insights into the mechanisms of macular degeneration associated

13
Insights into the mechanisms of macular degeneration associated with the R172W mutation in RDS Shannon M. Conley 1 , Michael W. Stuck 1 , Justin L. Burnett 1 , Dibyendu Chakraborty 1 , Seifollah Azadi 1 , Steven J. Fliesler 2,3 and Muna I. Naash 1, 1 Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Boulevard, BMSB 781, Oklahoma City, OK 73104, USA, 2 Research Service, Veterans Administration Western New York Healthcare System, Buffalo, NY 14215, USA and 3 Departments of Ophthalmology (Ross Eye Institute) and Biochemistry, University at Buffalo-S.U.N.Y, Buffalo, NY 14215, USA Received November 11, 2013; Revised and Accepted January 9, 2014 Mutations in the photoreceptor tetraspanin gene peripherin-2/retinal degeneration slow (PRPH2/RDS) cause both rod- and cone-dominant diseases. While rod-dominant diseases, such as autosomal dominant retinitis pig- mentosa, are thought to arise due to haploinsufficiency caused by loss-of-function mutations, the mechanisms underlying PRPH2-associated cone-dominant diseases are unclear. Here we took advantage of a transgenic mouse line expressing an RDS mutant (R172W) known to cause macular degeneration (MD) in humans. To facili- tate the study of cones in the heavily rod-dominant mouse retina, R172W mice were bred onto an Nrl 2/ 2 back- ground (in which developing rods adopt a cone-like fate). In this model the R172W protein and the key RDS-binding partner, rod outer segment (OS) membrane protein 1 (ROM-1), were properly expressed and traf- ficked to cone OSs. However, the expression of R172W led to dominant defects in cone structure and function with equal effects on S- and M-cones. Furthermore, the expression of R172W in cones induced subtle alterations in RDS/ROM-1 complex assembly, specifically resulting in the formation of abnormal, large molecular weight ROM-1 complexes. Fundus imaging demonstrated that R172W mice developed severe clinical signs of disease nearly identical to those seen in human MD patients, including retinal degeneration, retinal pigment epithlium (RPE) defects and loss of the choriocapillaris. Collectively, these data identify a primary disease-causing mo- lecular defect in cone cells and suggest that RDS-associated disease in patients may be a result of this defect coupled with secondary sequellae involving RPE and choriocapillaris cell loss. INTRODUCTION The product of the peripherin-2/retinal degeneration slow (PRPH2/RDS) gene, hereafter referred to as RDS, is a photorecep- tor-specific tetraspanin protein exclusively localized to the disc rims of both cone and rod outer segments (OSs) (1). The protein is critical for the proper formation and maintenance of OSs, and specifically for building the rim structure (2,3). To date over 80 dif- ferent mutations in the RDS gene have been linked with human dis- eases, including autosomal dominant retinitis pigmentosa (adRP), digenic RP, pattern dystrophy, adult vitelliform macular dystrophy, central areolar choroidal dystrophy and other forms of macular de- generation (MD) (http://www.retina-international.org/sci-news/ rdsmut.htm). Yet, no curative or preventative therapies for these conditions exist. RDS function relies on its assembly into protein complexes, which initially are either homotetramers or heterotetramers, the latter containing RDS’ non-glycosylated homologue ROM-1 (rod OS membrane protein-1); these complexes are held together via interactions between the second intradiscal (D2) loop of the two proteins (4,5). RDS and ROM-1 function together, assem- bling in the inner segment (cell body) of the photoreceptor into tetrameric core complexes (6). These complexes are then traf- ficked to the OS where they further assemble into higher order oligomeric structures, including hetero-octamers and RDS homo- oligomers (7). These larger complexes are held together by To whom correspondence should be addressed. Tel: +1 4052718001 ex 47969; Fax: +1 4052713548; Email: [email protected] # The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: [email protected] Human Molecular Genetics, 2014, Vol. 23, No. 12 3102–3114 doi:10.1093/hmg/ddu014 Advance Access published on January 25, 2014 Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898 by guest on 09 April 2018

Upload: vanthuy

Post on 11-Feb-2017

223 views

Category:

Documents


0 download

TRANSCRIPT

Insights into the mechanisms of maculardegeneration associated with the R172Wmutation in RDS

Shannon M. Conley1, Michael W. Stuck1, Justin L. Burnett1, Dibyendu Chakraborty1,

Seifollah Azadi1, Steven J. Fliesler2,3 and Muna I. Naash1,∗

1Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Boulevard, BMSB

781, Oklahoma City, OK 73104, USA, 2Research Service, Veterans Administration Western New York Healthcare

System, Buffalo, NY 14215, USA and 3Departments of Ophthalmology (Ross Eye Institute) and Biochemistry, University

at Buffalo-S.U.N.Y, Buffalo, NY 14215, USA

Received November 11, 2013; Revised and Accepted January 9, 2014

Mutations in the photoreceptor tetraspanin gene peripherin-2/retinal degeneration slow (PRPH2/RDS) causeboth rod- and cone-dominant diseases. While rod-dominant diseases, such as autosomal dominant retinitis pig-mentosa, are thought to arise due to haploinsufficiency caused by loss-of-function mutations, the mechanismsunderlying PRPH2-associated cone-dominant diseases are unclear. Here we took advantage of a transgenicmouse line expressing an RDS mutant (R172W) known to cause macular degeneration (MD) in humans. To facili-tate the study of cones in the heavily rod-dominant mouse retina, R172W mice were bred onto an Nrl2/2 back-ground (in which developing rods adopt a cone-like fate). In this model the R172W protein and the keyRDS-binding partner, rod outer segment (OS) membrane protein 1 (ROM-1), were properly expressed and traf-ficked to cone OSs. However, the expression of R172W led to dominant defects in cone structure and functionwith equal effects on S- and M-cones. Furthermore, the expression of R172W in cones induced subtle alterationsin RDS/ROM-1 complex assembly, specifically resulting in the formation of abnormal, large molecular weightROM-1 complexes. Fundus imaging demonstrated that R172W mice developed severe clinical signs of diseasenearly identical to those seen in human MD patients, including retinal degeneration, retinal pigment epithlium(RPE) defects and loss of the choriocapillaris. Collectively, these data identify a primary disease-causing mo-lecular defect in cone cells and suggest that RDS-associated disease in patients may be a result of this defectcoupled with secondary sequellae involving RPE and choriocapillaris cell loss.

INTRODUCTION

The product of the peripherin-2/retinal degeneration slow(PRPH2/RDS) gene, hereafter referred to as RDS, is a photorecep-tor-specific tetraspanin protein exclusively localized to the discrims of both cone and rod outer segments (OSs) (1). The proteinis critical for the proper formation and maintenance of OSs, andspecifically for building the rim structure (2,3). To date over 80 dif-ferent mutations in the RDS gene have been linked with human dis-eases, including autosomal dominant retinitis pigmentosa (adRP),digenicRP,patterndystrophy,adultvitelliformmaculardystrophy,central areolar choroidal dystrophy and other forms of macular de-generation (MD) (http://www.retina-international.org/sci-news/

rdsmut.htm). Yet, no curative or preventative therapies for theseconditions exist.

