18
BRAIN A JOURNAL OF NEUROLOGY Downregulation of apoptosis-inducing factor in Harlequin mice induces progressive and severe optic atrophy which is durably prevented by AAV2-AIF1 gene therapy Aicha Bouaita, 1,2,3,4 Se ´ bastien Augustin, 1,2,3,4, * Christophe Lechauve, 1,2,3,4, * He ´le ` ne Cwerman-Thibault, 1,2,3,4 Paule Be ´nit, 5 Manuel Simonutti, 1,2,3,4 Michel Paques, 1,2,3,4 Pierre Rustin, 5,6 Jose ´-Alain Sahel 1,2,3,4 and Marisol Corral-Debrinski 1,2,3,4 1 INSERM, U968, Paris, F-75012, France 2 UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France 3 CNRS, UMR_7210, Paris, F-75012, France 4 Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France 5 INSERM, UMR 676, Ho ˆ pital Robert Debre ´ , Paris, F-75019 France 6 Universite ´ Paris 7, Faculte ´ de Me ´ decine Denis Diderot, IFR02, Paris, France *These authors contributed equally to this work. Correspondence to: Marisol Corral-Debrinski, 17 rue Moreau, 75012 Paris, France E-mail: [email protected] The Harlequin mutant mouse, characterized by loss of function of apoptosis-inducing factor, represents a reliable genetic model that resembles pathologies caused by human mitochondrial complex I deficiency. Therefore, we extensively characterized the retinal morphology and function of Harlequin mice during the course of neuronal cell death leading to blindness, with the aim of preventing optic atrophy. Retinas and optic nerves from these mice showed an isolated respiratory chain complex I defect correlated with retinal ganglion cell loss, optic atrophy, glial and microglial cell activation. All of these changes led to irre- versible vision loss. In control mice, retinas AIF1 messenger RNA was 2.3-fold more abundant than AIF2, both messenger RNAs being sorted to the mitochondrial surface. In Harlequin mouse retinas, there was a 96% decrease of both AIF1 and AIF2 messenger RNA steady-state levels. We attained substantial and long-lasting protection of retinal ganglion cell and optic nerve integrity, the preservation of complex I function in optic nerves, as well as the prevention of glial and microglial responses after intravitreal administration of an AAV2 vector containing the full-length open reading frame and the 3 0 untranslated region of the AIF1 gene. Therefore, we demonstrate that gene therapy for mitochondrial diseases due to mutations in nuclear DNA can be achieved, so long as the ‘therapeutic gene’ permits the accurate cellular localization of the corresponding messenger RNA. Keywords: apoptosis-inducing factor; optic atrophy; AAV-mediated gene therapy; respiratory chain complex I; retinal ganglion cells; Harlequin mice Abbreviations: AIF = apoptosis inducing factor; GFAP = glial fibrillary acidic protein; GFP = green fluorescent protein; Hq = Harlequin; PCR = polymerase chain reaction doi:10.1093/brain/awr290 Brain 2012: 135; 35–52 | 35 Received May 19, 2011. Revised September 7, 2011. Accepted September 9, 2011. Advance Access publication November 26, 2011 ß The Author (2011). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: [email protected] at INSERM on July 12, 2012 http://brain.oxfordjournals.org/ Downloaded from

Downregulation of apoptosis-inducing factor in Harlequin mice induces progressive and severe optic atrophy which is durably prevented by AAV2-AIF1 gene therapy

Embed Size (px)

Citation preview

BRAINA JOURNAL OF NEUROLOGY

Downregulation of apoptosis-inducing factor inHarlequin mice induces progressive and severeoptic atrophy which is durably prevented byAAV2-AIF1 gene therapyAicha Bouaita,1,2,3,4 Sebastien Augustin,1,2,3,4,* Christophe Lechauve,1,2,3,4,*Helene Cwerman-Thibault,1,2,3,4 Paule Benit,5 Manuel Simonutti,1,2,3,4 Michel Paques,1,2,3,4

Pierre Rustin,5,6 Jose-Alain Sahel1,2,3,4 and Marisol Corral-Debrinski1,2,3,4

1 INSERM, U968, Paris, F-75012, France

2 UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France

3 CNRS, UMR_7210, Paris, F-75012, France

4 Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France

5 INSERM, UMR 676, Hopital Robert Debre, Paris, F-75019 France

6 Universite Paris 7, Faculte de Medecine Denis Diderot, IFR02, Paris, France

*These authors contributed equally to this work.

Correspondence to: Marisol Corral-Debrinski,

17 rue Moreau, 75012 Paris,

France

E-mail: [email protected]

The Harlequin mutant mouse, characterized by loss of function of apoptosis-inducing factor, represents a reliable genetic model

that resembles pathologies caused by human mitochondrial complex I deficiency. Therefore, we extensively characterized the

retinal morphology and function of Harlequin mice during the course of neuronal cell death leading to blindness, with the aim of

preventing optic atrophy. Retinas and optic nerves from these mice showed an isolated respiratory chain complex I defect

correlated with retinal ganglion cell loss, optic atrophy, glial and microglial cell activation. All of these changes led to irre-

versible vision loss. In control mice, retinas AIF1 messenger RNA was 2.3-fold more abundant than AIF2, both messenger

RNAs being sorted to the mitochondrial surface. In Harlequin mouse retinas, there was a 96% decrease of both AIF1 and AIF2

messenger RNA steady-state levels. We attained substantial and long-lasting protection of retinal ganglion cell and optic nerve

integrity, the preservation of complex I function in optic nerves, as well as the prevention of glial and microglial responses after

intravitreal administration of an AAV2 vector containing the full-length open reading frame and the 30 untranslated region of the

AIF1 gene. Therefore, we demonstrate that gene therapy for mitochondrial diseases due to mutations in nuclear DNA can be

achieved, so long as the ‘therapeutic gene’ permits the accurate cellular localization of the corresponding messenger RNA.

Keywords: apoptosis-inducing factor; optic atrophy; AAV-mediated gene therapy; respiratory chain complex I; retinal ganglion cells;Harlequin mice

Abbreviations: AIF = apoptosis inducing factor; GFAP = glial fibrillary acidic protein; GFP = green fluorescent protein;Hq = Harlequin; PCR = polymerase chain reaction

doi:10.1093/brain/awr290 Brain 2012: 135; 35–52 | 35

Received May 19, 2011. Revised September 7, 2011. Accepted September 9, 2011. Advance Access publication November 26, 2011

� The Author (2011). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved.

For Permissions, please email: [email protected]

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

IntroductionMitochondrial disorders, thought initially to be rare, now appear

to be relatively common (Schaefer et al., 2008). Despite spectacu-

lar progress in elucidating the molecular basis of mitochondrial

respiratory chain diseases in the last decade, a substantial

amount remains to be done regarding therapy (Kerr, 2010; Schon

et al., 2010). Thus, innovative approaches, including gene therapy,

which may offer the real prospect of developing treatments that

improve the underlying condition of patients, should be explored.

Various obstacles may limit success in this goal: (i) the scarcity of

animal models that truly resemble human diseases; (ii) conditional

tissue targeting by gene therapy so as to minimize systemic distri-

bution and avoid harmful side-effects; (iii) difficulty of efficient mito-

chondrial delivery of the gene product; and (iv) as for any Mendelian

disorder, the choice of vectors, delivery method and dosage.

Since the eye is particularly suitable as a target organ for gene

therapy (Colella et al., 2009) and ocular involvement is a frequent

feature in mitochondrial respiratory chain diseases (Carelli et al.,

2009; Yu-Wai-Man et al., 2009), we decided to develop a gene

therapy strategy for preventing optic atrophy in the Harlequin

(Hq) mouse. Hq mice exhibit common characteristics of human

neurodegenerative mitochondriopathies due to respiratory chain

complex I deficiency, such as the degeneration of the cerebellum,

retina, optic nerve, thalamic, striatal and cortical regions (Klein

et al., 2002; Vahsen et al., 2004; Benit et al., 2008). The Hq

phenotype is caused by a severe reduction of apoptosis-inducing

factor (AIF) gene expression due to a retroviral insertion in the first

intron of the gene (Klein et al., 2002). AIF is a flavoprotein

with nicotinamide adenine dinucleotide (NADH) oxidase activity

localized to the mitochondrial inner membrane and involved in

respiratory chain complex I biogenesis and/or stability.

Interestingly, AIF was first discovered as a caspase-independent

death effector able to induce features of apoptosis in isolated

nuclei (Susin et al., 1999; Ye et al., 2002). Recently, a homozy-

gous trinucleotide deletion in exon 5 of the human AIF gene,

leading to the ablation of an arginine residue at position 201 of

the AIF polypeptide, has been described as responsible for a severe

X-linked mitochondrial encephalopathy and oxidative phosphoryl-

ation (OXPHOS) failure in two infant patients (Ghezzi et al.). By

extensively evaluating Hq mouse eyes, we determined that retinal

ganglion cells reached up to 36% of loss relative to control mice,

associated with the disappearance of optic nerve fibres.

Furthermore, the residual fibres presented a clear respiratory

chain complex I deficiency. The overall injury evidenced in the

ganglion cell layer of Hq mice led to progressive vision loss;

thus, 10-month-old mice were permanently blind.

At least four different isoforms of AIF are possibly generated by

alternative splicing of the messenger RNA precursor; in the ner-

vous system AIF1 and AIF2 (Hangen et al., 2010) are the most

abundant. Here we show that in Hq retinas, AIF1 and AIF2

messenger RNA steady-state levels were 96% inferior to control

animals; in controls, AIF1 messenger RNA is 2.3-fold more

abundant than AIF2. Both AIF1 and AIF2 messenger RNAs are

sorted to the mitochondrial surface, as previously reported for

several nuclear transcripts encoding mitochondrial proteins in

human cells (Sylvestre et al., 2003). In an attempt to prevent

retinal ganglion cell loss and optic nerve atrophy in Hq mice we

have constructed a recombinant adeno-associated virus type 2

(AAV2/2_AIF1) which incorporates the mouse AIF1 open reading

frame with its full-length 30-UTR (untranslated region), ensuring

the sorting of the messenger RNA to the mitochondrial surface

and the efficient mitochondrial translocation of the protein. This

point is particularly important because of the pro-apoptotic prop-

erties of AIF if localized to the cytosol (Scovassi et al., 2009). The

prevention of retinal ganglion cell degeneration and the preserva-

tion of respiratory chain complex I activity in optic nerve were

durably achieved in eyes injected intravitreally with AAV2/

2_AIF1. Hence, we established the proof of principle that

rAAV2/2-mediated gene therapy can lead to the prevention of

respiratory chain complex I defects and optic atrophy in Hq mice.

