bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Ift172 conditional knockout mice exhibit rapid retinal degeneration and

trafficking defects

Priya R Gupta1,2, Nachiket Pendse1, Scott H Greenwald1, Mihoko Leon1, Qin

Liu1, Eric A Pierce1, Kinga M Bujakowska1

1Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of

Ophthalmology, Harvard Medical School, Boston, Massachusetts, 02114, USA

2Weill Cornell Medical College, New York, New York, 10021, USA

Corresponding author:

Kinga M Bujakowska:

Massachusetts Eye and Ear, 243 Charles Street, Boston, MA 02114

[email protected], Tel.: (617)-391-5933, Fax: (617)-573-

6901

1 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Abstract

Intraflagellar transport (IFT) is a bidirectional transport process that occurs along

primary cilia and specialized sensory cilia, such as photoreceptor outer-

segments. coding for various IFT components are associated with

ciliopathies. Mutations in IFT172 lead to diseases ranging from isolated retinal

degeneration to severe syndromic ciliopathies. In this study, we created a mouse

model of IFT172-associated retinal degeneration to investigate the ocular

disease mechanism. We found that depletion of IFT172 in rod photoreceptors

leads to a rapid degeneration of the , with severely reduced

electroretinography responses by one month and complete outer-nuclear layer

degeneration by two months. We investigated molecular mechanisms of

degeneration and show that IFT172 protein reduction leads to mislocalization of

specific photoreceptor outer-segments (RHO, RP1, IFT139), aberrant

light-driven translocation of alpha transducin and altered localization of glioma-

associated oncogene family member 1 (GLI1). This murine model recapitulates

the retinal phenotype seen in patients with IFT172-associated blindness and can

be used for in vivo testing of ciliopathy therapies.

2 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

INTRODUCTION

Primary cilia are non-motile -based organelles that are present in the

majority of vertebrate cell types. They are involved in cellular signaling that is

crucial for the development and functioning of most organs (1). Mutations in

genes coding for ciliary proteins lead to ciliopathies, rare genetic disorders that

may affect the retina, brain, olfactory epithelium, heart, liver, kidney, bone,

gonads and adipose tissues, in isolation or as part of specific syndromes (2–4).

Vision loss is a common feature of ciliopathies because the photoreceptor outer-

segment (OS) is a highly specialized sensory that is critical for

phototransduction (5–7).

The assembly and maintenance of the cilium requires

(IFT), the bidirectional protein trafficking that occurs along the that is

necessary for the growth and maintenance of the cilium (8). IFT is dependent on

two large protein complexes: IFT-B is responsible for anterograde transport to

the ciliary tip and IFT-A is responsible for retrograde transport and return of

materials to the basal region (9). Mutations in many of these IFT components,

including IFT172, have been associated with disease (10–21).

IFT172 is a peripheral IFT-B complex protein that is thought to mediate the

transition from anterograde to retrograde transport at the tip of the cilium (22–24).

Therefore, even though it is associated with the anterograde machinery, its

temperature sensitive mutants in Chlamydomonas display phenotypes typical of

3 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

retrograde proteins, including elongation of the cilium (24, 25). Elongation of the

cilium was also observed in cultured fibroblasts obtained from patients with

IFT172 associated disease (10). In contrast, node cells of mouse embryos

harboring a homozygous p.Leu1564Pro mutation in Ift172 (wim mutants) lacked

all cilia (26). Mice homozygous for loss of function Ift172 alleles wim and slb, or

the hypomorphic avc1 allele, die in utero or at birth respectively (26–28),

therefore photoreceptor degeneration in a murine Ift172 model has not been

described to date. However, homozygous ift172 zebrafish mutants have

abnormal photoreceptor development, where photoreceptor cilia fail to extend, in

addition to severe multi-systemic defects (29, 30).

Given IFT172’s role in the retinal disease, we created rod photoreceptor-specific

Ift172 knock-out mice by crossing mice that have a conditional Ift172 allele (31)

with mice that express Cre driven by the Rhodopsin promoter (32). We

generated and validated an IFT172-specific antibody, which allowed us to

monitor reduction of the IFT172 protein, and to demonstrate that depletion of

IFT172 in the mouse retina causes a rapid retinal degeneration. We also show

that decreasing levels of IFT172 lead to aberrant localization of specific proteins.