RDS function relies on its assembly into protein complexes,which initially are either homotetramers or heterotetramers, thelatter containing RDS’ non-glycosylated homologue ROM-1(rod OS membrane protein-1); these complexes are held togethervia interactions between the second intradiscal (D2) loop of thetwo proteins (4,5). RDS and ROM-1 function together, assem-bling in the inner segment (cell body) of the photoreceptor intotetrameric core complexes (6). These complexes are then traf-ficked to the OS where they further assemble into higher orderoligomeric structures, including hetero-octamers and RDS homo-oligomers (7). These larger complexes are held together by

∗To whom correspondence should be addressed. Tel: +1 4052718001 ex 47969; Fax: +1 4052713548; Email: [email protected]

# The Author 2014. Published by Oxford University Press. All rights reserved.For Permissions, please email: [email protected]

Human Molecular Genetics, 2014, Vol. 23, No. 12 3102–3114doi:10.1093/hmg/ddu014Advance Access published on January 25, 2014

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

intermolecular disulfide bonds mediated by a specific cysteineresidue (C150) (8,9), one of seven D2 loop cysteines (the othersare all involved in the intramolecular disulfide bonding necessaryfor proper folding of the D2 loop). In the absence of C150, RDSand ROM-1 tetramers form, but not higher order oligomers. Intransgenic mice that express C150S mutant RDS in the absenceof wild-type (WT) RDS, OSs fail to form, confirming that cova-lently bound, higher order RDS complexes are required forproper photoreceptor OS biogenesis (9). Although RDS/ROM-1complexes are similar in rods and cones (7), we have shown thatthe two cell types have differential requirements for RDS(10,11). While rods without RDS form no OSs (12), coneswithout RDS (e.g. in the Nrl2/2 background) form open OSsthat lack rim structures and conventional flattened membranouslamellae, but nonetheless retain appreciable levels of retinal func-tion (10). However, the reason why some RDS mutations result inrod-dominant retinal diseases (such as adRP), while others areassociated with cone-dominant diseases (such as MD) is notknown.

One of the most common RDS mutations is a substitution oftryptophan for arginine at position 172 (R172W), whichresults in autosomal dominant macular dystrophy. This mutationhas been described by multiple groups and occurs in a largenumber of families (13–16). Key patient phenotypes includecentral vision loss, clinically detected macular changes (e.g.by ophthalmoscopy) and atrophy of the choriocapillaris andretinal pigment epithelium (RPE) (13,15). While full-field elec-troretinograms (ERGs) can be normal, the multi-focal ERG isalmost always decreased. Affected individuals typicallypresent with faltering visual acuity in the third to fourth decadeof life, although macular changes can be detected in asymptom-atic children and young adults carrying the mutation (15). Al-though it is unclear why the R172W mutation causes thisdistinct cone-associated phenotype, having arginine at position172 in RDS is critical for cone structure and function. Each ofthe known substitutions at position 172 (R172W, R172Q)causes a cone- or a fovea-dominant defect in patients (15,17)in contrast to some other residues in which mutations at thesame site can cause both rod- and cone-dominant phenotypes(such as K153D) (18) or (N244H/K) (19,20).

Our previous studies have suggested that the underlyingdisease mechanism for RDS mutations that cause rod-dominantdisease (such as C214S) may be haploinsufficiency (21,22).However, the mechanisms underlying RDS-associated cone-dominant or macular disease are more complex. To study thesedisease mechanisms we generated and characterized transgenicmice carrying the R172W mutation on multiple rds genetic back-grounds (23,24). We showed that expression of R172W caused asevere dominant-negative defect in cone function, consistentwith patient phenotypes, while rod function was unaffected or,in some cases, improved (e.g. in R172W mice on the rds+/2 orrds2/2 backgrounds) (24). This dominant effect is not due to ex-cessive levels of RDS, since we have previously shown that over-expression of WT RDS does not have any structural or functionalconsequences to the retina (25). Biochemical studies showedthat the R172W RDS protein was slightly more susceptible totryptic digestion than WT RDS, but retained the ability to bindROM-1 and form proper RDS complexes in rods (24). Theseobservations are consistent with improved, rather than defective,rod structure and function in the presence of R172W protein.

However, the small number of cones in the WT mouse retina,which account for only �3% of the total photoreceptor popula-tion, precluded studies on the function of R172W protein incones.

Hence, we undertook additional studies to extend our under-standing of the mechanism of cone-dominant disease in thepresence of the R172W mutation in RDS. To facilitate character-ization of cones, we took advantage of the Nrl2/2 (neural retinalleucine zipper) knockout mouse, in which developing rods are con-verted to cone-like cells (26). Herein, we show that R172W miceon the Nrl2/2 background also exhibit defects in cone vision,consistent with their counterparts on the WT background. Weobserved that the formation of RDS/ROM-1 complexes in conesof the R172W mice was altered, suggesting that this moleculardefect may underlie the cell-type specific disease phenotype. Inaddition, we show that the R172W mice exhibited clinical signsof disease consistent with those seen in patients, suggesting thatthis model has potential utility for investigating the relationshipbetween molecular defects and the development of this type ofretinal degeneration.

RESULTS

Expression and localization of R172W in the Nrl2/2 retina

To facilitate our studies on the mechanism of disease in the caseof the R172W mutation, we cross-bred mice expressing theR172W transgene on different rds backgrounds onto theNrl2/2 genetic background (herein also referred to as the ‘cone-dominant background’). Throughout this study, we comparedthe characteristics of the R172W protein expressed on the rod-dominant background (i.e. WT, rds+/2 or rds2/2) with that onthe cone-dominant background (i.e. Nrl2/2, rds+/2/Nrl2/2 orrds2/2/Nrl2/2).

Western blot (WB) analysis of extracts from retinas containingonly WT RDS (WT/Nrl2/2) or mutant RDS (R172W/rds2/2,R172W/rds2/2/Nrl2/2) under reducing conditions demon-strated that the R172W protein had the correct size in both therod- and cone-dominant background (Fig. 1A). Our previousstudies showed that animals homozygous for the R172W trans-gene had R172W protein levels that were �75% of WT levels(24). Here, we observed that R172W levels in R172W homozy-gous mice on the rds2/2/Nrl2/2 genetic background were�50% of RDS levels in Nrl2/2 retinas (Fig. 1B, top, ∗∗P ,0.01). ROM-1 levels also were decreased in the R172W/rds2/2

retinas compared with WT and R172W/rds2/2/Nrl2/2 versusNrl2/2 retinas (Fig. 1B, bottom), although the difference inROM-1 levels was of smaller magnitude than the differencein RDS levels. Interestingly, these results also confirm an observa-tion that we made previously, namely that the ratio ofRDS:ROM-1 is different in rods (WT background) than incones (Nrl2/2 background) (27). RDS levels in the Nrl2/2

were decreased by �30% compared with WT levels, whileROM-1 levels in the Nrl2/2 line were decreased by �70% com-pared with WT levels (Fig. 1B).

We next assessed the localization of the R172W protein (here-after referred to as just R172W). Frozen retinal sections werecollected from R172W/Nrl2/2 or control Nrl2/2 eyes harvestedat P30 and immunolabeled with antibodies which are eitherspecific to transgenic R172W (mAB 3B6, green Fig. 1C) or

Human Molecular Genetics, 2014, Vol. 23, No. 12 3103

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

which recognize both endogenous and transgenic RDS isoforms(RDS-CT, red, Fig. 1C). Consistent with the specificity of theIRBP promoter, the expression of R172W was restricted tophotoreceptor cells and the mutant protein co-localized withWT RDS. To determine whether the transgene was expressedin either or both S- and M-cones, immuno-florescence (IF) label-ing was performed on tissue sections from Nrl2/2, R172W/

rds+/2/Nrl2/2 and R172W/rds2/2/Nrl2/2 eyes using mAB3B6 (R172W-specific; green, Fig. 1D) and either S- or M-coneopsin-specific antibodies (red, Fig. 1D). Co-localization withmAB 3B6 and S-/M-opsin antibodies confirmed that R172Wwas expressed in both cone cell types. While the imagesshown were taken of photoreceptors whose OSs were insidethe photoreceptor rosettes typical of the Nrl2/2 retina [10],

Figure 1. R172W protein is expressed and properly localized on the Nrl2/2 background. (A) Retinal extracts were harvested at P30 from the indicated genotypes.Shown are representative WBs probed with RDS-CT, ROM1-CT and actin antibodies (loading control) from reducing SDS-PAGE. (B) Quantification of WB data.Protein levels measured densitometrically and normalized to actin then to the mean WT value on each blot. N ¼ 6 retinas per genotype, shown are means+SEM.∗∗P , 0.01, ∗∗∗P , 0.001 by one-way ANOVA with Bonferroni’s post hoc comparison. (C and D) IF was performed on P30 retinal sections using mAB 3B6 (green, Cand D, for R172W RDS), and either RDS-CT (red, C, R172W and WT RDS), S-opsin (red, D, left) or M-opsin (red, D, right). Sections were counterstained with DAPI(blue). Images are single planes from a confocal stack. N ¼ 3–4 eyes per genotype. OS, outer segment; ONL, outer nuclear layer; INL, inner nuclear layer; R, rosette.Scale bars: 25 mm (C), and 10 mm (D).