Materials and methods

AnimalsHemizygous (Hq/Y) males were obtained by mating Hq/X females

with either Hq/Y or wild-type males from the colony shipped from

The Jackson Laboratory at �2 months of age.

The Hq strain was B6CBACaAw-J/A-Pdc8Hq/J (http://jaxmice.jax

.org/strain/000501.html). All mice used in this study were F1 mice

bred from founders having a mixed genetic background. Only hemizy-

gous (Hq/Y) males received evaluations and gene therapy; they were

compared exclusively with littermate males from the colony. The mice

were housed from one to four per cage in a temperature-controlled

environment, with a 12-h light/dark cycle and free access to food and

water. Studies were conducted in accordance with the statements on

the care and use of animals in research of the guidelines issued by the

French Ministry of Agriculture and the Veterinarian Department of Paris

(Permit number DF/DF_2010_PA1000298), the French Ministry of

Research (Approval number 5575) and the ethics committees of the

University Paris 6 and the INSERM, Institut National de la Sante et

de la Recherche Medicale (Authorization number 75-1710).

Adeno-associated viral vectors androute for ocular administrationThe entire Mus musculus apoptosis-inducing factor, mitochondrion-

associated 1 (Aifm1) messenger RNA sequence (http://www.ncbi

.nlm.nih.gov/nuccore/NM_012019) of 1926 base pairs (bp) was

synthesized by Genscript Corp, encompassing the original 87 bp of

the 50-UTR, the entire open reading frame encoding a 612 amino

acid-long protein and two restriction sites at the extremities: EcoR1

at the 50 and XhoI at the 30 for cloning into the pAAV internal ribo-

some entry site-humanized recombinant green fluorescent protein

(hrGFP) vector (Stratagene) in which we had earlier replaced the

hGH [human growth hormone 1 (MIM 139250)] polyadenylation

signal with the 176 bp full-length AIF1 30-UTR (http://www.ncbi.

nlm.nih.gov/nuccore/NM_012019) by using BglII and RsrII unique re-

striction sites. AIF1 transcription is under the control of the CMV pro-

moter and the b-globin intron to ensure high levels of expression. The

open reading frame is in frame with the 3� FLAG� sequence at the

C-terminus. The pAAV-internal ribosome entry site-humanized recom-

binant GFP vector also contains a dicistronic expression cassette in

which the humanized recombinant GFP is expressed as a second

36 | Brain 2012: 135; 35–52 A. Bouaita et al.

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

open reading frame translated from the encephalomyocarditis virus

internal ribosome entry site. The final vector, named AAV2/2_AIF1,

contains adeno-associated virus type 2 (AAV2) inverted terminal

repeats, which direct viral replication and packaging. The expression

cassettes, flanked by the two AAV2 inverted terminal repeats, were

encapsidated into AAV2 shells. The final construct was sequenced for

accuracy (Genome Express).

In a previous report, we demonstrated that intravitreal injection of

vector DNA followed by electroporation is successful in transducing rat

retinal ganglion cells and leads to a sustained transgene expression for

up to 90 days (Ellouze et al., 2008). In vivo electroporation of the

AIF1 vector DNA in control mice was efficient in transducing retinal

ganglion cells, which express AIF1 for at least 2 months without any

harm to retinal architecture, optic fibre density or visual function

(data not shown). Nevertheless, since our aim is to prevent durably

the absence of AIF in Hq mice and its deleterious effect on retinal

ganglion cell integrity, we need a protocol permitting both the efficient

transduction of retinal ganglion cells and the long-lasting expression of

the AIF1 transgene. It has been demonstrated that, to date, AAV2/2

represents the best choice in terms of overall transduction efficiency

and tropism for adult rodent retinal ganglion cells (Harvey et al., 2009;

Hellstrom et al., 2009). Moreover, it is well known that AAV2 shows

strong dependence for transduction on heparan sulphate proteogly-

cans that are present in retinal ganglion cells and in the inner limiting

membrane. The inner limiting membrane is a meshwork of extracellu-

lar matrix proteoglycans located at the interface of the vitreous and

the ganglion cell layer. Thus, this first physical barrier may also effect-

ively localize the virus and prevent it from being cleared from the

vitreous via the trabecular meshwork, thereby facilitating subsequent

retinal ganglion cell transduction (Dalkara et al., 2009). For all these

reasons, we decided to target retinal ganglion cells of Hq mice with

the AAV2/2_AIF1 vector, by administration into the vitreous body of

mouse eyes.

Vectors were produced in the Vector Core at the University Hospital

of Nantes (http://www.vectors.nantes.inserm.fr). The AAV titres were

determined by dot blot and expressed as vector genomes per millilitre.

During the course of this study we used two independent productions

with the following titres: 7.5 � 1010 and 2.37 � 1011 vector genomes/

ml. For intravitreal injections, after dilatation of the pupil with topical

1% tropicamide (CibaVision), mice were subjected to anaesthesia with

isoflurane (40 mg/kg body weight). The tip of a 15-mm 33-gauge

needle, mounted on a 10 ml Hamilton syringe (Hamilton Bonaduz

AG) was advanced through the sclera �1 mm posterior to the

corneoscleral limbus in the superior region of the eye, and 2–3 ml of

vector suspension was injected intravitreally, avoiding retinal structure

disruption, bleeding or lens injury. All the injections were performed

under visual control using an ophthalmic surgical microscope. Viral

particles were mixed with 1/10 000 of sterile fluorescein to follow

the homogenous dissemination of the suspension into the vitreous.

Forty-three mice (4- to 8-weeks old) were subjected to AAV2/

2_AIF1 administration; 33 animals received 2.25 � 108 vector gen-

omes/eye (lowest titre batch) and 10 received 7.11 � 108 vector gen-

omes/eye (highest titre batch). Two animals developed a cataract due

to the surgery and three animals died �7 weeks after eye surgery from

natural causes; these five animals were discarded from the study.

Tissue homogenate preparation andrespiratory chain enzymatic assaysAfter rapid and careful dissection, optic nerves or retinas were kept

frozen (�80�C). Samples were prepared at ice-melting temperature by

homogenization of tissues using a 1-ml hand-driven glass–glass Potter

in 200ml of extraction buffer (0.25 mM sucrose, 40 mM KCl, 2 mM

ethylene glycol tetraacetic acid (EGTA), 1 mg/ml bovine serum albu-

min and 20 mM Tris–HCl, pH 7.2). Large cellular debris was spun

down by a low speed centrifugation (3100 rpm, 8 min) and tissue

homogenates were used immediately. Respiratory chain complex I

and V activities were measured using a Cary 50 spectrophotometer

equipped with an 18-cell holder maintained at 37�C (Varian Australia)

as previously described (Benit et al., 2008). Each assay was made in

triplicate with 50 ml of the homogenates obtained. All chemicals were

of the highest grade from Sigma-Aldrich. Complex I

(NADH-ubiquinone oxidoreductase) activity values were either normal-

ized by complex V or converted to specific activities after protein

quantification by the Bradford method. Optic nerves were dissected

from 40 mice subjected to AAV2/2_AIF1 injection; optic nerves from

seven mice were subjected to histological studies. Respiratory chain

assessments obtained from only 28 mice were included in Table 3,

since we failed to measure complex I and complex V activities from

five mouse eye samples due to technical problems encountered during

the assessment.

Fundus imaging by confocal scanninglaser ophthalmoscopy and opticalcoherence tomographyA confocal scanning laser ophthalmoscope (Heidelberg Retina

Angiograph, Heidelberg Engineering) with green laser illumination

was used to examine mice before treatment and each month until

euthanasia. To monitor the nerve fibre layer at each session, up to

four pictures, each from an average of eight images, corresponding to

cardinal areas of the eye fundus were performed. The built-in software

was used for post-processing the images, including alignment,

adjustment of contrast, construction of a mean image and/or of a

composite image (Paques et al., 2006). Spectral-domain optical coher-

ence tomography was performed using the Spectralis� system from

Heidelberg Engineering. The acquisition time per tomographic image,

which includes summation of 10 raw images, was in the order of 1 s

(Grieve et al., 2004). Neural retina thickness for control and Hq eyes

was compared by measuring the distance from the anterior boundary

of the retinal nerve fibre layer and the posterior boundary of the

photoreceptors layer. Throughout the examination process, a quiet

ambiance with dim illumination was maintained. Pupil dilation was

performed with topical 1% tropicamide (CibaVision). Careful slit-lamp

examination before the procedure ruled out the presence of any

corneal or lens opacities. Mice were manually held in front of either

apparatus, in an upright position. As a rule, restraint and rest periods

alternated approximately every 30 s. The examination was interrupted

if the animal showed any sign of tiredness.

Retinal and optic nerve histologyAnimals were sacrificed under deep ketamine (50 mg/kg) xylazine

(10 mg/kg) anaesthesia. After the ocular globes were removed, they

were pierced in the cornea with a needle and fixed for 1 h in 4%

paraformaldehyde at 4�C. After removal of the cornea and lens,

eyecups were post-fixed overnight in 4% paraformaldehyde at 4�C,

cryoprotected by overnight incubation in phosphate-buffered saline

containing 30% sucrose (Sigma-Aldrich) at 4�C. Then, retinas were

embedded in Optimal Cutting Temperature medium (Neg 50;

Richard-Allan Scientific), frozen in liquid nitrogen, and stored at

�20�C. Optic nerves were carefully collected and fixed overnight in

Gene therapy to prevent optic neuropathy Brain 2012: 135; 35–52 | 37

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

4% paraformaldehyde at 4�C. Next day, after washes in phosphate-

buffered saline, tissues were cryoprotected by incubation overnight in

30% sucrose in phosphate-buffered saline. Subsequently, optic nerves

were incubated in a 7.5% gelatin solution from porcine skin; Type A

(Sigma-Aldrich) and 10% sucrose. Samples were frozen between

�50�C and �60�C in 2-methyl-butane solution and then stored at

�20�C. Sections of retinas and optic nerves (transverse and longitudin-

al, near the globe) with a thickness of 10 mm were cut on a cryostat

(Microm Microtech) at �20�C and mounted on SuperFrost�Plus slides.