4 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

RESULTS

Targeted disruption of Ift172 in rod photoreceptors leads to Ift172 protein

depletion

A rod photoreceptor-specific Ift172 knock-out mouse was generated by crossing

mice carrying a conditional knock-out allele (Ift172fl) (31) with transgenic mice

harbouring Cre recombinase under a Rhodopsin promoter (iCre) (32) (Figure

1A). Expression of iCre in the Ift172 knockout mouse (Ift172fl/fl/iCre) was specific

to the outer nuclear layer (ONL) and expressed in the majority of the rod

photoreceptors at postnatal day 25 (PN25) (Figure 1B). iCre is necessary for

generating the Ift172 knock-out allele (Ift172fliCre) and as a consequence for

depletion of the IFT172 protein. Therefore, the time-course of IFT172 protein

depletion was monitored by immunostaining of mouse with a specific anti-

IFT172 antibody, developed to the C-terminus of the protein (Figure 1C).

Complete depletion of the IFT172 protein in rod photoreceptors was observed at

PN28 (Figure 1D).

IFT172 deficient mouse retinas degenerate rapidly

Retinas of the Ift172fl/fliCre mice showed thinning of the ONL at one month and

complete degeneration of the ONL by two months, whereas age-matched

littermate controls did not show this phenotype (Figure 2A-C, Supplementary

Figure 1). Electroretinography (ERG) testing concordantly demonstrated that at

one month Ift172fl/fliCre mice showed a significantly reduced (84% reduction) rod-

driven b-wave amplitude (32.3 µV ±12 µV) compared to the wild-type controls

(196.9 µV ±12 µV). Mixed rod/cone and cone-isolated responses were reduced

5 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

by half at the same age, indicating a secondary cone degeneration (Figure 2D).

By two months of age, the ERG was undetectable in Ift172fl/fliCre mice across the

stimulus conditions (Figure 2D). Mice homozygous for the floxed allele (Ift172fl/fl)

or heterozygous retina specific knock-out mice (Ift172wt/fliCre) were followed up to

6 months and maintained normal retinal function and structure (Figure 2D,

Supplementary Figure 1).

Ultrastructural analyses of the retina in the Ift172fl/fliCre mice showed no signs of

photoreceptor degeneration at PN21 (Figure 3). However, at PN25, shortening of

the OS was observed, and at PN28, OS disc disorganization and accumulation of

extracellular debris was seen (Figure 3). At these stages, no changes in

axoneme length were observed. At PN31 the outer-segments were disorganized

and they appeared to have lost the connection with the axoneme and the inner

segment of the (Figure 3). Based on these results, further

studies were performed on mice up to PN28.

Depletion of IFT172 leads to mislocalization of Rp1 and Ift139

Disruption of the outer-segment structure led us to investigate the localization of

the axoneme-associated proteins RP1 and acetylated tubulin (acTub)(33). AcTub

showed no differences in localization between the mutant and the control retinas

up to PN28. RP1 staining, however, showed shortening of the signal relative to

acTub at PN28 in mutant mice compared with the controls (Figure 4A), and it

was observed to mislocalize to the synaptic terminals (Figure 4B).

6 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Since IFT172 is thought to be involved in the switching between the anterograde

and retrograde transport, mislocalization of the IFT components is expected. For

example, before the complete cessation of IFT, accumulation of the IFT particles

at the tip of the cilium would be likely, as seen in Chlamydomonas (24, 25). We

therefore tested localization of two components of the retrograde IFT (IFT43 and

IFT139) and a Bbsome protein (Bbs9). Of these three, only IFT139 showed

mislocalization at the synaptic terminals of rods, however no changes in location

at the axoneme were seen (Figure 4B, Supplementary Figure 2).

Mislocalization of photoreceptor outer segment proteins in Ift172fl/fliCre

mice

Depletion of a component of the intraflagellar transport machinery is expected to

affect IFT overall and thus alter trafficking of the outer-segment proteins.