3104 Human Molecular Genetics, 2014, Vol. 23, No. 12

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

similar results were observed in photoreceptors whose OSs facedthe RPE (Supplementary Material, Fig. S1). RDS functions as acomplex with ROM-1; hence, to confirm that ROM-1 localiza-tion was not altered in the presence of R172W, retinal sectionsfrom Nrl2/2 and R172W/rds2/2/Nrl2/2 were probed by IFusing both RDS-CT and ROM1-2H5 antibodies (Fig. 2A). Weobserve co-immunolocalization of the two proteins and no dif-ferences in their distribution between the two genotypes.Finally, to confirm that RDS and ROM-1 were trafficking tothe OS and not accumulating abnormally in the inner segmentsof photoreceptors, tissue sections were probed with antibodiesto an inner segment marker (Na+K+-ATPase, green, Fig. 2B)and either RDS-CT or ROM1-CT (red, Fig. 2B). We observedthat the green IF labeling was in a distinct layer (correspondingto photoreceptor inner segments) from the red IF labelingobtained with the anti-RDS/ROM-1 antibodies (confined toOSs) in both the Nrl2/2 and R172W/rds2/2/Nrl2/2 mouselines. Hence, both R172W and ROM-1 properly trafficked tothe OS on the given genetic backgrounds. These data are in con-trast to prior observations regarding some other cone-dominantRDS mutations, which cause mislocalization of mutant RDSprotein and cone opsins throughout the outer nuclear and synap-tic (outer plexiform) layers (9,28). In the present study, all fourproteins (R172W, ROM-1, S-opsin and M-opsin) were exclu-sively localized to the OS (Fig. 1D, Fig. 2B).

S- and M-cone functions is similarly affectedby the R172W mutation

Previously, we observed that R172W caused a severe dominantdefect in cone ERG amplitudes, even when the transgene wasexpressed in the presence of WT level of RDS (24). In contrast,rod function was only slightly decreased (in R172W/WT miceversus WT) or was improved (in R172W/rds+/2 versus rds+/2,as shown in (24) and Supplementary Material, Figure S2). Herewe asked whether this dominant cone defect was specific to a par-ticular cone subtype. Full-field spectral photopic ERG was con-ducted at P30 on R172W or non-transgenic (Non-T) mice oneither the rod-dominant (Fig. 3A) or cone-dominant (Fig. 3B)background. S- (Photopic B-UV) and M- (Photopic B-Green)cone function, as measured by maximum photopic b-wave ampli-tudes, were significantly reduced in R172W/WT and R172W/Nrl2/2 compared with their non-transgenic counterparts (WT/Nrl2/2, respectively, Fig. 3A and B), confirming that the cone-dominant defect associated with the R172W mutation is recapitu-lated in the Nrl2/2 model. In the rod-dominant background,S- and M-cones were similarly affected: green photopic b-waveamplitudes were reduced by 59%, while UV-photopic b-waveamplitudes were reduced by 57% (R172W/WT versus WT,Fig. 3A). In the Nrl2/2 cone-dominant background, M-cone pho-topic b-wave amplitudes were slightly more affected than S-coneb-wave amplitudes (65 and 47% decrease between Nrl2/2 andR172W/Nrl2/2, respectively; Fig. 3B).

Interestingly, when the R172W transgene was expressed inthe presence of only one allele of WT RDS (i.e. the rds+/2 back-ground), there was a dramatic decrease in S- and M- cone b-waveamplitudes on the rod-dominant background (R172W/rds+/2

versus rds+/2, Fig. 3A), while amplitudes were unchanged onthe cone-dominant background (R172W/rds+/2/Nrl2/2

versus rds+/2/Nrl2/2, Fig. 3B). This observation highlights

the fact that cones in the retina of the Nrl2/2 mouse line(which is devoid of rods) may not behave in precisely the sameway as WT mouse cones (which are surrounded by rods),although they are quite similar in structure and function.Finally, we examined cone function in cone-dominant micelacking any WT RDS (R172W/rds2/2/Nrl2/2). WhileM-cone b-wave amplitudes were not improved compared withnon-transgenic controls (rds2/2Nrl2/2), mean S-cone b-waveamplitudes tended to be higher than in WT controls (althoughthe difference was not statistically significant, Fig. 3B). This

Figure 2. R172W and ROM-1 properly traffic to the OS. (A) Frozen retinal sec-tions from transgenic and non-transgenic animals were immunolabeled withROM1-2H5 (green) and RDS-CT (red) antibodies. (B). Sections were labeledwith the inner segment marker Na+K+ ATPase (ATPase, green) and eitherRDS-CT or ROM1-CT (red). N ¼ 3 eyes per genotype. Abbreviations as inFigure 1. Scale bar: 10 mm.

Human Molecular Genetics, 2014, Vol. 23, No. 12 3105

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

small improvement in S-cone b-wave amplitudes is consistentwith our previously reported small improvement in non-spectral(i.e. white light-stimulated) photopic b-wave amplitudes inR172W mice lacking endogenous RDS (24), and again supportsthe utility of the Nrl2/2 line as a cone-dominant model. Thisobservation also suggests that while R172W is not capable ofsupporting normal cone photoreceptor function, it apparentlyis better than no RDS at all.

Cone OS ultrastructure reflects cone functionin R172W mice

Due to the low number of cones in the rod-dominant WT mouseretina, we previously had not been able to assess the effects ofthe R172W mutation on cone OS ultrastructure. Therefore, inthe present study, we conducted histological analyses at thelight and EM levels on 1-month old R172W mice on the Nrl2/2

background. No gross changes in retinal morphology wereobserved at the light level in R172W mice compared with non-transgenic controls (Fig. 4A), although consistent with our previ-ous observation (10), we did observe fewer rosettes in the rds2/2/Nrl2/2 retina (and in the R172W/rds2/2/Nrl2/2) than in theNrl2/2 retina (Fig. 4A).

At the ultrastructural level, however, defects became morereadily apparent. Cone OSs in the Nrl2/2 line exhibit nicelystacked lamellae; however, while some similar stacks of lamellaeappeared in retinas in the R172W/Nrl2/2 line, many OSs con-tained open, unflattened membranous structures (black arrow,

Fig. 4B; see also additional representative images in Supplemen-tary Material,Fig. S3).Consistent with the lack of detectable func-tional (ERG) difference, the ultrastructure of OSs of R172W/rds+/2/Nrl2/2 mice were indistinguishable from their non-transgenic rds+/2/Nrl2/2 counterparts, both exhibiting largeswirls of membranous lamellae (11) that were not properlyoriented. Finally, as reported previously (10), we observe thatthe OSs of rds2/2/Nrl2/2 retinas completely lack disc rims andlamellae, and instead were just open, somewhat amorphous mem-branous structures. However, consistent with the small improve-ment we observed in the ERG function, we also detected someimprovements in OS ultrastructure in the R172W/rds2/2/Nrl2/2 mouse retina, including nascent lamellae formation andmembrane stacking (white arrow, Fig. 4B). EM immunogold(IG) labelingwithS-andM-opsinantibodies (SupplementaryMa-terial, Fig. S4A and B) did not identify any ultrastructural differ-ences in S- versus M-cones in transgenic versus non-transgenicanimals. Finally, EM IG labeling with RDS-CT antibodies con-firmed that RDS (both mutant and WT isoforms) was located inthese abnormal OS structures (Supplementary Material, Fig. 4C).

