiments. C.J.H.,

B.L., Z.N.B., and S.C.L. analyzed the data. S.C.L. drafted the manuscript

and P.A.C., T.W., A.M.E., S.J.S., F.D., C.T.M., and M.J.P. provided editorial

feedback.

ACKNOWLEDGMENTS

We wish to acknowledge Drs. Joe Prohaska (University of Minnesota Medical

School), Dennis Winge (University of Utah), Dennis Thiele (Duke University),

Jonathan Gitlin (Marine Biological Laboratory), and Jaekwon Lee (University

of Nebraska-Lincoln) for helpful discussions; Drs. Dennis Thiele, Joe Pro-

haska, and Jack Kaplan (University of Chicago) for CTR1 or CCS1 antiserum;

and Dr. Eric Shoubridge (McGill University) for Lrrprc liver samples. Grants-in-

aid of research to S.C.L. from the Saskatchewan Health Research Foundation,

942 Cell Reports 10, 933–943, February 17, 2015 ª2015 The Authors

the Canadian Institutes for Health Research (CIHR), and the Canadian Gene

Cure Foundation, and to P.A.C. from the National Science Foundation funded

this research. S.C.L. is a New Investigator of the CIHR, and T.W. is a long-term

fellow of the Human Frontiers Science Program.

Received: July 28, 2014

Revised: December 15, 2014

Accepted: January 8, 2015

Published: February 12, 2015

REFERENCES

Aigner, E., Strasser, M., Haufe, H., Sonnweber, T., Hohla, F., Stadlmayr, A.,

Solioz, M., Tilg, H., Patsch, W., Weiss, G., et al. (2010). A role for low hepatic

copper concentrations in nonalcoholic Fatty liver disease. Am. J. Gastroen-

terol. 105, 1978–1985.

Bull, P.C., Thomas, G.R., Rommens, J.M., Forbes, J.R., and Cox, D.W. (1993).

The Wilson disease gene is a putative copper transporting P-type ATPase

similar to the Menkes gene. Nat. Genet. 5, 327–337.

Caruano-Yzermans, A.L., Bartnikas, T.B., and Gitlin, J.D. (2006). Mechanisms

of the copper-dependent turnover of the copper chaperone for superoxide

dismutase. J. Biol. Chem. 281, 13581–13587.

Cerutti, R., Pirinen, E., Lamperti, C., Marchet, S., Sauve, A.A., Li, W., Leoni, V.,

Schon, E.A., Dantzer, F., Auwerx, J., et al. (2014). NAD(+)-dependent activation

of Sirt1 corrects the phenotype in a mouse model of mitochondrial disease.

Cell Metab. 19, 1042–1049.

Cobine, P.A., Pierrel, F., andWinge, D.R. (2006). Copper trafficking to themito-

chondrion and assembly of copper metalloenzymes. Biochim. Biophys. Acta

1763, 759–772.

Desai, V., and Kaler, S.G. (2008). Role of copper in human neurological disor-

ders. Am. J. Clin. Nutr. 88, 855S–858S.

Diaz, F., Garcia, S., Hernandez, D., Regev, A., Rebelo, A., Oca-Cossio, J., and

Moraes, C.T. (2008). Pathophysiology and fate of hepatocytes in a mouse

model of mitochondrial hepatopathies. Gut 57, 232–242.

DiMauro, S., Tanji, K., and Schon, E.A. (2012). The many clinical faces of cyto-

chrome c oxidase deficiency. Adv. Exp. Med. Biol. 748, 341–357.

Dodani, S.C., Leary, S.C., Cobine, P.A., Winge, D.R., and Chang, C.J. (2011). A

targetable fluorescent sensor reveals that copper-deficient SCO1 and SCO2

patient cells prioritize mitochondrial copper homeostasis. J. Am. Chem. Soc.

133, 8606–8616.

Eisses, J.F., and Kaplan, J.H. (2005). The mechanism of copper uptake medi-

ated by human CTR1: a mutational analysis. J. Biol. Chem. 280, 37159–37168.

Festa, R.A., and Thiele, D.J. (2011). Copper: an essential metal in biology. Curr.

Biol. 21, R877–R883.

Festa, R.A., and Thiele, D.J. (2012). Copper at the front line of the host-path-

ogen battle. PLoS Pathog. 8, e1002887.