Localization of four different outer-segment proteins was investigated: two

phototransduction proteins dependent on IFT (rhodopsin (Rho) and guanylate

cyclase-1 (Gc-1)) (34–36), one protein supporting disk structure (peripherin

(Prph2))(37) and one phototransduction protein independent of IFT (transducin)

(38, 39). Early signs of RHO mislocalization became apparent at PN21 and

PN25, where the protein was detected around the nuclei. This pattern became

stronger at PN28 (Figure 5). Depletion of IFT172, however, had no visible effect

on localization of GC-1 and PRPH2 (Figure 5).

Localization of transducin in rods is light dependent, where in the darkness,

transducin is predominantly present in the outer-segments and upon bright light

7 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

stimulation it is translocated to the inner-segment (40, 41). This light-driven

translocation is an adaptation of rods to be able to respond to different light

conditions and it is widely believed to occur by diffusion (38, 39, 42–44). To

establish if depletion of IFT172 had an effect on the translocation of the alpha

subunit of transducin (Gαt), we tested the localization of this protein in dark-

adapted and in photobleached retinas from Ift172fl/fliCre and Ift172fl/fl control

littermates at PN21 and PN25 (Figure 6). These two time-points were chosen

because the mice show only early signs of retinal degeneration in which

alteration of the photoreceptor structure and early signs of RHO mislocalization

(Figure 3 and 5) are mild or absent. At PN21 the location of Gαt followed the

expected pattern, with the protein detected only in the outer-segments of the

dark-adapted photoreceptors and distributed throughout the photoreceptors in

the light-adapted tissue (Figure 6A,B). At PN25 however, Gαt is detectable in the

cell bodies of the dark-adapted photoreceptors of the Ift172fl/fliCre to a greater

extent than in the Ift172fl/fl control mice, however after Bonferroni-Sidak mutli-

comparison corrections, this difference was not statistically significant (Figure 6C,

E). In the light, more Gαt protein was seen in the inner-segment of the

Ift172fl/fliCre than the control mice (Figure 6D). Quantification of the relative

fluorescence signal in the outer-segments compared to the total fluorescence of

the retina, revealed that the Gαt content in outer-segment is lower in the mutant

mice compared to the controls by 36.3% (p=0.0003) (Figure 6E).

8 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

IFT172 deficient mouse retinas show early mislocalization of hedgehog

signaling pathway protein Gli1

Primary cilia that modulate developmental signaling events, such as Hedgehog

(Hh) signaling and all three hedgehog signaling proteins (Sonic, Desert, and

Indian), have been shown to be expressed by the retinal ganglion cells or the

retinal pigment epithelium in the developing and adult murine eye (45). Moreover,

depletion of IFT172 has been shown to affect expression of Hh components in

the developing brain (28). We therefore investigated the role of Hh signaling in

the degenerating photoreceptors due to abnormal IFT. Using

immunofluorescence analyses, we investigated the localization of smoothened

(Smo) and the glioma-associated oncogene (Gli) family members 1, 2, and 3 (46,

47). Of the four proteins, only Gli1 showed differing localization in the Ift172fl/fliCre

retinas compared to the control mice. In control mice, Gli1 was predominantly

detected in the outer segments, while in Ift172fl/fliCre mice Gli1 localized to the

inner-segments at PN25 and at PN28. At PN28, the mutant photoreceptors had a

substantially reduced expression of Gli1, as well (Figure 7). Smo localized to the

base of the photoreceptor cilia in the control and the mutant mice, Gli2 showed a

diffuse inner-segment staining, and Gli3 expression was seen in the inner

nuclear layer and ganglion cells but not in the photoreceptors (Supplementary

Figure 3).