R172W causes defects in ROM-1 complex formation

One of the most useful aspects of Nrl2/2 mice is the ability tostudy RDS biochemistry in cones in this model. Our previousdata from the oligomerization-incompetent C150S RDSmutant suggest that RDS/ROM-1 complexes may assemble dif-ferently in rods versus cones (9,28). Therefore, we conducted

Figure 3. R172W causes a dominant defect in cone function. Full-field spectral photopic ERG was performed on P30 R172W transgenic mice or age-matched non-transgenic (Non-T) controls. Shown are maximum photopic b-wave amplitudes from mice in the rod-dominant (A) or cone-dominant (Nrl2/2, B) background. ERGsweremeasured in response to green (left, M-cones)or UV(right, S-cones) light.N ¼ 8–12 mice pergenotype, shown are means+SEM. ∗∗P , 0.01, ∗∗∗P , 0.001byone-way ANOVA with Bonferroni’s post hoc comparison.

3106 Human Molecular Genetics, 2014, Vol. 23, No. 12

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

non-reducing velocity sedimentation analysis using continuous5–20% sucrose gradients on retinal extracts from R172Wmice in either the rod-dominant (R172W/rds2/2) or cone-dominant (R172W/rds2/2/Nrl2/2) background followed by re-ducing SDS-PAGE/WB analysis (Fig. 5). To prevent cysteineshuffling and the formation of post-extraction disulfide bonds,N-ethylmaleimide was included in the protein extractionbuffer for all biochemical experiments. RDS/ROM-1 core com-plexes are tetrameric (primarily found in sedimentation fractions6–9), and once in the OS these complexes further assemble viaintermolecular disulfide bonds into hetero-octamers (fractions4–5) and higher order RDS homo-oligomers (fractions 1–3).Figure 5A and B shows representative RDS/ROM-1 immuno-blots and graphs plotting the percent of total RDS (solid line)or ROM-1 (dashed line) found in each fraction; no significantdifferences were observed with respect to either RDS orROM-1 distribution between transgenic and non-transgenicmice. Consistent with our prior observations (28), retinas fromcone-dominant animals (Nrl2/2 and R172W/rds2/2/Nrl2/2,Fig. 5B) had a greater percentage of their total RDS in higherorder oligomeric complexes (fractions 1–3) than did retinasfrom rod-dominant animals.

RDS complexes are formed via both covalent (disulfide) andnon-covalent bonds, and intermolecular disulfide bonding is re-sponsible for the assembly of octameric and higher order oligo-meric complexes. The relative contribution of covalently boundversus non-covalently bound forms of RDS/ROM-1 to the totalpool of RDS/ROM-1 complexes can be assessed by examiningfractions obtained from non-reducing sucrose gradients that sub-sequently are analyzed by non-reducing SDS-PAGE/WBs.Shown in Figure 6A are representative non-reducing RDSWBs and companion quantification graphs (as in Fig. 5) plottingthe percent of monomer (dashed line, non-covalently boundform) and dimer (solid line, covalently bound form). Most ofthe RDS was found in the dimeric form, but there were no signifi-cant differences between genotypes. In contrast, when these non-reducing blots were probed with anti-ROM-1 antibodies, an

anomalous third band was detected in addition to the monomericand dimeric forms (Fig. 6B). These abnormal, large molecularweight complexes (dotted line, Fig. 6B) were found primarilyin the octameric fractions, and were highly enriched inR172W/rds2/2/Nrl2/2 retinal extracts. These complexes alsowere observed when R172W was expressed in conjunctionwith WT RDS in the cone-dominant background (R172W/rds+/2/Nrl2/2, Supplementary Material, Fig. 5A). Importantly,these data indicate that while RDS complexes assemble properlyin the presence of R172W, the ability of ROM-1 to complex nor-mally in cones expressing R172W is impaired.

As a result of the appearance of this aberrant third type ofROM-1 complex, we wanted to confirm that ROM-1 stillretained the ability to interact with RDS in the presence ofR172W transgenic protein. Therefore, we conducted reciprocalco-immunoprecipitation (IP) with antibodies against RDS andROM-1 (bound fractions are shown in Fig. 7A; input andunbound fractions are in Supplementary Material, Fig. 5B). Al-though RDS and ROM-1 protein levels are lower in R172W/rds2/2/Nrl2/2 and R172W/rds2/2 retinas than in controls,the mutant RDS was able to pull down ROM-1 (and viceversa). We next asked whether the abnormal large molecularweight ROM-1 complexes identified in the velocity sedimenta-tion studies retained the ability to bind R172W. Using non-reducing SDS-PAGE/WB analysis following IP of retinalextracts with anti-RDS antibodies, we confirmed that this aber-rant large complex does bind to RDS (Fig. 7B).

R172W mice exhibit RPE defects and degeneration of thechoriocapillaris consistent with patient phenotypes

Although vision defects in patients carrying the R172W RDSmutation are primarily attributed to defective cones, the maculo-pathies seen clinically suggest that such defects may be a com-bination of direct effects on cones coupled with secondarysequellae, such as toxicity to the RPE, which firsts affects themacula due to the higher demands on the RPE in that region

Figure 4. R172W leads to dominant defects in cone OS ultrastructure. Eyes from the indicated genotypes were collected and fixed at P30 for histology at the light (A)and EM (B) levels. RPE, retinal pigment epithelium; ONL, outer nuclear layer; INL, inner nuclear layer; IS, inner segment; OS, outer segment. Scale bars: 25 mm (A)and 1 mm (B). N ¼ 3 eyes per genotype.

Human Molecular Genetics, 2014, Vol. 23, No. 12 3107

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

Figure 5. R172W does not alter the distribution of RDS complexes. Retinal extracts collected at P30 from transgenic or non-transgenic controls in the rod-dominant(A) or cone-dominant (Nrl2/2, B) background were fractionated on continuous 5–20% non-reducing sucrose gradients. Fractions were separated on reducingSDS-PAGE and blots were probed for RDS or ROM-1. Graphs show the amount of RDS (solid line) or ROM-1 (dashed line) in each fraction as a percent of totalimmunoreactivity. Plotted are means+SEM. Peak fractions for molecular weight markers in the gradient are shown with arrows: apoferritin (443 kD), beta-amylase(200 kD), alcohol dehydrogenase (150 kD), bovineserum albumin (66 kD) and carbonicanhydrase (29 kD). Two retinas werepooled foreach extraction/fractionationand three to six independent experiments were performed per genotype.

Figure 6. R172W leads to the formation of abnormal ROM-1 complexes in the Nrl2/2 background. Retinal extracts from the non-reducing gradients shown in Figure 5were separated on non-reducing SDS-PAGE. Blots were probed with RDS (A) or ROM-1 (B). Graphs show the amount of RDS/ROM-1 found as monomer (dashedline, A and B), dimer (solid line, A and B) or in the case of ROM-1, an abnormal large MW complex (dotted line, B) as a percent of total immunoreactivity. Plotted aremeans+SEM. Peak fractions for molecular weight markers in the gradient are shown with arrows as in Figure 5. Two retinas were pooled for eachextraction/fractionation and three six independent experiments were performed per genotype.

3108 Human Molecular Genetics, 2014, Vol. 23, No. 12

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

compared with other retinal regions. Clinically, this manifests asperipapillary atrophy, RPE atrophy and loss of the choriocapil-laris, in addition to other pronounced phenotypic changes inthe macula (RPE granularity, bull’s eye maculopathy, etc.)(13,15), all of which are visible by fundus imaging and fluores-cein angiography (FA). Because mice lack a macula, they arenot a good model for such characteristic central retinal defects.However, to determine whether R172W transgenic miceexhibit any of the other clinical signs, we conducted fundusimaging and FA at 2 months of age. The Nrl2/2 model isknown to degenerate over time (10) and the rosettes commonlyseen in this model appear on fundus images, potentially cloudinginterpretation of the phenotype, and so these studies wererestricted to mice that had the rod-dominant background.