For immunochemistry, sections of retinas and optic nerves were

washed with phosphate buffer pH 7.4, permeabilized with 1%

Triton X-100 in phosphate buffer for 15 min at room temperature

and treated with 10% normal goat serum, 3% bovine serum albumin,

0.5% Triton and 0.05% Tween-20 in phosphate buffer for 1 h. They

were then incubated with primary antibody overnight at 4�C. The next

day, sections were washed three times in phosphate buffer and

incubated with the appropriate secondary antibodies and DAPI (40,6-

diamidino-2-phenylindole; 2 mg/ml) for 90 min at room temperature

with 3% normal goat serum, 3% bovine serum albumin, 0.5%

Triton and 0.05% Tween-20 in phosphate buffer. Finally, they were

washed three times in phosphate buffer, rinsed with sterile water and

mounted on a glass slide. Primary and secondary antibodies used are

shown in Supplementary Table 1.

Microscopic observations and retinalganglion cell countsImage acquisition of fluorescence labelling was monitored with a fluor-

escence microscope (Leica DM 6000 B) using the MetaVue software

for image acquisitions or a confocal laser scanning microscope

Olympus FV1000. Images were captured with Olympus software

(Olympus). Images were then analysed with Adobe Photoshop� CS

and ImageJ (National Institutes of Health) software. Slides from retinas

and optic nerve sections were also scanned with the Hamamatsu

Nanozoomer Digital Pathology (NDP) 2.0 HT, its fluorescence unit

option (L11600-05) and the NanoZoomer’s 3-CCD TDI camera

(Hamamatsu Photonics). A resolution of 0.92 mm/pixel (�40) was

routinely used.

Retinal ganglion cell counts were performed from each retina on

three entire consecutive cryostat sections with depth at �400 mm

from the optic nerve, using BRN3A immunolabelling as previously

described (Ellouze et al., 2008). BRN3A is a transcription factor

expressed in �80% of all retinal ganglion cells in mouse retina

(Badea et al., 2009) and 490% in rat retina (Nadal-Nicolas et al.,

2009). Manual counting of immunopositive cells was performed after

capturing each whole section in 13–15 images; the identity of each

mouse was unknown for the counting. BRN3A-positive cell numbers

were further validated, for a subset of animals in each group, by per-

forming two additional immunostainings of different retinal sections

(four sections apart from the one used for the first labelling). Retinal

ganglion cell numbers was estimated as a mean for 20 control eyes,

26 eyes from Hq untreated mice and both eyes from 16 Hq mice

subjected to AAV2/2_AIF1 administration in one of their eyes

(12 out of 16 received the vector in their left eyes).

Optomotor testsThe head-tracking method is based on an optomotor test (Thaung

et al., 2002). Vertical black-and-white lines of three varying widths,

subtending 0.0625, 0.125 and 0.25 cycle/�, were presented to the

animal and rotated alternatively clockwise and counterclockwise,

each for 60 s with an interval of 10 s between the two drum rotations.

Head movements were recorded with a video camera mounted above

the apparatus. Animals were scored only when the speed of the head

turn corresponded with the speed of rotation of the stripes (12�/s).

Light levels were kept constant (240 lux).

RNA extractions and real-timequantitative polymerase chain reactionassayRNA preparations from mitochondrion-bound polysomes and free-

polysomes were obtained from 12 brains (cortical region) from

8-week-old C57Bl/6NCrlBR male mice with some modifications of

the protocol previously published for HeLa cells (Sylvestre et al.,

2003). Briefly, after euthanasia, tissues were rinsed in phosphate-

buffered saline before homogenization with a glass Potter homogen-

izer in the freshly prepared extraction buffer [0.32 M sucrose, 30 mM

Tris–HCl pH 7.5, 5 mM AcMg, 100 mM KCl, 1% (vol/vol) protease

inhibitor cocktail (Sigma), 5 mM b-mercapthoethanol, 1 mM PMFS

(Fluka), 0.1% (w/vol) bovine serum albumin fraction V in ultra-pure

distilled water]. The homogenate was centrifuged (1000 rpm, 10 min,

4�C) to eliminate nucleus and unbroken cells. The supernatant was

conserved at 4�C and the pellet was subjected to two additional hom-

ogenizations for optimal cell lysis. The three supernatants were pooled

and were centrifuged at slow speed twice to remove nucleus and

unbroken cells. The supernatants obtained were then centrifuged at

12 500 rpm for 30 min at 4�C; the pellet contained mitochondria and

the supernatant represented the cytosolic fraction. The mitochondrial

fraction was washed three times in the extraction buffer (lacking

bovine serum albumin). The last mitochondrial pellet, containing poly-

somes bound to the outer mitochondrial membrane, was conserved

at �80�C. The cytosolic fraction was gently deposed on a SW41 tube

that contained two layers of 2.0 M sucrose and 0.5 M sucrose

prepared in the extraction buffer. SW41 tubes were centrifuged at

4�C for 20 h at 40 000 rpm. The pellet represented the free-polysome

fraction.

Total RNAs were prepared from mitochondrion-bound polysomes,

free-polysome fractions as well as from wild-type or Hq mouse retinas

using the RNA isolation kit (Qiagen). RNA concentration was deter-

mined with the Nanodrop ND-2000 Spectrophotometer (Thermo

Fisher Scientific) and RNA integrity was verified using agarose gel

electrophoresis. Real-time polymerase chain reaction (PCR) analyses

using mitochondrion-bound polysome and free-polysome fractions

were performed with the Superscript III one-step real-time PCR kit

with Platinum Taq (Invitrogen, Life Technologies).

For real-time quantitative PCR analyses, 1 mg of total RNA from

wild-type or Hq mouse retinas was reverse-transcribed with oligo-dT

in a 20ml final reaction volume using Superscript� II Reverse

Transcriptase (Invitrogen, Life Technologies) following the manufactur-

er’s instructions. The mouse AIF1, AIF2 and AIF1tr (AIF1 messenger

RNA produced from the vector) primers were customized to be highly

specific for each messenger RNA specie. RNAs were purified from

21 mice subjected to AAV2/2_AIF1 intravitreal injection, 13 received

the lowest vector dose and eight received the highest vector dose.

One of the samples from the lowest dose was discarded, since a mix-

ture with another sample led to a confusion of its real identity. Thus,

the results for 20 mice subjected to AAV2/2_AIF1 administration were

presented. In treated mice, the measurement of AIF1 corresponds to

the overall amount of AIF1 messenger RNA (endogenous gene and

AAV2/2_AIF1 driven expression) and the measurement AIF1tr repre-

sents only the abundance of messenger RNA transcribed from the

38 | Brain 2012: 135; 35–52 A. Bouaita et al.

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

vector since one of the primers used recognizes the FLAG epitope

sequence. The primers used in this study are shown in

Supplementary Table 2 and were synthesized by Invitrogen, Life

Technologies.

Quantitative real-time PCR reactions were performed using ABI

7500 Fast (Applied Biosystems). For microplate experiments, 1/100

and 1/500 of the reverse transcription product was used per gene

as template for quantitative PCR reactions with Power Sybr� green

PCR Master Mix (Applied Biosystems, Life Technologies,). Real-time

PCR amplifications were carried out as follows: 10 min 95�C incubation

followed by 40 cycles of 15 s at 95�C and 1 min at 60�C. Each bio-

logical sample was subjected to the assay in triplicate. Obtained Ct

values for each sample allowed AIF1, AIF2, AIF1tr (AIF1 messenger

RNA synthesized from the AAV2/2_AIF1 vector), BRN3A and

gamma-synuclein (SNCG) messenger RNA quantifications using

7500 v.2.0.4 software (Applied Biosystems). The mitochondrial ATP6

gene was selected as the most stable, regarding messenger RNA

steady-state levels, to normalize the messenger RNA species evaluated

and to obtain relative messenger RNA abundance in Hq retinas.

Several reports indicated that BRN3A and SNGC messenger RNA

levels can be used to assess retinal ganglion cell number (Soto et al.,

2008; Surgucheva et al., 2008; Torero Ibad et al., 2011). Therefore,

their estimation in both Hq and control retinas aimed at completing

retinal ganglion cell counts obtained by histological evaluation of

retinal sections.

Statistical analysesValues are expressed as mean � SEM. Data were compared using

Student’s t-test (http://www.u707.jussieu.fr/biostatgv/index.html).

P-values of 50.05 were considered as significant, and 50.01 as

highly significant.

Results

Phenotypic hallmarks of Harlequinmouse eyesWe first studied the viability of retinal ganglion cells in Hq mice

since their long axons form the optic nerve. We examined

eye fundus at different ages using a full-field optical coherence

tomograph (Grieve et al., 2004) (Fig. 1). In control mice of any

age, the nerve fibre bundles converge towards the optic nerve disc

and radiating vessels were noticed in the x–y images (Fig. 1A).

By contrast, stacks of Hq retina x–y images collected from animals

older than 6 months showed a significant diminution of the

number of nerve fibre bundles converging to the optic nerve

disc (Fig. 1B). In reconstructed x–z retinal images (Fig. 1A and

B; right), the nuclear and plexiform layers were recognizable.

Retinal thickness (mm � SEM) was 234.5 � 14.5mm in control

mice independently of their age (n = 4); while in Hq mice values

were 205.75 � 21.75 mm and 143.5 � 9.5 mm for 2- and

13-month-old animals, respectively (n = 3 per age group). The

significant reduction of retinal thickness in old Hq mice compared

with age-matched controls may reflect the retinal neuron loss in all

cell layers previously described in Hq animals older than 8 months

(Klein et al., 2002).

To quantify retinal ganglion cell somas in Hq mice, cryostat

sections of retinas were counted for BRN3A-positive cells in

the ganglion cell layer, since BRN3A is a transcription factor

expressed in �80% of all retinal ganglion cells in mouse retina

Figure 1 In vivo analysis and retinal layering with spectral domain optical coherence tomography imaging of control and Hq retinas.

Representative fundus photographs (left) of control (A) and Hq (B) mice aged between 2 and 14 months. The right panels of A and B

illustrate cross-sectional optical coherence tomography images distal to the optic disk. Retinal thickness was measured by positioning

built-in callipers between the anterior boundary of the retinal nerve fibre layer (NFL) and the posterior boundary of the photoreceptor

layer excluding the retinal pigment epithelium (RPE).