9 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

DISCUSSION

We have generated a murine model of retinal degeneration due to mutations in

IFT172, a previously implicated in a syndromic and a non-syndromic retinal

degeneration (10, 11, 21). Since general disruption of Ift172 is embryonic lethal

(27, 28), we used post-natal Cre-mediated excision of exons 2 and 3 in rods. Cre

recombinase under control of the Rho promoter (iCre) showed increasing

expression through to PN25, which led to undetectable levels of the IFT172

protein by PN28. Of note, the iCre recombinase expression was observed later

than previously reported (32), which may be due to a lower copy of the transgene

caused by numerous mouse crosses and due to chromatin modifications that

may affect the transgene expression timeline (48–50).

The mutant Ift172 mice showed a rapid retinal degeneration with severely

reduced retina thickness and ERG responses by one month and absent

photoreceptor cells by two months. The first signs of photoreceptor outer-

segment shortening were seen before the complete IFT172 depletion, at PN25

and within six days, by PN31, the outer-segments were fully degenerated. The

degeneration occurs therefore faster than the normal 10 day cycle of the outer-

segment regeneration in mice (51). This indicates that shortening of the outer-

segments is not only due to the cessation of transport of the proteins necessary

for the outer-segment regeneration, but implies an additional function of IFT in

the photoreceptors.

10 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Other selected outer-segment/cilia related proteins, such as RP1 and IFT139

showed mislocalization, which may indicate a general protein trafficking

disturbance in the cell. RHO accumulation in the bodies of the mutant

mice was first observed at PN21, before any visible signs of structural changes to

the outer-segments. This was the first symptom of the failing IFT machinery that

was observed in the Ift172fl/fliCre mice. RHO mislocalization has been reported in

other murine models, where anterograde complex B (Ift88) and retrograde

complex A (Ift140) genes were perturbed (35, 52). The result obtained in this

study adds further evidence supporting IFT’s role in opsin transport.

Localization of two other outer-segment proteins, GC1 and PRPH2 was not

altered in the mutant mice up to PN28 (Figure 5). PRPH2, is a structural protein,

necessary for the outer-segment disc rim formation (42). Previous studies have

indicated that PRPH2 and RHO utilize separate methods of transport to reach

the outer segment, which is in agreement with our findings (37, 42, 53). The lack

of GC1 mislocalization was, however, surprising since it requires RHO for

trafficking to the outer-segments (53). One of the reasons for this result could be

a low abundance of the GC1 protein and the low sensitivity of the

immunohistochemical assay. Of note, GC1 accumulation was also absent in the

rod cell bodies of the Rho-/- retinas (53).

Unexpectedly, decreasing levels of IFT172 affected light-driven translocation of

alpha transducin at PN25. Specifically, transducin showed increased localization

11 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

within the inner segment and around the photoreceptor nuclei after light

adaptation (Figure 6). It is thought that light-mediated translocation of transducin

and the reciprocal translocation of arrestin occur via diffusion and thus are IFT-

independent (38–44). Therefore, it is not clear how perturbed IFT can alter

protein diffusion through the transition zone. It is plausible that at the PN25 time

point, the structure of the axoneme starts to disintegrate, forming a barrier

against diffusion. A similar conclusion has been drawn from studies in mouse

retinal explants, where drug-mediated disruption of actin filaments and

led to impaired distribution of transducin and arrestin during the

dark adaptation (54). Involvement of cytoskeleton in light-mediated protein

translocation was also observed in Xenopus laevis (55, 56). Another possible

explanation is that the transducin mislocalization observed in our model is

secondary to the impaired trafficking of rhodopsin. Current evidence suggests

that light activated rhodopsin and intact disc membranes are needed to act as

“sinks” for arrestin/transducin transport (43, 57, 58). Therefore it is plausible that

perturbations in rhodopsin transport would lead to secondary mislocalization of

transducin. The timeline of this theory also matches our findings of rhodopsin

mislocalization at PN21, preceding transducin mislocalization detectable at

PN25.