Fundus images from R172W/rds2/2 were often grosslynormal, although some exhibited an abnormal ring around theoptic nerve (Fig. 8A, left column, yellow arrows), as well as alack of the normal star pattern of retinal vasculature. In contrast,the phenotype was quite striking upon FA; transgenic mice exhib-ited extensive loss of the choriocapillaris (Fig. 8A, right column,only choroidal arterioles are visible, capillary beds are gone). Thisphenotype is nearly identical to that seen in RDS-associatedmaculopathy patients (13,15). In the R172W/rds2/2 eyes,retinal vasculature was difficult to see, but appeared to be severelyattenuated, and also was found in a very different imaging plane(observe faint out-of-focus retinal vessels in some specimens,red arrows Fig. 8A) from the newly exposed, abnormal choroidalvasculature. Interestingly, in mice (as in patients), this FAphenotype exhibited incomplete penetrance: half (4 of 8 eyesexamined) exhibited the severe phenotype shown in the top tworows of Figure 8A, while the other eyes (4 of 8) showed no oronly minor changes (Fig. 8A, bottom row). In addition, the pheno-type was not uniformly bilateral: some mice exhibited this chor-oidal atrophy in both eyes, while in others only one eyeappeared to be affected. None of the age-matched WT animalsexamined (Fig. 8B) showed any fundus or FA abnormalities. Inaddition, these atrophic changes were specific to the R172Wmutation and not merely due to RDS deficiency as they werenot observed in age-matched non-transgenic rds+/2 (Fig. 8C)or rds2/2 eyes (Supplementary Material, Fig. 6).

A subset of R172W/rds2/2 animals were aged out to observethe FA phenotype at later time points (5 months of age). FA andhistological sections from R172W/rds2/2 eyes shown in

Figure 8D are the same eyes imaged in Figure 8A (controlimages in Fig. 8E and F are age matched but not the sameanimals as in Fig. 8B and C). Vascular phenotypes in eyes exhi-biting abnormalities at 2 months of age persisted or worsened at 5months of age: for example, the eye shown in the top row ofFigure 8D showed further degeneration of choroidal vesselsfrom 2 to 5 months of age. In contrast, the eye which showedno FA abnormalities at 2 months of age continued to exhibitnormal vasculature at 5 months of age (Fig. 8D, bottom row).As before, WT and rds+/2 controls exhibited no FA abnormal-ities at 5 months of age. These severe choroidal abnormalitieswere also observed in histological sections. In the WT andrds+/2 eyes, the choroid consists of a continuous plexus withthe small vessels of the choriocapillaris readily visible (Fig. 8Eand F, white arrows in right panels). In contrast, in R172W/rds2/2 eyes, it was often impossible even to detect the chorioca-pillaris and the remaining choroidal tissue itself was largelydegenerated with large open spaces (Fig. 8D, right panels).This phenotype correlated with the FAs; eyes with severe FA ab-normalities exhibited the worst choroidal structure upon histo-logical examination of tissue sections.

Given these observed choroidal abnormalities, we next askedwhether the neural retina exhibited degeneration at 5 months ofage, especially in view of the photoreceptor-specific expressionof the mutant protein. We conducted morphometric assessmentof the thickness of the outer nuclear layer (ONL, a measure ofphotoreceptor degeneration) and found that the R172W/rds2/2

eyes exhibited dramatic thinning of the ONL compared withboth the WT and rds+/2 (Fig. 9A and B). The mean ONL thick-ness in R172W/rds2/2 mice was reduced to �45% of WT thick-ness with similar magnitude losses across the retina (Fig. 9B).Again, this deterioration is not due to deficiency of the RDSprotein, but rather is a specific gain-of-function of the R172W mu-tation, since we have previously shown that mice homozygous forthe transgene express �75% of WT levels of RDS protein (incomparison to �50% in the rds+/2). Interestingly, we did notobserve the same variability in ONL thickness that we observedin FA. All R172W/rds2/2 animals studied showed thinning ofthe ONL throughout the retina. Patients typically exhibit fundussigns of RPE atrophy and so we next examined the RPE structure(Fig. 9C). R172W/rds2/2 exhibited large vacuoles in the RPE(Fig. 9C, black arrows and 8D, white arrowheads). These wereonly observed in the central retina and were mostly pronounced

Figure 7. R172W binds to ROM-1 in cones. (A) Retinal extracts underwent reciprocal co-IP with RDS-CT (top) or ROM1-CT (bottom). Shown are reducingSDS-PAGE/WBs on the bound fraction (input and unbound fractions are found in Supplementary Material, Fig. S4) probed with RDS-CT or ROM1-CT. (B) Extractsunderwent IP with RDS-CT and non-reducing SDS-PAGE/WBs are shown (probed with RDS-CT or ROM-1 mAB 2H5). I, input; B, bound; U, unbound. Two retinaswere pooled for each IP and three independent experiments were performed per genotype.

Human Molecular Genetics, 2014, Vol. 23, No. 12 3109

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

in the peripapillary region near the optic nerve. Interestingly, incontrast to the ONL thinning, this RPE defect was only seen inanimals that exhibited FA abnormalities.

DISCUSSION

Here we present exciting new evidence that sheds significantlight on the mechanisms of RDS-associated cone-dominantdisease. We showed that the cone-dominant functional effectsof the R172W are recapitulated in the Nrl2/2 background, sup-porting its use for analysis of the effects of R172W on cone

photoreceptors. The effects of R172W impact both S- andM-cones, i.e. they are not cone subtype specific; consistent withthe fact that most murine cones express both M- and S-opsin. Im-portantly,wefound that the expressionofR172Wincones leads toabnormal RDS/ROM-1 complex formation, specifically changesin the biophysical behavior of ROM-1. This abnormality also wasdetected when R172W was co-expressed with WT RDS, consist-ent with thedominant natureof the defect inmaculopathypatients.Furthermore, we observed striking clinical signs of RPE defectsand degeneration of the choriocapillaris, consistent with compar-able findings in human RDS-associated disease.

Figure 8. R172W causes RPE atrophy and degeneration of the choriocapillaris. R172W mice (A) or age-matched non-transgenic controls (WT: B, rds+/2: C) under-went fundus imaging (left column) and FA (right column) at 2 months of age. (A) Images from three different R172W/rds2/2 eyes to illustrate the varied phenotype inthis background. Images in the left column are from the same eye as those in the right column. Yellow arrows in (A) show fundus abnormalities and red arrows showshadows of retinal vasculature in a different imaging plane. (D–F) Shown are fluorescein angiograms (right) and histological sections (left) from the indicated geno-types at 5 months of age. White arrows point to small vessels of the choriocapillaris and white arrowheads show RPE vacuoles. N ¼ 6/8 eyes per genotype. Scale bar,10 mm. Sc, sclera; Ch, choroid; RPE, retinal pigment epithelium; ChC, choriocapillaris.

3110 Human Molecular Genetics, 2014, Vol. 23, No. 12

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

Defects in RDS complex formation may contribute directly orindirectly to many different RDS-associated retinal disease phe-notypes, a fact which undoubtedly accounts for the observationthat the majority of RDS disease-causing mutations, includingR172W, reside in the D2 loop (the region of RDS/RDS andRDS/ROM-1 interactions) (4,5). The defect in complex forma-tion we observed here was relatively minor. The distribution ofRDS complexes is completely normal in R172W retinas, bothin the rod- and cone-dominant background. The only changewe observed was the appearance of abnormal, large molecularweight ROM-1 complexes in the cones of R172W transgenicretina. These complexes are not likely to be misfolded aggre-gates awaiting degradation in the inner segment (where proteinis synthesized), for three reasons. First, we did not detect anyROM-1 in the inner segment, suggesting that the protein is traf-ficked out of the ER/Golgi to the OS. Secondly, the abnormalcomplexes were reduced to monomers in the presence of redu-cing agent. In the case of RDS mutants that aggregate and aredegraded, such as C214S (21,29), we have observed that thelarge aggregate band seen on immunoblots persisted even inthe presence of reducing agent (29). Thirdly, the abnormallarge-molecular-weight ROM-1 complex was found in gradientfractions associated with octamers. Again, RDS mutant isoformsthat remain in the ER and aggregate (forming higher order oligo-mers) tend to accumulate in the heaviest fractions of the gradient(29), rather than in the intermediate fractions.