Gene therapy to prevent optic neuropathy Brain 2012: 135; 35–52 | 39

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

Figure 2 Immunofluorescence analysis of retinal and optic nerves from control and Hq mice. (A) Immunostaining for GFAP (green)

and BRN3A (red) of retinas from control and Hq mice aged 1 month; GFAP expression was restricted to the ganglion cell layer (GCL).

The nuclei were contrasted with DAPI (blue). (B) With ageing, Hq mice exhibited a progressive increase in GFAP expression as illustrated in

Hq mice aged 7 and 13 months, respectively. By contrast, age-matched controls only showed a GFAP signal restricted to the ganglion cell

layer. It also appears in this figure that with ageing the Hq mouse inner nuclear layer (INL) and outer nuclear layer (ONL) have thinned

considerably. Scale bar = 50mm. (C) Low magnification images of retinas from 5-month-old and 8-month-old Hq mice and age-matched

controls, GFAP (green) and DAPI (blue) staining are shown. Scale bar = 1 mm. In merge reconstitutions, a 3-fold zoom of the same region is

also illustrated. (D) Independent proximal optic nerve transversal sections from 8-month-old control and Hq mice were immunolabelled with

antibodies against the heavy chain (200 kDa) subunit of neurofilaments, (NF200, green), the tubulin b-III, a microtubule protein (b-III, red)

and the basic myelin protein (MBP, red). Immunoreactivity for the two axonal markers (NF200 and tubulin b-III) shows that axons are largely

lost in Hq mice relative to age-matched controls. Myelin basic protein immunoreactivity was also significantly reduced in Hq mouse optic

nerves, suggesting myelin alterations due to the absence of AIF1 in these mice. Scale bars = 100 and 50mm. IPL = inner plexiform layer.

40 | Brain 2012: 135; 35–52 A. Bouaita et al.

(continued)

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

Figure

2C

ontinued

.

Gene therapy to prevent optic neuropathy Brain 2012: 135; 35–52 | 41

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

(Badea et al., 2009) and 490% in rat retina (Nadal-Nicolas et al.,

2009). We found that retinal ganglion cell somas were signifi-

cantly reduced in Hq retinas: 149 � 27 (n = 26) relative to

233 � 39 in control retinas (n = 20), a diminution of 436% of

the total amount of BRN3A-positive cells in the ganglion cell layer

(P =2.2 � 10�7). Accordingly, the total number of cells in the

ganglion cell layer was also significantly lower in Hq mice:

502.7 � 50.8 relative to 608.4 � 76 in control retinas

(P = 0.00021).

Glial cell activation is an important factor contributing to retinal

ganglion cell death in experimental models of glaucoma (Jiang

et al., 2007; Zhou et al., 2009). In retinas from 1-month-old

control and Hq mice, glial fibrillary acidic protein (GFAP), which

is a sensitive marker of glial activation immunofluorescence, was

confined exclusively to the ganglion cell layer, presumably corres-

ponding to the end-feet of Muller cells and a few astrocytes (Fig.

2A). An obvious increase in GFAP immunoreactivity across the

entire thickness of the retina was observed in Hq retinas from

7 - and 13-month-old mice; while 7- and 13-month-old control

retinas showed GFAP immunostaining restricted to the ganglion

cell layer (Fig. 2B). Interestingly, in retinal sections from 7- and 13-

month-old Hq mice, the diminished number of BRN3A-positive

cells in the ganglion cell layer and the thinness of both the inner

nuclear layer and outer nuclear layer are noticeable, confirming

the neuron cell loss noted in eye fundus images (Fig. 1) and re-

ported earlier (Klein et al., 2002). The reconstruction of whole

retinal sections confirmed the extensive glial response in 5- and

8-month-old Hq mice relative to age-matched controls as well as

the retinal thickness reduction in Hq eyes (Fig. 2C). Next, optic

nerve cross-sections were immunolabelled to observe axonal pro-

files and to search for myelin pathology in Hq mice. Figure 2D

illustrates a recognizable reduction in immunopositive dots in

7-month-old Hq mice relative to age-matched controls when

two antibodies against integral axonal proteins were used (b-III

tubulin and NF200). Moreover, the immunostaining with the anti-

body against the myelin basic protein, one of the most abundant

proteins of the myelin sheath, shows a very weak signal in Hq

mice; whereas intact and strong labelled red rings (myelin basic

protein) were observed in age-matched control (Fig. 2D). Thus,

AIF depletion in the optic nerve induces severe loss of axons along

with oligodendrocyte death; this latter could ultimately lead to

impaired myelination. We also evaluated longitudinal sections of

wild-type and Hq mice for gliosis and microglial activation. For the

latter, we used IBA1 (ionized calcium binding adaptor molecule 1)

since it is upregulated in activated microglia and is involved in cell

migration and phagocytosis (Sasaki et al., 2001). We observed a

weak immunoreactivity for GFAP in optic nerves from a 12-

month-old control, while a strong signal was apparent in optic

nerves from an age-matched Hq mouse. Further, it appears that

IBA1-positive cells were present in higher numbers and with an

increased staining intensity in optic nerves from Hq eyes compared

with the few scattered IBA1-positive cells observed in optic nerves

dissected from an age-matched control (Supplementary Fig. 1).

Additionally, microglial cells in control optic nerves were consist-

ently more ramified (resting state); while IBA1 immunostaining in

optic nerves from Hq mice revealed both ramified cells and round

somata cells with simplified processes (activated state). Hence,

similar to rodent models of optic nerve injury (Bosco et al.,

2008; Sivilia et al., 2009; Morrison et al., 2010) optic nerves

from Hq mice hosted activated astrocytes, microglial cells, as

well as structural disruption of myelination.

Studies of respiratory chain functionFirst, we assessed complex I and complex V activities and

estimated the complex I/complex V ratios in retinas from eight

8-month-old Hq mice and seven age-matched controls

(Table 1). A significant 60% decrease relative to control values

was observed, in both complex I-specific activity and complex

I/complex V ratio, while complex V activity was identical between

Hq and control mice. Secondly, optic nerves were collected either

from animals sacrificed before 8 months of age or older than

8 months. In control optic nerves, complex I, complex V and

their ratio did not change with age, being similar to the ones

measured in retinas (Table 1). Optic nerves from Hq mice aged

58 months already had an �50% decrease of complex I activity,

Table 1 Detection of respiratory chain complex I deficiency in retinas and optic nerves from Hq mice

Ocular tissues Specific activities (nmol/min/mg protein) � SEM Activity ratios � SEM

Complex I Complex V Complex I/V

Retinas from 8-month-old mice (number of eyes tested)

Controls (14) 16.44 � 3.7 68.48 � 8.24 0.278 � 0.071

Harlequin (16) 6.91 � 0.88 75.73 � 11.8 0.090 � 0.014

P-value controls/Harlequin 5.41 � 10�5 0.13 9.15 � 10�6

Optic nerves (age; number of eyes tested)

Controls (2–7 months; 32) 14.12 � 4.53 72.24 � 20.9 0.235 � 0.05

Harlequin (2–7 months; 28) 7.22 � 1.91 70.64 � 29.37 0.143 � 0.025

P-value controls/Harlequin 0.0025 0.898 7.35 � 10�9

Controls (8–13 months; 20) 13.94 � 4.18 70.93 � 17.4 0.243 � 0.055

Harlequin (8–13 months; 28) 3.78 � 1.64 72.33 � 16.9 0.0706 � 0.0176

P-value controls/Harlequin 4.67 � 10�6 0.891 2.58 � 10�10

Measurements of complex I and V activities were obtained by spectrophotometry. Complex I and complex V activities were expressed as nanomoles of oxidized NADH/min/mg protein. In retinas and optic nerves, a complex I defect was shown in young Hq mice; the activity declined in Hq with age. Values represent the mean of triplicatesper each eye sample evaluated.

42 | Brain 2012: 135; 35–52 A. Bouaita et al.

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

a decrease that worsened with age (470% decrease after

8 months of age). Complex V activity was not different between

controls and Hq mice in either age group examined. Thus, in Hq

mice not only did a significant proportion of nerve fibre bundles

disappear (Fig. 1) but complex I enzymatic activity in residual

nerve fibres was severely compromised.

Harlequin mouse visual functionWe next studied the visual function of the Hq mice by the

optokinetic drum test (Thaung et al., 2002; Davies et al., 2007).

We first examined sighted littermate controls (B6CBAC) at two

different ages (43 months and between 6 and 12 months).

They were capable of tracking the moving acuity square-wave

gratings of the three frequencies examined, by making several

head-tracking movements in the same direction and speed as

the drum (Table 2). Hq mice younger than 3 months showed

head-tracking of all three square-wave gratings, indicating that

they were not functionally blind despite their reduced visual per-

formance. Hq mice aged between 4 and 8 months of age spent

much less time tracking across the test period at all three

square-wave gratings relative to age-matched controls. Thus, by

this age there was a significant effect of genotype in the three

grating frequencies, for both the clockwise and counterclockwise

drum rotations (Table 2). Finally, eight Hq mice older than

11 months failed to make head-tracking movements at any of

the grating frequencies in either clockwise or counterclockwise

drum rotation, and could be considered totally blind. The complete

loss of visual function in animals older than 1 year is certainly the

consequence of retinal ganglion cell degeneration, the disappear-

ance of photoreceptors (Figs 1 and 2B) and the disruption of

myelin in the optic nerves (Fig. 2D).