Gitlin, D., and Janeway, C.A. (1960). Turnover of the copper and protein moi-

eties of ceruloplasmin. Nature 185, 693.

Glerum, D.M., Shtanko, A., and Tzagoloff, A. (1996a). Characterization of

COX17, a yeast gene involved in copper metabolism and assembly of cyto-

chrome oxidase. J. Biol. Chem. 271, 14504–14509.

Glerum, D.M., Shtanko, A., and Tzagoloff, A. (1996b). SCO1 and SCO2 act as

high copy suppressors of a mitochondrial copper recruitment defect in

Saccharomyces cerevisiae. J. Biol. Chem. 271, 20531–20535.

Guo, Y., Smith, K., Lee, J., Thiele, D.J., and Petris, M.J. (2004). Identification of

methionine-rich clusters that regulate copper-stimulated endocytosis of the

human Ctr1 copper transporter. J. Biol. Chem. 279, 17428–17433.

Hamza, I., Schaefer, M., Klomp, L.W., and Gitlin, J.D. (1999). Interaction of the

copper chaperone HAH1 with the Wilson disease protein is essential for cop-

per homeostasis. Proc. Natl. Acad. Sci. USA 96, 13363–13368.

Hasan, N.M., and Lutsenko, S. (2012). Regulation of copper transporters in hu-

man cells. Curr. Top. Membr. 69, 137–161.

Page 12: The Mitochondrial Metallochaperone SCO1 Is …Cell Reports Article The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter

Jaksch, M., Ogilvie, I., Yao, J., Kortenhaus, G., Bresser, H.G., Gerbitz, K.D.,

and Shoubridge, E.A. (2000). Mutations in SCO2 are associated with a distinct

form of hypertrophic cardiomyopathy and cytochrome c oxidase deficiency.

Hum. Mol. Genet. 9, 795–801.

Kim, B.E., Nevitt, T., and Thiele, D.J. (2008). Mechanisms for copper acquisi-

tion, distribution and regulation. Nat. Chem. Biol. 4, 176–185.

Kim, H., Son, H.Y., Bailey, S.M., and Lee, J. (2009). Deletion of hepatic Ctr1

reveals its function in copper acquisition and compensatory mechanisms

for copper homeostasis. Am. J. Physiol. Gastrointest. Liver Physiol. 296,

G356–G364.

Lamb, A.L., Torres, A.S., O’Halloran, T.V., and Rosenzweig, A.C. (2001). Het-

erodimeric structure of superoxide dismutase in complex with its metallocha-

perone. Nat. Struct. Biol. 8, 751–755.

Leary, S.C. (2010). Redox regulation of SCO protein function: controlling cop-

per at a mitochondrial crossroad. Antioxid. Redox Signal. 13, 1403–1416.

Leary, S.C., Hill, B.C., Lyons, C.N., Carlson, C.G., Michaud, D., Kraft, C.S., Ko,

K., Glerum, D.M., and Moyes, C.D. (2002). Chronic treatment with azide in situ

leads to an irreversible loss of cytochrome c oxidase activity via holoenzyme

dissociation. J. Biol. Chem. 277, 11321–11328.

Leary, S.C., Kaufman, B.A., Pellecchia, G., Guercin, G.H., Mattman, A.,

Jaksch, M., and Shoubridge, E.A. (2004). Human SCO1 and SCO2 have inde-

pendent, cooperative functions in copper delivery to cytochrome c oxidase.

Hum. Mol. Genet. 13, 1839–1848.

Leary, S.C., Cobine, P.A., Kaufman, B.A., Guercin, G.H., Mattman, A., Palaty,

J., Lockitch, G., Winge, D.R., Rustin, P., Horvath, R., and Shoubridge, E.A.

(2007). The human cytochrome c oxidase assembly factors SCO1 and

SCO2 have regulatory roles in the maintenance of cellular copper homeosta-

sis. Cell Metab. 5, 9–20.

Leary, S.C., Sasarman, F., Nishimura, T., and Shoubridge, E.A. (2009). Human

SCO2 is required for the synthesis of CO II and as a thiol-disulphide oxidore-

ductase for SCO1. Hum. Mol. Genet. 18, 2230–2240.