The origin of these large-molecular-weight ROM-1 complexesin R172W-expressing cones is not clear. However, the fact that

they are seen in the presence of WT RDS (R172W/rds+/2/Nrl2/2) suggests that they may be clinically relevant, sinceaffected patients express one copy of mutant and one copy ofWT RDS. Periodically, we have seen a small amount of the abnor-mal complexes in R172W transgenic retinas in the R172W/rds2/2 background. Although this could represent complexescoming fromthe small number of cones in the rod-dominant back-ground, the fact that we also sometimes detect a small quantity ofthese complexes in the Nrl2/2 retina makes that unlikely to be thesole factor. What is more likely is that these complexes areforming in response to subtle stresses in the photoreceptor. Wehave previously shown that rod OSs are more resistant to the pres-ence of mutant RDS than are cones, requiring only sufficient RDSprotein to prevent haploinsufficiency phenotype (21,23,25). Thereason for this differential photoreceptor subtype sensitivity isunknown, but may be tied to the different structural organizationof cone OSs (which have lamellae/rims exposed to the extracellu-lar space) and rods (in which discs are fully enclosed by the OSplasma membrane). It also may be tied to differences incomplex assembly in the two cell types. Our data from mice carry-ing the C150S RDS mutation suggest that RDS complexes mayassemble differently in the inner segments of rods versus cones(although the nature of these differences is not known) (9,28).In any case, when mutant R172W is expressed in cone OSs, theRDS complexes become just sufficiently abnormal to causealterations in ROM-1.

However, the reason that R172W (and possibly other RDSmutations) causes MD is likely more complex than a simple

Figure 9. R172W/rds2/2 eyes exhibit pan-retinal degeneration and RPE defects. (A) Shown are representative 40× micrographs captured from the indicated regionfrom eyes harvested at 5 months of age. (B) Shown is the quantification of the thickness of the ONL from sections as in (A). N ¼ 4–5 eyes/group. ∗P , 0.05, ∗∗∗ P ,

0.001. (C) Shown are higher magnification sections of the RPE in the region near the optic nerve head (ONH). Observe vacuoles in the RPE. Scale bar, 10 mm. Ch,choroid; RPE, retinal pigment epithelium; OS, outer segments; IS, inner segments; ONL, outer nuclear layer; INL, inner nuclear layer; GCL, ganglion cell layer.

Human Molecular Genetics, 2014, Vol. 23, No. 12 3111

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

molecular defect in cones. A key phenotype of R172W-associateddisease in patients, often observable before overt vision loss orERG changes are detectable, is maculopathy and the developmentof atrophic patches in the RPE, which are visible by fundus exam-ination (13). This phenotype is not uniformly associated withdefects in cones. For example, mutations in the cone phototrans-duction ion channel protein CNGA3 cause a complete lack ofcone function and color vision from birth (30), but except inrare cases in older individuals, do not cause alterations in fundusappearance or RPE (31). As a result, we hypothesize that thesevere MD coupled with choroidal/RPE atrophy seen in patientscarrying the R172W mutation is due to a combination ofprimary cone photoreceptor molecular defects coupled with sec-ondary sequellae that impact RPE as well as choriocapillaris via-bility and structural integrity. Precedence for this hypothesisexists. For example, mutations in the photoreceptor geneABCA4 lead to accumulation of abnormal retinoids (derivedfrom the visual cycle), which are toxic to the RPE, resulting inStargardt’s MD (32,33). Importantly, we showed in the presentstudy that the transgenic R172W RDS mouse line recapitulatesthe RPE/choriocapillaris defects so prevalent in maculopathypatients, even though the mouse lacks a macula. The fact thatthis phenotype is not observed in rds+/2 mice indicates that it isspecific to the R172W mutation, and also is not merely causedby ongoing degeneration, OS loss or RDS-associated defects invision. Interestingly, in common with the case in patients, wesee striking animal-to-animal variability in fluorescein angiogramand RPE phenotypes in R172W/rds2/2, while degeneration ofphotoreceptors is fairly consistent among all animals. This sug-gests that the clinical disease severity is determined less by theprimary defect in photoreceptors and more by the varyingability of neighboring tissues (RPE and choroid) to cope withsuch a defect. Furthermore, we observe significant pan-retinal de-generation of the choriocapillaris in animals who exhibit onlymodest, centrally localized defects in RPE cells and no sign ofovert loss of RPE cells. That death of the RPE is not a prerequisitefor degeneration of the neighboring choroid suggests that moresubtle toxic changes in the RPE may influence the adjacent vascu-lature, and in-depth assessment of the RPE will be part of futurework.

In conclusion, we have provided new findings that may be in-formative with regard to the mechanisms underlying MD in thecase of the R172W mutation in RDS. We hope to capitalize onthese new insights to develop rationally designed, mechanism-based therapeutics that will target disease processes ongoingboth in photoreceptors as well as other affected cell types. Thisexcellent mouse model will facilitate future work to moreclosely evaluate the interplay between photoreceptor defectsand resulting loss in RPE structural and functional integrity.Of particular importance will be an assessment of the role ofthe abnormal RDS/ROM-1 complexes we have described herein the development of the associated RPE and choriocapillarisdefects.

MATERIALS AND METHODS

Animal care and use

R172W transgenic mice were generated and characterized asdescribed previously (24). Transgene expression is driven by

the photoreceptor-specific interphotoreceptor retinoid-bindingprotein promoter, and the murine RDS cDNA carries the patho-genic R172W point mutation. It also carries a P341Q modifica-tion to facilitate antibody recognition (see below), which wehave previously shown has no effect on protein function orgross biochemical properties (25). R172W mice were bredonto either an Nrl2/2 background (generously shared by DrAnand Swaroop, National Eye Institute), or a rds2/2/Nrl2/2

background, as described and characterized previously (10).Animals were maintained under dim cyclic light (12 h/12 hL:D cycle; �30 lux). All experimental protocols and procedureswere approved by the Institutional Animal Care and Use Com-mittee at the University of Oklahoma Health Sciences Center,and conformed to the Statement for the Use of Animals in Oph-thalmic and Vision Research as set forth by the Association forResearch in Vision and Ophthalmology and the NIH Guide forthe Care and Use of Laboratory Animals.

Antibodies

RDS-CT and ROM1-CT rabbit polyclonal antibodies against theC-terminus of RDS were generated in-house and characterized aspreviously described (9,10,24). RDS-CT recognizes both en-dogenous RDS and the R172W mutant RDS isoform, and wasused at a dilution of 1:1000 for WB and immuno-fluorescence(IF) microscopy or at 1:10 for EM IG microscopy. ROM1-CTwas used at 1:1000 dilution (WB and IF). ROM-1 monoclonalantibody (mAB) 2H5 also was generated in-house and used at1:10 dilution for WB. mAB 3B6 (generously provided by DrRobert Molday, University of British Columbia) specificallyrecognizes the transgenic (R172W) RDS isoform, but not WTmurine RDS, due to the P341Q silent mutation and was used at1:100 dilution for IF. Rabbit polyclonal antibodies against short(S-) and medium (M-) wavelength cone opsins (generouslyshared by Dr Cheryl Craft, University of Southern California)were used at 1:10 dilution for IG or at 1:30 000 dilution for IF.HRP-conjugated anti- actin polyclonal antibodies (Sigma-Aldrich, St. Louis, MO, USA) were used at 1:50 000 dilution.The following secondary antibodies were employed: anti-rabbit/anti-mouse HRP-conjugated IgG (KPL, Gaithersburg, MD,USA, at 1:50 000 dilution for WB); Alexafluor 488-, 555- or647-conjugated anti-mouse/anti-rabbit IgG (Life Technologies,Grand Island, NY, USA, at 1:1000 dilution for IF); andAuroProbe

TM

10 nm colloidal gold-conjugated anti-rabbit IgG(GE Healthcare, Pittsburgh,PA, USA,at 1:10 dilution for EMIG).