Relative abundance of AIF1 and AIF2messenger RNAs in Hq retinasThe relative abundance of AIF1 and AIF2 transcripts (Hangen

et al., 2010) in retinas from control and Hq mice was determined

by quantitative real-time PCR using the comparative ��Ct

method and the mitochondrial ATP6 as a reference gene, since

its messenger RNA steady-state levels remained almost unchanged

in all the samples evaluated (data not shown). RNA preparations

from 24 retinas of littermate controls aged between 6 and

7 months were examined. AIF1 messenger RNA is 2.3-fold

more abundant than AIF2 in retinas from control mice

(P = 8.2 � 10�12). The difference between AIF1 and AIF2 messen-

ger RNA relative abundance was preserved in 31 Hq retinas, while

the total amount of both messenger RNAs fell to 54% of control

values (Fig. 3A). As SNCG messenger RNA is expressed at high

levels in most if not all retinal ganglion cells and in no other cells

within the adult mouse retina (Soto et al., 2008; Surgucheva et al.,

2008), we determined the relative amount of SNCG and BRN3A

messenger RNAs (Fig. 3B). The steady-state levels of these two

specific markers of retinal ganglion cells were reduced to �50%

of control values in Hq retinas (P-values of 2.3 � 10�8 and

1.8 � 10�11 for BRN3A and SNCG, respectively), confirming ourTab

le2

Opto

moto

rte

sts

of

contr

ol

and

Har

lequin

mic

e

Anim

als

Tra

ckin

gfe

ature

sLi

tter

mat

eco

ntr

ol

mic

e(5

3m

onth

sof

age)

n=

10

Litt

erm

ate

contr

ol

mic

e(6

–12

month

sof

age)

n=

10

Young

Hq

mic

e(5

3m

onth

sof

age)

n=

12

Hq

mic

e(4

–8m

onth

sof

age)

n=

10

Gra

tings

freq

uen

cy(c

ycle

s/�)

0.0

625

0.1

25

0.2

50.0

625

0.1

25

0.2

50.0

625

0.1

25

0.2

50.0

625

0.1

25

0.2

5

Clo

ckw

ise

4.3�

1.9

5.3�

1.7

6�

29.8�

29.3�

2.5

8.9�

2.2

2.6�

1.6

4.2

5�

24.8�

22.1�

1.7

82.3

5�

1.7

3.3�

1.7

5

Counte

r-cl

ock

wis

e5.4�

2.1

6.3�

0.8

58�

1.4

10.4�

2.5

8�

2.3

9.6�

2.2

4.6�

2.1

3.7

5�

1.7

53.7�

1.7

1.7

5�

1.5

52.4�

1.5

82.7�

1.9

Four

diffe

rent

gro

ups

of

mic

ew

ere

eval

uat

edfo

rhea

dtr

acki

ng

move

men

tat

anan

gula

rsp

eed

iden

tica

lto

that

of

the

dru

mro

tation.

The

mic

ew

ere

pre

sente

dw

ith

a2�,4�

and

8�

gra

ting

(corr

espondin

gto

0.2

5,0.1

25

and

0.0

625

cycl

es/�

,re

spec

tive

ly).

They

mad

ese

vera

lhea

dtr

acki

ng

move

men

tsin

the

sam

edirec

tion

and

spee

das

the

dru

m.H

ead

move

men

tssh

ow

nin

Tab

le2

are

pro

vided

per

min

ute

.D

ata

colle

cted

from

Hq

mic

eag

edbet

wee

n4

and

8m

onth

sat

the

thre

egra

ting

freq

uen

cies

for

both

the

clock

wis

ean

dco

unte

r-cl

ock

wis

edru

mro

tations

wer

esi

gnifi

cantly

diffe

rent

toth

ose

of

litte

rmat

eco

ntr

olm

ice

(6–1

2m

onth

sof

age)

:St

uden

t’s

t-te

st;

P=

3.8�

10�

6,

3�

10�

6,

0.0

015,

0.0

0094,

0.0

106,

0.0

0019.

Eight

Hq

mic

eof

about

1ye

arof

age

wer

eal

sosu

bje

cted

toth

ete

stth

ree

tim

esw

ithin

a2-w

eek

tim

ein

terv

al:

they

tota

llyfa

iled

tom

ake

hea

dtr

acki

ng

move

men

tsat

any

of

the

gra

ting

freq

uen

cies

for

eith

ercl

ock

wis

eor

counte

r-cl

ock

wis

edru

mro

tation.

Gene therapy to prevent optic neuropathy Brain 2012: 135; 35–52 | 43

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

data on retinal ganglion cell loss and nerve fibre degeneration

(Fig. 1).

Design of a gene therapy strategy forpreventing retinal ganglion cell andoptic nerve injuryTo efficiently prevent retinal ganglion cell loss and optic nerve

atrophy in Hq mice, AIF protein must be translocated to the mito-

chondrial inner membrane. By studying RNAs isolated from

mitochondrion-bound polysomes and free cytosolic polysomes

isolated from mouse brains we found an enrichment of AIF1

and AIF2 messenger RNAs in mitochondrion-bound polysomes;

indeed, �70% of the overall signal of AIF1 or AIF2 messenger

RNA was found in this fraction (Fig. 4). Next, we selected AIF1 for

gene therapy since its messenger RNA is 2.3-fold more abundant

than AIF2 messenger RNA. We constructed a recombinant AAV

vector containing both the full-length AIF1 open reading frame

and the 30-UTR of the mouse gene to ensure messenger RNA

sorting to the mitochondrial surface. The recombinant vector

was generated using the genome and capsid proteins from sero-

type 2, which has been described as able to efficiently transduce

retinal ganglion cells in mice (Guy et al., 2009; Zhou et al., 2009).

For preventing optic atrophy, 4- to 8-week-old animals were sub-

jected to a single intravitreal injection since at this age the extent

of retinal ganglion cell injury did not lead to noticeable nerve fibre

degeneration (Fig. 1) or vision loss (Table 2).

Effect of rAAV2/2_AIF1 administrationon retinal ganglion cell integrityFour to five months after AAV2/2_AIF1 treatment, we evaluated

the relative amount of the transduced AIF1 messenger RNA by

real-time quantitative PCR in retinas from 20 Hq mice. Twelve

mice received 2.25 � 108 vector genomes; the remaining eight

were treated with 3.16-fold more of AAV2/2_AIF1. We detected

specific messenger RNA species in retina preparations of all the

injected eyes; the steady-state levels of the transduced AIF1

(AIF1tr) messenger RNA contributed to a 14% increase of the

overall amount of AIF1 messenger RNA when 2.25 � 108 vector

genomes were used. Remarkably, mouse retinas from eyes

injected with the highest dose of the vector showed 600-fold

more of the transduced AIF1 messenger RNA. Consequently, the

total amount of AIF1 messenger RNA in retinas from transduced

eyes reached 25.25% of the control value (Fig. 5A). We next

estimated the relative amounts of BRN3A and SNCG messenger

RNAs in retinas from treated and untreated eyes. Independently of

the AAV2/2_AIF1 dose used, a consistent but not statistically sig-

nificant increase of 13.5 and 20%, respectively in their relative

abundance was measured between treated and untreated eyes,

whereas rhodopsin (RHO) messenger RNA abundance remained

unchanged (Fig. 5B). The high variability observed could be

ascribed to the well-known interindividual phenotypic heterogen-

eity, unevenness due to transduction efficiency, but also to the use

of whole retinas in which retinal ganglion cells represent 55% of

the overall cell population. Therefore, we decided to estimate ret-

inal ganglion cell number in retinal sections from 16 mice treated

in one of their eyes with AAV2/2_AIF1 and euthanized �4

months after vector administration. Retinal ganglion cell number

in treated eyes was 31.6% higher than in contralateral untreated

eyes from the same animals (P = 1.4 � 10�9, Fig. 5C). AIF1 ex-

pression driven by the AAV2/2 vector in Hq mice led to an in-

crease of �39% for retinal ganglion cells and 15% for ganglion

cell layer cell population relative to the values obtained in the

overall untreated Hq eye population (P = 1.2 � 10�8 and

0.00034, respectively). Thus, AAV2/2_AIF1 intravitreal administra-

tion led to a remarkable preservation of retinal ganglion cell

Figure 3 AIF1, AIF2, SNCG and BRN3A messenger RNA (mRNA) steady-state levels in control and Hq retinas. Relative fold variations

were calculated using the comparative Ct method and normalized to the mitochondrial ATP6 messenger RNA steady-state levels, since

no variations for the latter were observed among all the RNA preparations evaluated. Data obtained (A and B) correspond to three

independent quantifications (mean � SEM) of RNA preparations from mouse retinas (control n = 24, Hq n = 31). AIF1 and AIF2

messenger RNA steady-state levels are significantly different (Student’s t-test; P = 8.2 � 10�12). The steady-state levels of both BRN3A

and SNCG messenger RNAs were significantly diminished in Hq retinas relative to age-matched controls (P-values of 2.3 � 10�8 and

1.8 � 10�11, respectively). PCR amplifications were performed using specific primers for each gene (Supplementary Table 2).

44 | Brain 2012: 135; 35–52 A. Bouaita et al.

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

integrity since their number reached 89% of the control value

despite remaining statistically different (P = 0.042).

AIF1 expression prevents nerve fibreloss in Hq miceTo determine whether AAV2/2_AIF1 administration could lead to

a better preservation of nerve fibres in Hq mice, eyes fundus

images were collected using confocal scanning laser ophthalmos-

copy (Paques et al., 2006) and optical coherence tomography.

Experiments were performed before vector administration and

monthly until euthanasia, permitting us to follow over time the

disappearance of nerve fibres and look for any change related to

AIF1 transgene expression in treated eyes. Striations of the nerve

fibre layer radiating from the optic nerve disc were clearly visible

for different areas of the eye fundus (nasal, temporal, inferior,

superior). Figure 6A shows a mouse eye fundus image before

(6 weeks) and at different times after vector administration.

A diffuse loss of nerve fibre bundles is clearly visible in the

untreated eye of the mouse at the age of 22 weeks, especially

in the superior and temporal superior areas. In the bottom panel

of Fig. 6A, images collected for the eye that received AAV2/

2_AIF1 are illustrated, for the areas shown, tracks of axons were

preserved. Thus, axon loss gives the impression of being less

important than in the untreated eye, indicating that AIF1 expres-

sion protected against nerve fibre degeneration. Figure 6B illus-

trates fundus imaging obtained by optical coherence tomography

in Hq mouse before euthanasia; it is noticeable that nerve fibre

bundles are more abundant in the treated eye than in the untreat-

ed eye. Moreover, the reconstructed x–z retinal images show irre-

gularities in the retinal structure in the untreated eye, especially in

the nerve fibre layer and ganglion cell layer which appear as

broader regions of high scattering intensity, while the disorgan-

ization of cell architecture is less notable in the treated eye.