Leary, S.C., Antonicka, H., Sasarman, F., Weraarpachai, W., Cobine, P.A.,

Pan, M., Brown, G.K., Brown, R., Majewski, J., Ha, K.C., et al. (2013a). Novel

mutations in SCO1 as a cause of fatal infantile encephalopathy and lactic

acidosis. Hum. Mutat. 34, 1366–1370.

Leary, S.C., Cobine, P.A., Nishimura, T., Verdijk, R.M., de Krijger, R., de Coo,

R., Tarnopolsky, M.A., Winge, D.R., and Shoubridge, E.A. (2013b). COX19me-

diates the transduction of a mitochondrial redox signal from SCO1 that regu-

lates ATP7A-mediated cellular copper efflux. Mol. Biol. Cell 24, 683–691.

Lee, J., Pena, M.M., Nose, Y., and Thiele, D.J. (2002). Biochemical character-

ization of the human copper transporter Ctr1. J. Biol. Chem. 277, 4380–4387.

Lutsenko, S., Barnes, N.L., Bartee, M.Y., and Dmitriev, O.Y. (2007). Function

and regulation of human copper-transporting ATPases. Physiol. Rev. 87,

1011–1046.

McKenzie, M., Lazarou, M., Thorburn, D.R., and Ryan, M.T. (2007). Analysis of

mitochondrial subunit assembly into respiratory chain complexes using Blue

Native polyacrylamide gel electrophoresis. Anal. Biochem. 364, 128–137.

Mendelsohn, B.A., Yin, C., Johnson, S.L., Wilm, T.P., Solnica-Krezel, L., and

Gitlin, J.D. (2006). Atp7a determines a hierarchy of copper metabolism essen-

tial for notochord development. Cell Metab. 4, 155–162.

Mercer, J.F., Livingston, J., Hall, B., Paynter, J.A., Begy, C., Chandrasekhar-

appa, S., Lockhart, P., Grimes, A., Bhave, M., Siemieniak, D., et al. (1993).

Isolation of a partial candidate gene for Menkes disease by positional cloning.

Nat. Genet. 3, 20–25.

Merchant, S.S., Allen, M.D., Kropat, J., Moseley, J.L., Long, J.C., Tottey, S.,

and Terauchi, A.M. (2006). Between a rock and a hard place: trace element

nutrition in Chlamydomonas. Biochim. Biophys. Acta 1763, 578–594.

Nose, Y., Kim, B.E., and Thiele, D.J. (2006). Ctr1 drives intestinal copper ab-

sorption and is essential for growth, iron metabolism, and neonatal cardiac

function. Cell Metab. 4, 235–244.

Ohrvik, H., Nose, Y., Wood, L.K., Kim, B.E., Gleber, S.C., Ralle, M., and Thiele,

D.J. (2013). Ctr2 regulates biogenesis of a cleaved form of mammalian Ctr1

Ce

metal transporter lacking the copper- and cisplatin-binding ecto-domain.

Proc. Natl. Acad. Sci. USA 110, E4279–E4288.

Papadopoulou, L.C., Sue, C.M., Davidson, M.M., Tanji, K., Nishino, I., Sad-

lock, J.E., Krishna, S., Walker, W., Selby, J., Glerum, D.M., et al. (1999). Fatal

infantile cardioencephalomyopathy with COX deficiency and mutations in

SCO2, a COX assembly gene. Nat. Genet. 23, 333–337.

Petris, M.J., Smith, K., Lee, J., and Thiele, D.J. (2003). Copper-stimulated

endocytosis and degradation of the human copper transporter, hCtr1.

J. Biol. Chem. 278, 9639–9646.

Postic, C., Shiota, M., Niswender, K.D., Jetton, T.L., Chen, Y., Moates, J.M.,

Shelton, K.D., Lindner, J., Cherrington, A.D., and Magnuson, M.A. (1999).

Dual roles for glucokinase in glucose homeostasis as determined by liver

and pancreatic beta cell-specific gene knock-outs using Cre recombinase.

J. Biol. Chem. 274, 305–315.

Potting, C., Tatsuta, T., Konig, T., Haag, M., Wai, T., Aaltonen, M.J., and

Langer, T. (2013). TRIAP1/PRELI complexes prevent apoptosis by mediating

intramitochondrial transport of phosphatidic acid. Cell Metab. 18, 287–295.