IF microscopy

Eyes were enucleated, fixed and dissected as described previous-ly (21,28). After cryoprotection in serial sucrose dilutions (28),frozen retinal tissue sections (10-mm thickness) were collectedonto glass microscope slides and IF was performed as describedin detail previously (28). In brief, sections were incubated in 1%NaBH4, washed in PBS, blocked in PBS containing 5% (w/v)BSA (Sigma-Aldrich), 1% (w/v) fish gelatin (Sigma-Aldrich),20% (v/v) donkey serum (Jackson ImmunoResearch, WestGrove, PA, USA) and 1% (v/v) Triton X-100 (VWR). Sectionswere then incubated overnight with primary antibodies dilutedin blocking buffer, then rinsed, incubated with secondary anti-bodies, rinsed again and mounted using ProlongGold

TM

3112 Human Molecular Genetics, 2014, Vol. 23, No. 12

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

mounting medium containing 4′,6-diamidino-2-phenylindole(DAPI; Life Technologies). Images were captured on a BX-62spinning disk confocal microscope (Olympus, Japan) and ana-lyzed with Slidebook

TM

4.2 software (Intelligent Imaging Inno-vations, Denver, CO, USA). All images are presented as singleplanes from a confocal stack unless otherwise specified. Expos-ure times and display settings for all images were normalized toa control section where primary antibody was omitted duringprocessing.

WB/IP/velocity sedimentation

For protein chemistry, retinas were added to solubilizationbuffer [PBS, pH 7.0, containing 1% (v/v) TX-100, 5 mMEDTA, 5 mg/ml N-ethyl maleimide and protease inhibitors],briefly sonicated, then incubated at 48C for 1 h. After pelletinginsoluble material in a microfuge, soluble protein content inextracts was quantified using Bradford reagent (Bio-Rad, Teme-cula, CA, USA) as per the manufacturer’s directions. IP (100 mgretinal protein extract per sample) and reducing/non-reducingSDS-PAGE/WB analyses (15 mg retinal protein extract perlane) were performed as described previously (9,24). Non-reducing velocity sedimentation was performed on wholeretinal extracts using continuous 5–20% (w/v) sucrose gradientsas described previously (7,9,28): 100 mg protein per gradientwas loaded for WT, R172W/rds2/2 and Nrl2/2 retinal extracts,while 200 mg protein per gradient was used for R172W/rds2/2/Nrl2/2 retinal extracts (due to the lower RDS levels).

Histology and electron microscopy

Tissue collection, dissection, fixation and plastic embedment forlight microscopy (histology), conventional transmission electronmicroscopy (EM) and EM IG cytochemistry were as describedpreviously (10,21). Thin sections (600–800 A) were collectedon copper (EM) or nickel (IG) grids and stained prior to beingimaged on a JEOL 100CX electron microscope (9). Morphomet-ric measurements were made on 40× micrographs using AdobePhotoshop CS5. Three measurements were made in each imageand 4–5 eyes/genotype were analyzed. Differences betweengenotypes and location (peripheral/central) were assessed viatwo-way ANOVA with Bonferroni’s post hoc comparisons.

Electroretinography

Full-field scotopic and spectral photopic electroretinography(ERG) was performed at postnatal day (P) 30 as described previ-ously (28) using a BigShotw Ganzfeld and UTAS system (LKC,Gaithersburg, MD, USA). Briefly, the eyes of dark-adapted,anesthetized animals were dilated (1% Cyclogylw; Pharmaceut-ical Systems, Inc., Tulsa, OK, USA), and scotopic measurementswere recorded in response to a single strobe flash at 157 cd s/m2.For photopic recordings, animals were light adapted for 5 min(29.03 cd/m2), then were exposed to 25 flashes at 530 nm(M-cones, 12.5 cd s/m2). Finally, after a 1 min recoveryperiod, they were exposed to 25 flashes at 365 nm (S-cones,0.79 cd s/m2). Differences between groups were analyzed byone-way ANOVA with Bonferroni’s post hoc comparisons(http://www3.nd.edu/~rwilliam/stats1/x53.pdf).

Fundus imaging

Fundus imaging was performed on anesthetized, dilated miceusing a Micron III system (Phoenix Research Laboratories,Pleasanton, CA, USA) as described previously (34,35). Aftercollecting bright-field fundus images, animals were injectedintraperitoneally with 100 ml of 1% (w/v) fluorescein sodium(Sigma-Aldrich)andFAimageswerecapturedusinganexcitationfilter of 486 nm and an emission filter of 436 nm. All images werecaptured using StreamPixw software (Phoenix Research Labs).

SUPPLEMENTARY MATERIAL

Supplementary Material is available at HMG online.

ACKNOWLEDGEMENTS

The authors would like to thank Ms Maggie Yoder and Ms BarbNagel for their technical assistance with fundus images and EM,respectively. The contents described herein do not reflect theviews of the Veterans Administration or the US Government;no official endorsement by the Veterans Administration or theUS Government of the findings reported herein is either madeor implied.

Conflicts of Interest statement. None of the authors has any con-flict of interest to declare.

FUNDING

This work was supported by grants from the National Eye Institute(EY010609 to M.I.N., EY018512 to S.M.C. and EY007361 toS.J.F.), the Foundation Fighting Blindness (M.I.N.) and the Okla-homa Center for the Advancement of Science and Technology(S.M.C. and M.I.N.), an Unrestricted Grant from Research toPreventBlindness (S.J.F.) and by resources and facilities providedby the Veterans Administration Western New York HealthcareSystem, Department of Veteran Affairs, Veterans Health Admin-istration, Office of Research and Development (S.J.F.).

REFERENCES

1. Arikawa, K., Molday, L.L., Molday, R.S. and Williams, D.S. (1992)Localization of peripherin/rds in the disk membranes of cone and rodphotoreceptors: relationship to disk membrane morphogenesis and retinaldegeneration. J. Cell Biol., 116, 659–667.

2. Sanyal, S., De Ruiter, A. and Hawkins, R.K. (1980) Development anddegeneration of retina in rds mutant mice: light microscopy. J. Comp.Neurol., 194, 193–207.

3. Sanyal, S. and Jansen, H.G. (1981) Absence of receptor outer segments in theretina of rds mutant mice. Neurosci. Lett., 21, 23–26.

4. Ding, X.Q., Stricker, H.M. and Naash, M.I. (2005) Role of the secondintradiscal loop of peripherin/rds in homo and hetero associations.Biochemistry, 44, 4897–4904.

5. Goldberg, A.F., Fales, L.M., Hurley, J.B. and Khattree, N. (2001) Foldingand subunit assembly of photoreceptor peripherin/rds is mediated bydeterminants within the extracellular/intradiskal EC2 domain: implicationsfor heterogeneous molecular pathologies. J. Biol. Chem., 276, 42700–42706.

6. Loewen, C.J. and Molday, R.S. (2000) Disulfide-mediated oligomerizationof Peripherin/Rds and Rom-1 in photoreceptor disk membranes.Implications for photoreceptor outer segment morphogenesis anddegeneration. J. Biol. Chem., 275, 5370–5378.

Human Molecular Genetics, 2014, Vol. 23, No. 12 3113

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018

7. Chakraborty, D., Ding, X.Q., Fliesler, S.J. and Naash, M.I. (2008) Outersegment oligomerization of Rds: evidence from mouse models andsubcellular fractionation. Biochemistry, 47, 1144–1156.