Haematoxylin and eosin staining of AAV2/2_AIF1 treated Hq

eye and contralateral untreated eye confirms the optical coherence

tomography results since it appears that the nerve fibre layer is

thicker in the treated eye, as if more retinal ganglion cell axons are

converging to the optic nerve head. The axonal profiles, detected

by immunohistochemistry for NF200 and b-III tubulin, in optic

nerve cross-sections confirmed that treated Hq eyes presented

an increase in neurofilament and microtubule-immunopositive

signals relative to untreated eyes (Fig. 6C). This indicates that

AAV2/2_AIF1 administration prevents loss of axons within the

nerve and substantiates retinal ganglion cell number estimations

in eyes subjected AAV2/2_AIF1 treatment (Fig. 5C). Conversely,

no changes in the myelin basic protein-immunostaining were

noticed in treated eyes relative to untreated ones. This result

indicates that oligodendrocytes residing in the optic nerve cannot

be transduced by the vector after intravitreal administration.

Remarkably, optic nerve sections immunolabelled for GFAP and

IBA1 showed weaker fluorescence signals in the treated eye rela-

tive to the untreated one. Immunofluorescence for both GFAP and

IBA1 in the optic nerve section shown were comparable between

the eye subjected to vector administration and a control sample

from a mouse of approximately the same age (Supplementary

Fig. 2). Accordingly, AIF1 expression driven by AAV2/2_AIF1

appears to lead to a beneficial effect on optic fibre integrity and

a diminution of gliosis and microglial activation in optic nerves.

Complex I activity is maintained in opticnerves from AAV2/2_AIF1-treatedanimalsWe next assessed complex I enzymatic activity in optic nerves

from mice expressing the AIF1 transgene to determine whether

the beneficial effects observed in both retinal ganglion cell overall

number and nerve fibre density could result from preserved

Figure 4 Subcellular distribution of AIF1 and AIF2 messenger

RNAs in mouse brains. Steady-state levels of different

messenger RNAs were compared in the mitochondrion-bound

polysome (M-P) and free-polysomes (F-P) fractions depending

on the gene examined, as follows: mitochondrial ATP6: 50 ng

of RNA for each fraction and 18 cycles; glyceraldehyde-3-

phosphate dehydrogenase, GAPDH: 50 ng for each fraction

and 23 cycles; adenylate kinase isoenzyme 4, AK4: 100 ng

for each fraction and 30 cycles; apoptosis-inducing factor,

mitochondrion-associated 1 (AIF1) encoding the isoform AIF1:

250 ng for each fraction and 30 cycles; apoptosis-inducing

factor, mitochondrion-associated 2 (AIF2) encoding the isoform

AIF2: 250 ng for each fraction and 30 cycles. PCR amplifications

were performed using specific primers for each gene

(Supplementary Table 2) and three independent RNA purifica-

tions. Each specific primer leads to the amplification of a PCR

product of 150 bp. Densitometric analyses were performed with

signals revealed by electrophoresis (top: 1/10 of the real-time

PCR reaction for the ATP6 messenger RNA and 1/5 for the other

messenger RNAs). Bar graphs (bottom) represent the amount of

each messenger RNA species in the mitochondrion-bound

polysome fraction relative to the total signal measured in

mitochondrion-bound polysome and free-polysome fractions.

The intensity of each signal was quantified using the Quantity

One� 1-D Analysis Software (Bio-Rad laboratories).

Gene therapy to prevent optic neuropathy Brain 2012: 135; 35–52 | 45

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

Figure 5 Evaluation of AIF1 transgene expression and its impact on retinal ganglion cell integrity. (A) Relative quantification of

endogenous AIF1 messenger RNA and AIF1 messenger RNA transcribed from the AAV2/2_AIF1 vector in Hq retinas. For vector

administration, intravitreal injections were performed in one eye with either 2.25 � 108 vector genomes (VG) or 7.11 � 108 vector

genomes. (B) The impact of AAV2/2_AIF1 treatment on retinal ganglion cell integrity was estimated by the evaluation of SNCG and

46 | Brain 2012: 135; 35–52 A. Bouaita et al.

(continued)

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

complex I activity. Twenty-eight mice were subjected to intravi-

treal injection of AAV2/2_AIF1 at ages between 4 and 8 weeks;

eight of these animals received the highest dose of the vector. All

the mice were euthanized at 16–20 weeks after vector adminis-

tration. AIF1 expression driven by AAV2/2_AIF1 led to a signifi-

cant protection against complex I defect, since both complex I

activity and complex I/complex V ratios in treated eyes attained

the values measured in age-matched controls and were statistically

different to those measured in the untreated fellow eyes (Table 3).

By contrast as expected, values measured in untreated eyes were

very similar to the ones measured in untreated Hq mice of

approximately the same age (group 2–7 months; Table 1) con-

firming that the observed difference is a direct consequence of the

increased AIF protein level in treated eyes. Noticeably, in all the

samples assessed, complex V enzymatic activities were unchanged.

Thus, AAV2/2_AIF1 administration to Hq mouse eyes, before an

extensive retinal ganglion cell loss, neutralized the deleterious

effect of AIF depletion on complex I function; and this was

independent of vector dose used, showing that a 14% increase

in the overall amount of cellular AIF1 messenger RNA (Fig. 5A)

is sufficient to protect against complex I deficiency.

DiscussionBecause the above experiments target the multi-faceted AIF

protein, a number of conclusions can be drawn from this study.

They first deal with AIF function. Throughout the study, it is clear

that AIF depletion in the Hq retinas and optic nerves, far from

reducing cell death processes, rather generates retinal ganglion

cell death; this latter process is robustly counteracted by AAV2/

2_AIF1. Besides, the consequence of AIF depletion seems to be

directly correlated with severely defective respiratory chain com-

plex I activity. Thus, these results fully confirm the previous

observation that the alternative function of AIF, as an assembly

factor for the respiratory chain, determines the in vivo mouse

phenotype associated with AIF depletion (Vahsen et al., 2004;

Benit et al., 2008). A similar observation was recently reported

in humans, where a deleterious mutation in AIF triggers a typical

mitochondrial respiratory chain disease hallmarked by severe

encephalopathy (Ghezzi et al., 2010).

It has been suggested that the AIF2 isoform mediates physio-

logical function only in the differentiating neurons (Hangen et al.,

2010). We observed that increased levels of AIF1, the major iso-

form we have identified in mouse retinas, are sufficient to protect

retinal ganglion cell and optic nerve integrity in Hq mice. So far,

in the context of eye diseases resulting from mitochondrial dys-

function, two approaches have been used to get round the ab-

sence of available experimental models. First, mitochondrial

dysfunction has been triggered by blockade of complex I by rote-

none. Complex I inhibition was subsequently counteracted by

the allotopic expression of the yeast Ndi1 protein which success-

fully counteracts the effect of rotenone by providing a bypass

for matrix NADH oxidation (Marella et al., 2010). Next, impair-

ing complex I activity by the allotopic expression of a mutant

ND4 gene was shown to be readily neutralized by expressing

its wild-type counterpart (Ellouze et al., 2008). While these

approaches demonstrate the feasibility of gene therapy in

these particular models of complex I-associated eye diseases,

the Hq mouse strain provides an authentic genetic model for

a mitochondrial respiratory chain complex I disease affecting

the eye.

Down regulation of AIF gene expression in Hq mice is respon-

sible for a severe respiratory chain complex I defect and leads to

the progressive degeneration of neurons in the cerebellum, thal-

amus and striatum with a massive glial reaction (El Ghouzzi et al.,

2007). We have shown in Hq mice that, despite variable pheno-

typic progression across mice, the loss of retinal ganglion cells

reached up to 36% of control values in Hq mice aged 6–9

months. The decrease of retinal ganglion cell number is associated

with a noticeable diminution in the density of optic fibre bundles

converging to the optic nerve disc and within the optic nerve the

number of axons was greatly reduced. As in the brain (El Ghouzzi

et al., 2007), Hq retinas and optic nerves exhibited a strong gliosis

reaction that occurred early in the course of retinal ganglion cell

degeneration. We also demonstrated a marked activation of

microglia in Hq optic nerves. In this respect, Hq optic nerve path-

ology bears a resemblance to rodent models of glaucoma induced

by acute intraocular hypertension; in these animals an early and

widespread GFAP overexpression in both retina and brain occurred

(Zhang et al., 2009) as well as a marked activation of microglia in

the retina, optic nerve and optic tract (Ebneter et al., 2010). It can

be hypothesized that impairment of mitochondrial function is a

key event during the course of glaucoma as suggested recently

in the DBA/2 J mouse model. Hence, their optic nerves possess a

lower bioenergetic reserve that correlates with exposure to raised

intraocular pressure (Baltan et al., 2010). We also demonstrated

that the deleterious impact of AIF depletion in Hq optic nerves

starts in young animals, as the respiratory chain complex I defect

Figure 5 ContinuedBRN3A messenger RNA steady-state levels in untreated or treated Hq eyes. AAV2/2_AIF1 administration did not influence the

relative RHO messenger RNA amounts as shown. Data obtained correspond to three independent experiments per RNA preparation

(mean � SEM) of mouse retina [control n = 20, Hq untreated or treated eyes (2.25 � 108 vector genomes; n = 12) and Hq untreated or

treated eyes (7.11 � 108 vector genomes; n = 8)]. (C) Retinal ganglion cell (RGC) number estimations were performed after immuno-

labelling for BRN3A antibody and DAPI staining; this latter allowed estimation of total nuclei in the ganglion cell layer (GCL). The

histograms showed the means � SEM obtained after all of the total and BRN3A cells in the ganglion cell layer were counted in three

independent retinal sections per animal. Thirty-two Hq mouse eyes were evaluated: 16 were subjected to AAV2/2_AIF1 administration in

one eye (12 left eyes and four right eyes); their fellows were untreated. Animals were euthanized between 16 and 20 weeks after vector

administration. Values were compared with retinal ganglion cell number obtained for 20 controls and 26 Hq mice that were not subjected

to gene therapy.

Gene therapy to prevent optic neuropathy Brain 2012: 135; 35–52 | 47

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

Figure 6 Protection against retinal ganglion cell damage in Hq eyes treated with AAV2/2_AIF1. (A) Eye fundus images obtained by

scanning laser ophthalmoscopy from an Hq mouse, in which one eye has been subjected to AAV2/2_AIF1 injection when the animal was

6 weeks old. Images were collected from both eyes before vector injection and at different times until euthanasia, performed 20 weeks

after vector administration. Different regions of the retina were illustrated: face up, NI (nasal inferior), T (temporal), S (superior) and

TI (temporal inferior). It appears that the untreated eye may have undergone a more severe degenerative process of the nerve fibres

relative to the treated eye. (B) Spectral domain optical coherence tomography and light microscopic images of treated (left) and untreated

(right) eyes from one Hq mouse in which AAV2/2_AIF1 administration was performed at the age of 1 month; the animal was sacrificed

16 weeks later. En face images show more nerve fibres bundles in the treated eye than in the untreated one (left). Low-magnification

images from the treated and untreated eyes of the same Hq mouse after haematoxylin and eosin staining of retinal sections are also

illustrated. Note that more retinal ganglion cell axons converged to the optic nerve head in the treated eye relative to the untreated eye.