Puig, S., Lee, J., Lau, M., and Thiele, D.J. (2002). Biochemical and genetic

analyses of yeast and human high affinity copper transporters suggest a

conserved mechanism for copper uptake. J. Biol. Chem. 277, 26021–26030.

Sasarman, F., Brunel-Guitton, C., Antonicka, H.,Wai, T., and Shoubridge, E.A.;

LSFC Consortium (2010). LRPPRC and SLIRP interact in a ribonucleoprotein

complex that regulates posttranscriptional gene expression in mitochondria.

Mol. Biol. Cell 21, 1315–1323.

Sato, M., and Gitlin, J.D. (1991). Mechanisms of copper incorporation during

the biosynthesis of human ceruloplasmin. J. Biol. Chem. 266, 5128–5134.

Schindelin, J., Arganda-Carreras, I., Frise, E., Kaynig, V., Longair, M., Pietzsch,

T., Preibisch, S., Rueden, C., Saalfeld, S., Schmid, B., et al. (2012). Fiji: an

open-source platform for biological-image analysis. Nat. Methods 9, 676–682.

Stiburek, L., Vesela, K., Hansikova, H., Hulkova, H., and Zeman, J. (2009). Loss

of function of Sco1 and its interaction with cytochrome c oxidase. Am. J.

Physiol. Cell Physiol. 296, C1218–C1226.

Turski, M.L., and Thiele, D.J. (2009). New roles for copper metabolism in cell

proliferation, signaling, and disease. J. Biol. Chem. 284, 717–721.

Valnot, I., Osmond, S., Gigarel, N., Mehaye, B., Amiel, J., Cormier-Daire, V.,

Munnich, A., Bonnefont, J.P., Rustin, P., and Rotig, A. (2000). Mutations of

the SCO1 gene in mitochondrial cytochrome c oxidase deficiency with

neonatal-onset hepatic failure and encephalopathy. Am. J. Hum. Genet. 67,

1104–1109.

Vest, K.E., Leary, S.C., Winge, D.R., and Cobine, P.A. (2013). Copper import

into the mitochondrial matrix in Saccharomyces cerevisiae is mediated by

Pic2, a mitochondrial carrier family protein. J. Biol. Chem. 288, 23884–23892.

Viscomi, C., Bottani, E., Civiletto, G., Cerutti, R., Moggio, M., Fagiolari, G.,

Schon, E.A., Lamperti, C., and Zeviani, M. (2011). In vivo correction of COX

deficiency by activation of the AMPK/PGC-1a axis. Cell Metab. 14, 80–90.

Vulpe, C., Levinson, B., Whitney, S., Packman, S., and Gitschier, J. (1993).

Isolation of a candidate gene for Menkes disease and evidence that it encodes

a copper-transporting ATPase. Nat. Genet. 3, 7–13.

Wernimont, A.K., Huffman, D.L., Lamb, A.L., O’Halloran, T.V., and Rose-

nzweig, A.C. (2000). Structural basis for copper transfer by the metallochaper-

one for the Menkes/Wilson disease proteins. Nat. Struct. Biol. 7, 766–771.

White, C., Kambe, T., Fulcher, Y.G., Sachdev, S.W., Bush, A.I., Fritsche, K.,

Lee, J., Quinn, T.P., and Petris, M.J. (2009). Copper transport into the secre-

tory pathway is regulated by oxygen in macrophages. J. Cell Sci. 122, 1315–

1321.

Yamaguchi, Y., Heiny, M.E., and Gitlin, J.D. (1993). Isolation and characteriza-

tion of a human liver cDNA as a candidate gene for Wilson disease. Biochem.

Biophys. Res. Commun. 197, 271–277.

Yang, H., Brosel, S., Acin-Perez, R., Slavkovich, V., Nishino, I., Khan, R., Gold-

berg, I.J., Graziano, J., Manfredi, G., and Schon, E.A. (2010). Analysis of

mouse models of cytochrome c oxidase deficiency owing to mutations in

Sco2. Hum. Mol. Genet. 19, 170–180.

ll Reports 10, 933–943, February 17, 2015 ª2015 The Authors 943