8. Goldberg, A.F., Loewen, C.J. and Molday, R.S. (1998) Cysteine residues ofphotoreceptor peripherin/rds: role in subunit assembly and autosomaldominant retinitis pigmentosa. Biochemistry, 37, 680–685.

9. Chakraborty, D., Ding, X.Q., Conley, S.M., Fliesler, S.J. and Naash, M.I.(2009) Differential requirements for retinal degeneration slowintermolecular disulfide-linked oligomerization in rods versus cones. Hum.Mol. Genet., 18, 797–808.

10. Farjo, R., Skaggs, J.S., Nagel, B.A., Quiambao, A.B., Nash, Z.A., Fliesler,S.J. and Naash, M.I. (2006) Retention of function without normal discmorphogenesis occurs in cone but not rod photoreceptors. J. Cell Biol., 173,59–68.

11. Farjo, R., Fliesler, S.J. and Naash, M.I. (2007) Effect of Rds abundance oncone outer segment morphogenesis, photoreceptor gene expression, andouter limiting membrane integrity. J. Comp. Neurol., 504, 619–630.

12. Jansen, H.G. and Sanyal, S. (1984) Development and degeneration of retinain rds mutant mice: electron microscopy. J. Comp. Neurol., 224, 71–84.

13. Downes, S.M., Fitzke, F.W., Holder, G.E., Payne, A.M., Bessant, D.A.,Bhattacharya, S.S. and Bird, A.C. (1999) Clinical features of codon 172 RDSmacular dystrophy: similar phenotype in 12 families. Arch. Ophthalmol.,117, 1373–1383.

14. Wickham, L., Chen, F.K., Lewis, G.P., Uppal, G.S., Neveu, M.M., Wright,G.A., Robson, A.G., Webster, A.R., Grierson, I., Hiscott, P. et al. (2009)Clinicopathological case series of four patients with inherited maculardisease. Invest. Ophthalmol. Vis. Sci., 50, 3553–3561.

15. Wroblewski, J.J., Wells, J.A. III, Eckstein, A., Fitzke, F., Jubb, C., Keen, T.J.,Inglehearn, C., Bhattacharya, S., Arden, G.B., Jay, M. et al. (1994) Maculardystrophy associated with mutations at codon 172 in the human retinaldegeneration slow gene. Ophthalmology, 101, 12–22.

16. Michaelides, M., Holder, G.E., Bradshaw, K., Hunt, D.M. and Moore, A.T.(2005) Cone-rod dystrophy, intrafamilial variability, and incompletepenetrance associated with the R172W mutation in the peripherin/RDSgene. Ophthalmology, 112, 1592–1598.

17. Wells, J., Wroblewski, J., Keen, J., Inglehearn, C., Jubb, C.,Eckstein,A., Jay,M., Arden, G., Bhattacharya, S., Fitzke, F. et al. (1993) Mutations in thehuman retinal degeneration slow (RDS) gene can cause either retinitispigmentosa or macular dystrophy. Nat. Genet., 3, 213–218.

18. Weleber, R.G., Carr, R.E., Murphey, W.H., Sheffield, V.C. and Stone, E.M.(1993) Phenotypic variation including retinitis pigmentosa, patterndystrophy, and fundus flavimaculatus in a single family with a deletion ofcodon 153 or 154 of the peripherin/RDS gene. Arch. Ophthalmol., 111,1531–1542.

19. Nakazawa, M., Kikawa, E., Kamio, K., Chida, Y., Shiono, T. and Tamai, M.(1994) Ocular findings in patients with autosomal dominant retinitispigmentosa and transversion mutation in codon 244 (Asn244Lys) of theperipherin/RDS gene. Arch. Ophthalmol., 112, 1567–1573.

20. Nakazawa, M., Kikawa, E., Chida, Y. and Tamai, M. (1994) Asn244Hismutation of the peripherin/RDS gene causing autosomal dominant cone-roddegeneration. Hum. Mol. Genet., 3, 1195–1196.

21. Stricker, H.M., Ding, X.Q., Quiambao, A., Fliesler, S.J. and Naash, M.I.(2005) The Cys214--.Ser mutation in peripherin/rds causes aloss-of-function phenotype in transgenic mice. Biochem. J., 388, 605–613.

22. Cheng, T., Peachey, N.S., Li, S., Goto, Y., Cao, Y. and Naash, M.I. (1997)The effect of peripherin/rds haploinsufficiency on rod and conephotoreceptors. J. Neurosci., 17, 8118–8128.

23. Conley, S., Nour, M., Fliesler, S.J. and Naash, M.I. (2007) Late-onset conephotoreceptor degeneration induced by R172W mutation in Rds and partialrescue by gene supplementation. Invest. Ophthalmol. Vis. Sci., 48, 5397–5407.

24. Ding, X.Q., Nour, M., Ritter, L.M., Goldberg, A.F., Fliesler, S.J. and Naash,M.I. (2004) The R172W mutation in peripherin/rds causes a cone-roddystrophy in transgenic mice. Hum. Mol. Genet., 13, 2075–2087.

25. Nour, M., Ding, X.Q., Stricker, H., Fliesler, S.J. and Naash, M.I. (2004)Modulating expression of peripherin/rds in transgenic mice: critical levelsand the effect of overexpression. Invest. Ophthalmol. Vis. Sci., 45, 2514–2521.

26. Mears, A.J., Kondo, M., Swain, P.K., Takada, Y., Bush, R.A., Saunders,T.L., Sieving, P.A. and Swaroop, A. (2001) Nrl is required for rodphotoreceptor development. Nat. Genet., 29, 447–452.

27. Conley, S.M., Stuck, M.W. and Naash, M.I. (2012) Structural and functionalrelationships between photoreceptor tetraspanins and other superfamilymembers. Cell. Mol. Life Sci., 69, 1035–1047.

28. Chakraborty, D., Conley, S.M., Stuck, M.W. and Naash, M.I. (2010)

Differences in RDS trafficking, assembly and function in cones versus rods:insights from studies of C150S-RDS. Hum. Mol. Genet., 19, 4799–4812.

29. Conley, S.M., Stricker, H.M. and Naash, M.I. (2010) Biochemical analysisof phenotypic diversity associated with mutations in codon 244 of the retinal

degeneration slow gene. Biochemistry, 49, 905–911.

30. Wissinger, B., Gamer, D., Jagle, H., Giorda, R., Marx, T., Mayer, S.,Tippmann, S., Broghammer, M., Jurklies, B., Rosenberg, T. et al. (2001)

CNGA3 mutations in hereditary cone photoreceptor disorders. Am. J. Hum.

Genet., 69, 722–737.

31. Kohl, S., Jagle, H., Sharpe, L.T. and Wissinger, B. (1993) Achromatopsia. InPagon, R.A., Adam, M.P., Bird, T.D., Dolan, C.R., Fong, C.T. and Stephens,

K. (eds), GeneReviews. University of Washington, Seattle.

32. Weng, J., Mata, N.L., Azarian, S.M., Tzekov, R.T., Birch, D.G. and Travis,G.H. (1999) Insights into the function of Rim protein in photoreceptors andetiology of Stargardt’s disease from the phenotype in abcr knockout mice.

Cell, 98, 13–23.

33. Strauss,O. (2005) The retinal pigmentepitheliumin visual function.Physiol.

Rev., 85, 845–881.

34. Koirala, A., Makkia, R.S., Conley, S.M., Cooper, M.J. and Naash, M.I.(2013) S/MAR-containing DNA nanoparticles promote persistent RPE geneexpression and improvement in RPE65-associated LCA. Hum. Mol. Genet.,22, 1632–1642.

35. Han, Z., Guo, J., Conley, S.M. and Naash, M.I. (2013) Retinal angiogenesisin the Ins2Akita mouse model of diabetic retinopathy. Invest. Ophthalmol.

Vis. Sci., 54, 574–584.

3114 Human Molecular Genetics, 2014, Vol. 23, No. 12

Downloaded from https://academic.oup.com/hmg/article-abstract/23/12/3102/695898by gueston 09 April 2018