(C) Immunofluorescence analysis of optic nerve cross-sections from control and Hq mice. Independent proximal optic nerve transversal

sections (near the globe) from a 7-month-old control animal and an Hq mouse were immunolabelled with antibodies against the heavy

chain (200 kDa) subunit of neurofilaments, (NF200, green), the tubulin b-III, a microtubule protein (b-III, red) and the basic myelin protein

(MBP, red). The Hq mouse shown was subjected to a single intravitreal injection of AAV2/2_AIF1 (7.11 � 108 vector genomes) in its left

eye at 8 weeks of age and euthanized 5 months later. Immunoreactivity for the two axonal markers (NF200 and tubulin b-III) shows

a noticeable prevention of axon loss in the treated eye relative to the untreated one. Indeed, the density of either green (NF200) or red

(b-III) dots was similar to the one observed in control mouse. Conversely, myelin basic protein immunofluorescence signal did not

change by the AAV2/2_AIF1 administration. Scale bars = 100 and 50 mm.

48 | Brain 2012: 135; 35–52 A. Bouaita et al.

(continued)

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

Figure 6 Continued.

Gene therapy to prevent optic neuropathy Brain 2012: 135; 35–52 | 49

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

was significantly reduced in mice younger than 8 months relative

to age-matched controls (�50% of control values); the deficiency

reached its greatest level in mice older than 8 months (�30%

activity relative to controls). The significance of these observations

is 2-fold: (i) depletion of the AIF protein in retinal ganglion

cells leads to early cell death correlated with the disappearance

of optic fibres; and (ii) when animals presented up to 36% of

retinal ganglion cell loss, in the residual axons the respiratory

chain complex I defect was aggravated, suggesting that, eventu-

ally, the majority of retinal ganglion cells and their axons could

completely disappear. Retinal ganglion cell injury, optic nerve

degeneration and respiratory chain complex I defect certainly

contributed to vision loss; indeed, visual capabilities of Hq mice

progressively decline with age and 1-year-old mice can be con-

sidered completely and irreversibly blind. Moreover, in adult Hq

mice (6–8 months of age); myelin basic protein expression is

strongly diminished in optic nerves, indicating an impaired mylina-

tion process that may cause disruption of the optic nerve integrity

and contribute to visual function loss.

Interestingly, patients harbouring an AIF mutation (Ghezzi et al.,

2010) had abnormal visual evoked potentials: a delay in conduc-

tion and abnormal cortical waves with distortion of the recorded

potentials (G. Uziel and M. Zeviani, personal communication).

Since visual evoked potential measures the conduction of the

visual pathway from the optic nerve to the occipital cortex,

abnormal amplitudes can reflect retinal ganglion cell loss and

optic atrophy, as observed in Hq mice. Our data on retinal gan-

glion cell physiopathology and onset were an incentive to envisage

a gene therapy in Hq eyes before the disappearance of retinal

ganglion cells and their axons become measurable.

A number of conclusions emerge from our successful attempt at

gene therapy in Hq mouse optic atrophy. First, we strengthened

our previous observation that the specific sorting of the ‘thera-

peutic gene’ messenger RNA to the mitochondrial surface works

efficiently to ensure the delivery of the therapeutic protein inside

the organelle. This is particularly obvious in the case of AIF, whose

abnormal cytosolic distribution is known to readily trigger

apoptosis (Scovassi et al., 2009). In keeping with this, since AIF

messenger RNAs localize preferentially to the mitochondrial

surface in mouse brain, we have designed an AAV2/2 vector

specifying the AIF1 protein but also possessing the full-length

30-UTR of the AIF gene, to ensure the sorting of the correspond-

ing messenger RNA to the mitochondrial surface. Then by using

serotype 2 we confirmed numerous reports demonstrating that it

displays a high retinal ganglion cell transduction efficiency after

intravitreal injection (Harvey et al., 2002; Leaver et al., 2006;

MacLaren et al., 2006; Hellstrom et al., 2009). Thus, the present

study describes the first successful example of the ocular targeting

of a mitochondrial protein in the context of AAV-mediated gene

delivery. AIF1 gene expression driven by the vector appears stable

for up to 5 months post-injection without any negative effect on

retinal architecture or function. AIF1 messenger RNA steady-state

levels in transduced retinas increased depending on the vector

doses used; the low dose induced an �14% increase in the overall

messenger RNA AIF1 abundance while with the highest dose AIF1

messenger RNA reached �25% of the value measured in control

mouse retinas. Consequently, we assume that retinal ganglion cell

transduction yield was high and generated sustained AIF1 gene

expression. Importantly, AIF1 transgene expression was effect-

ive in: (i) improving retinal ganglion cell survival; (ii) preserving

nerve fibre integrity; (iii) reducing microglial activation and

gliosis reaction; and (iv) stabilizing respiratory chain complex I

function in optic nerves. Undeniably, retinal ganglion cell loss

was almost completely prevented since eyes subjected to

AAV2/2_AIF1 administration presented a retinal ganglion cell

population reaching �89% of the value measured in control

mice. Moreover in optic nerves from treated eyes, which axon

number was clearly superior to that in untreated animals, respira-

tory chain complex I activity was not statistically different com-

pared with the one measured in age-matched controls (P = 0.33;

Table 3).

In conclusion, the work presented here corroborates our previ-

ous study in a rat Leber’s hereditary optic neuropathy (LHON)

model in which replacement gene therapy, based on messenger

RNA sorting to the mitochondrial surface, was effective in pre-

venting the deleterious effects on retinal ganglion cell and optic

nerve integrity caused by a mutation in the mitochondrial ND4

gene (Ellouze et al., 2008).

Table 3 Complex I and complex V activity assessment in optics nerves from Harlequin mice treated withAAV2/2_AIF1

Optic nerves (n = 28) Complex I � SEM Complex V � SEM Complex I/V � SEM

Untreated eyes 7.43 � 1.98 69.12 � 14.3 0.128 � 0.042

Treated eyes 12.08 � 3.04 72.21 � 11.2 0.20 � 0.061

Student test: P-values

Treated/untreated 0.00074 0.625 0.00014

Treated/Hq 0.00043 0.89 0.0006

Treated/Controls 0.335 0.99 0.07

Untreated/Hq 0.82 0.89 0.21

The results from 28 animals subjected to intravitreal administration of AAV2/2_AIF1 at an approximate age of 6 weeks are shown. The averagetime for euthanasia was 4–5 months after the ocular intervention; thus mice were all younger than 8 months at the time of the assessment.Complex I and complex V activities were expressed as nanomoles of oxidized NADH/min/mg protein. Data were compared using Student’s t-test(http://www.u707.jussieu.fr/biostatgv/index.html). The comparison between animals subjected to vector administration and the whole controlor Hq population was performed using the values shown in Table 2 for the group of animals younger than 8 months.

50 | Brain 2012: 135; 35–52 A. Bouaita et al.

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

With a view towards future clinical trials in patients with mito-

chondrial impairment, the main objective of this study was to test

and validate preventive gene therapy of AIF1 driven by AAV2/2.

This has proven largely successful, as the vector clearly forced

expression of AIF1 in the recipient Hq mouse retinas, protected

mitochondrial respiratory chain complex I function and prevented

retinal ganglion cells and their axons from degeneration.

We have thus established the proof of principle that: (i) mes-

senger RNA sorting to the mitochondrial surface represents a

powerful tool for ensuring the efficient delivery of therapeutic

proteins inside the organelle; and (ii) the prevention of OXPHOS

failure is sufficient to protect against retinal ganglion cell and optic

nerve degeneration in Hq mouse strain; which is a faithful model

of human disease. Pivotally, since ocular involvement is a frequent

feature in mitochondrial diseases (Fraser et al., 2010; Yu-Wai-Man

et al., 2010) and since clinical trials based on AAV-mediated

gene therapy for retinal dystrophies are ongoing in three labo-

ratories (Bainbridge et al., 2008; Maguire et al., 2008;

Cideciyan, 2010) our study opens the door to counteracting

mitochondrial dysfunction in future clinical studies.

AcknowledgementsThe authors thank Dr Aubrey de Grey for careful reading and

feedback on final version of the manuscript. We are grateful

to David Godefroy and Stephane Fouquet (Cellular Imaging

Facility of the Institut de la Vision), respectively, for their help in

Nanozoomer� slides scanning, confocal laser scanning microscopy

and for useful discussions and comments on the histological

evaluations of the study.

FundingINSERM; the CNRS; l’Agence Francaise pour la Recherche (ANR);

l’Association Francaise contre les Myopathies (AFM); l’Agence

Francaise contre les Maladies Mitochondriales (AMMi); la

Fondation Voir et Entendre.

Supplementary materialSupplementary material is available at Brain online.

ReferencesBadea TC, Cahill H, Ecker J, Hattar S, Nathans J. Distinct roles of tran-

scription factors brn3a and brn3b in controlling the development,

morphology, and function of retinal ganglion cells. Neuron 2009;

61: 852–64.Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K,

et al. Effect of gene therapy on visual function in Leber’s congenital

amaurosis. N Engl J Med 2008; 358: 2231–9.

Baltan S, Inman DM, Danilov CA, Morrison RS, Calkins DJ, Horner PJ.

Metabolic vulnerability disposes retinal ganglion cell axons to dysfunc-

tion in a model of glaucomatous degeneration. J Neurosci 2010; 30:

5644–52.

Benit P, Goncalves S, Dassa EP, Briere JJ, Rustin P. The variability of the

harlequin mouse phenotype resembles that of human mitochondrial-

complex I-deficiency syndromes. PLoS ONE 2008; 3: e3208.

Bosco A, Inman DM, Steele MR, Wu G, Soto I, Marsh-Armstrong N,

et al. Reduced retina microglial activation and improved optic nerve

integrity with minocycline treatment in the DBA/2J mouse model of

glaucoma. Invest Ophthalmol Vis Sci 2008; 49: 1437–46.

Carelli V, La Morgia C, Valentino ML, Barboni P, Ross-Cisneros FN,

Sadun AA. Retinal ganglion cell neurodegeneration in mitochondrial

inherited disorders. Biochim Biophys Acta 2009; 1787: 518–28.Cideciyan AV. Leber congenital amaurosis due to RPE65 mutations and

its treatment with gene therapy. Prog Retin Eye Res 2010; 29:

398–427.Colella P, Cotugno G, Auricchio A. Ocular gene therapy: current progress

and future prospects. Trends Mol Med 2009; 15: 23–31.Dalkara D, Kolstad KD, Caporale N, Visel M, Klimczak RR, Schaffer DV,

et al. Inner limiting membrane barriers to AAV-mediated retinal

transduction from the vitreous. Mol Ther 2009; 17: 2096–102.Davies VJ, Hollins AJ, Piechota MJ, Yip W, Davies JR, White KE, et al.

Opa1 deficiency in a mouse model of autosomal dominant optic at-

rophy impairs mitochondrial morphology, optic nerve structure and

visual function. Hum Mol Genet 2007; 16: 1307–18.

Ebneter A, Casson RJ, Wood JP, Chidlow G. Microglial activation in the

visual pathway in experimental glaucoma: spatiotemporal characteriza-

tion and correlation with axonal injury. Invest Ophthalmol Vis Sci

2010; 51: 6448–60.

El Ghouzzi V, Csaba Z, Olivier P, Lelouvier B, Schwendimann L,

Dournaud P, et al. Apoptosis-inducing factor deficiency induces early

mitochondrial degeneration in brain followed by progressive multifocal

neuropathology. J Neuropathol Exp Neurol 2007; 66: 838–47.Ellouze S, Augustin S, Bouaita A, Bonnet C, Simonutti M, Forster V, et al.

Optimized allotopic expression of the human mitochondrial ND4

prevents blindness in a rat model of mitochondrial dysfunction. Am J

Hum Genet 2008; 83: 373–87.

Fraser JA, Biousse V, Newman NJ. The neuro-ophthalmology of

mitochondrial disease. Surv Ophthalmol 2010; 55: 299–334.

Ghezzi D, Sevrioukova I, Invernizzi F, Lamperti C, Mora M, D’Adamo P,

et al. Severe X-linked mitochondrial encephalomyopathy associated

with a mutation in apoptosis-inducing factor. Am J Hum Genet

2010; 86: 639–49.

Grieve K, Paques M, Dubois A, Sahel J, Boccara C, Le Gargasson JF.

Ocular tissue imaging using ultrahigh-resolution, full-field optical

coherence tomography. Invest Ophthalmol Vis Sci 2004; 45: 4126–31.

Guy J, Qi X, Koilkonda RD, Arguello T, Chou TH, Ruggeri M, et al.

Efficiency and safety of AAV-mediated gene delivery of the human

ND4 complex I subunit in the mouse visual system. Invest Ophthalmol

Vis Sci 2009; 50: 4205–14.

Hangen E, De Zio D, Bordi M, Zhu C, Dessen P, Caffin F, et al. A

brain-specific isoform of mitochondrial apoptosis-inducing factor:

AIF2. Cell Death Differ 2010; 17: 1155–66.Harvey AR, Hellstrom M, Rodger J. Gene therapy and transplantation in

the retinofugal pathway. Prog Brain Res 2009; 175: 151–61.Harvey AR, Kamphuis W, Eggers R, Symons NA, Blits B, Niclou S, et al.

Intravitreal injection of adeno-associated viral vectors results in the

transduction of different types of retinal neurons in neonatal and

adult rats: a comparison with lentiviral vectors. Mol Cell Neurosci

2002; 21: 141–57.

Hellstrom M, Ruitenberg MJ, Pollett MA, Ehlert EM, Twisk J,

Verhaagen J, et al. Cellular tropism and transduction properties of

seven adeno-associated viral vector serotypes in adult retina after

intravitreal injection. Gene Ther 2009; 16: 521–32.

Jiang C, Moore MJ, Zhang X, Klassen H, Langer R, Young M. Intravitreal

injections of GDNF-loaded biodegradable microspheres are

neuroprotective in a rat model of glaucoma. Mol Vis 2007; 13:

1783–92.Kerr DS. Treatment of mitochondrial electron transport chain disorders: a

review of clinical trials over the past decade. Mol Genet Metab 2010;

99: 246–55.

Gene therapy to prevent optic neuropathy Brain 2012: 135; 35–52 | 51

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from

Klein JA, Longo-Guess CM, Rossmann MP, Seburn KL, Hurd RE,Frankel WN, et al. The Harlequin mouse mutation downregulates

apoptosis-inducing factor. Nature 2002; 419: 367–74.

Leaver SG, Cui Q, Plant GW, Arulpragasam A, Hisheh S, Verhaagen J,

et al. AAV-mediated expression of CNTF promotes long-term survivaland regeneration of adult rat retinal ganglion cells. Gene Ther 2006;

13: 1328–41.

MacLaren RE, Buch PK, Smith AJ, Balaggan KS, MacNeil A, Taylor JS,

et al. CNTF gene transfer protects ganglion cells in rat retinaeundergoing focal injury and branch vessel occlusion. Exp Eye Res

2006; 83: 1118–27.

Maguire AM, Simonelli F, Pierce EA, Pugh EN Jr, Mingozzi F, Bennicelli J,et al. Safety and efficacy of gene transfer for Leber’s congenital

amaurosis. N Engl J Med 2008; 358: 2240–8.

Marella M, Seo BB, Thomas BB, Matsuno-Yagi A, Yagi T. Successful

amelioration of mitochondrial optic neuropathy using the yeast NDI1gene in a rat animal model. PLoS One 2010; 5: e11472.

Morrison JC, Cepurna Ying Guo WO, Johnson EC. Pathophysiology of

human glaucomatous optic nerve damage: insights from rodent

models of glaucoma. Exp Eye Res 2010; 93: 156–64.Nadal-Nicolas FM, Jimenez-Lopez M, Sobrado-Calvo P, Nieto-Lopez L,

Canovas-Martinez I, Salinas-Navarro M, et al. Brn3a as a marker of

retinal ganglion cells: qualitative and quantitative time course studies in

naive and optic nerve-injured retinas. Invest Ophthalmol Vis Sci 2009;50: 3860–8.

Paques M, Simonutti M, Roux MJ, Picaud S, Levavasseur E, Bellman C,

et al. High resolution fundus imaging by confocal scanning laserophthalmoscopy in the mouse. Vision Res 2006; 46: 1336–45.

Sasaki Y, Ohsawa K, Kanazawa H, Kohsaka S, Imai Y. Iba1 is an

actin-cross-linking protein in macrophages/microglia. Biochem

Biophys Res Commun 2001; 286: 292–7.Schaefer AM, McFarland R, Blakely EL, He L, Whittaker RG, Taylor RW,

et al. Prevalence of mitochondrial DNA disease in adults. Ann Neurol

2008; 63: 35–9.

Schon EA, DiMauro S, Hirano M, Gilkerson RW. Therapeutic prospectsfor mitochondrial disease. Trends Mol Med 2010; 16: 268–76.

Scovassi AI, Soldani C, Veneroni P, Bottone MG, Pellicciari C. Changes of

mitochondria and relocation of the apoptosis-inducing factor duringapoptosis. Ann N Y Acad Sci 2009; 1171: 12–7.

Sivilia S, Giuliani A, Fernandez M, Turba ME, Forni M, Massella A, et al.

Intravitreal NGF administration counteracts retina degeneration

after permanent carotid artery occlusion in rat. BMC Neurosci 2009;10: 52.

Soto I, Oglesby E, Buckingham BP, Son JL, Roberson ED, Steele MR,

et al. Retinal ganglion cells downregulate gene expression and lose

their axons within the optic nerve head in a mouse glaucomamodel. J Neurosci 2008; 28: 548–61.

Surgucheva I, Weisman AD, Goldberg JL, Shnyra A, Surguchov A.

Gamma-synuclein as a marker of retinal ganglion cells. Mol Vis

2008; 14: 1540–8.Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM,

et al. Molecular characterization of mitochondrial apoptosis-inducing

factor. Nature 1999; 397: 441–6.Sylvestre J, Margeot A, Jacq C, Dujardin G, Corral-Debrinski M. The role

of the 3’ untranslated region in mRNA sorting to the vicinity of

mitochondria is conserved from yeast to human cells. Mol Biol Cell

2003; 14: 3848–56.Thaung C, Arnold K, Jackson IJ, Coffey PJ. Presence of visual head

tracking differentiates normal sighted from retinal degenerate mice.

Neurosci Lett 2002; 325: 21–4.

Torero Ibad R, Rheey J, Mrejen S, Forster V, Picaud S, Prochiantz A,et al. Otx2 promotes the survival of damaged adult retinal ganglion

cells and protects against excitotoxic loss of visual acuity in vivo.

J Neurosci 2011; 31: 5495–503.

Vahsen N, Cande C, Briere JJ, Benit P, Joza N, Larochette N, et al. AIFdeficiency compromises oxidative phosphorylation. EMBO J 2004; 23:

4679–89.

Ye H, Cande C, Stephanou NC, Jiang S, Gurbuxani S, Larochette N, et al.DNA binding is required for the apoptogenic action of apoptosis

inducing factor. Nat Struct Biol 2002; 9: 680–4.

Yu-Wai-Man P, Griffiths PG, Chinnery PF. Mitochondrial optic neuropa-

thies - disease mechanisms and therapeutic strategies. Prog Retin EyeRes 2010; 30: 81–114.

Yu-Wai-Man P, Griffiths PG, Hudson G, Chinnery PF. Inherited

mitochondrial optic neuropathies. J Med Genet 2009; 46: 145–58.

Zhang S, Wang H, Lu Q, Qing G, Wang N, Wang Y, et al. Detection ofearly neuron degeneration and accompanying glial responses in the

visual pathway in a rat model of acute intraocular hypertension.

Brain Res 2009; 1303: 131–43.Zhou X, Li F, Kong L, Chodosh J, Cao W. Anti-inflammatory effect of

pigment epithelium-derived factor in DBA/2J mice. Mol Vis 2009; 15:

438–50.

52 | Brain 2012: 135; 35–52 A. Bouaita et al.

at INSE

RM

on July 12, 2012http://brain.oxfordjournals.org/

Dow

nloaded from