Numerous studies have shown the importance of ciliogenesis and IFT on Hh

signaling during development; however, little is known about how the Hh

signaling pathway functions in cilia after development and during degeneration

12 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

(26, 59–63). Generally, activation of the Hh pathway in vertebrate cells involves

binding of the ligand (Sonic, Desert or Indian Hedgehog) by the Patched

(PTCH1) receptor present at the ciliary membrane and subsequent activation

and translocation of SMO to the cilium. SMO subsequently blocks GLI repressors

(mostly GLI3) and triggers GLI transcriptional activators (mostly GLI2 and GLI1),

which translocate from the cilium to the nucleus (62, 63). Double mutant mouse

experiments have demonstrated that IFT acts downstream of the Hh receptor

PTCH1 and SMO, but upstream of the GLI effectors (26, 60, 62, 63). We

therefore used immunofluorescence analyses to determine the localization of key

Hh pathway components in the degenerating retina. Lack of differences in

localization of SMO, GLI2, and GLI3 indicate that Hh pathway was not activated

in the degenerating retina of the Ift172fl/fliCre mice. Immunofluorescence analysis

of GLI1 however, gave contrary results, as this protein translocated from the

cilium to the inner segments and the peri-nuclear region of the degenerating

photoreceptors. Functional differences between GLI1 and GLI2/GLI3

transcription factors have been noted before. For example in contrast to GLI2

and GLI3, GLI1 lacks a repressor domain and homozygous Gli1 knock-out mice

develop normally (46, 64). GLI1 has also been reported to be activated in a Hh-

independent fashion (65), which is likely the case in the present study. Currently,

the role of GLI1 in developed photoreceptors is unknown. Whether the observed

translocation of GLI1 from the outer segment has a protective function or if it

enhances photoreceptor degeneration remains to be determined. Further

13 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

research will be necessary to fully answer the question about its role in normal

and degenerating retinas.

The results of this study highlight the crucial nature of transition from anterograde

to retrograde IFT within the rod photoreceptors and expand our understanding of

IFT’s role in protein trafficking within these cells. A profound degeneration of the

outer segments occurred within just a matter of days after IFT172 was depleted,

which was shown by a thorough characterization of the degeneration between

PN21 and PN28. Phenotypic and molecular results from this time-course

analysis indicate that the hypomorphic condition present in patients with IFT172

associated retinal degeneration is most accurately modeled at PN25, when

depletion of some but not all of the IFT172 protein leads to the first signs of

photoreceptor degeneration. This model is therefore useful to understand the

IFT-associated disease and for the development of potential treatments for

patients with the IFT172-associated disease.

14 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

MATERIALS AND METHODS

Ethics Statement: The in-vivo experiments performed on mice were performed

according to protocols approved by the Animal Care Committee from the

Massachusetts Eye and Ear. All procedures were performed to minimize

suffering in accordance with the animal care rules in our institution in compliance

with the Animal Welfare Act, the Guide for the Care and Use of Laboratory

Animals, and the Public Health Service Policy on Humane Care and Use of

Laboratory Animals.

Mouse husbandry and genotyping: Mice were housed in the animal facility of

the Schepens Eye Research Institute, provided a standard rodent diet and

housed in standard 12 hour alternating light/dark cycles.

Genomic DNA was isolated from ear biopsies (Allele-In-One reagent, Allele

Biotechnology, San Diego, CA, USA). Two genotyping PCR reactions (Hot

FirePol DNA polymerase, Solis Biodyne, Tartu, Estonia) were performed for the

Ift172 allele (F: 5’-GAAGAGTTGGGTGTAAGAAATGC -3’ and R: 5’-

CTGGAGCTACATCAAAGACAG-3’) and the iCre allele (F: 5’-

TCAGTGCCTGGAGTTGCGCTGTGG-3’ and R: 5’-

CACAGACAGGAGCATCTTCCAG-3’). Both PCR reactions were performed in

0 2.0mM MgCl2 with the thermocycling protocol as follows: 95 C for 15 minutes; 35

cycles of 940C for 30 seconds, 600C for iCre/620C for Ift172 for 30 seconds; 720C

for 1 minute; final extension: 720C for 5 minutes. Gel electrophoresis was

performed on the PCR products. The Ift172 floxed allele produced a 380 base

15 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

pair band, whereas the wild type ift172 allele produced a 176 band.

Presence of the iCre allele produced a 200 base pair band.

Electroretinography (ERG): Full-field ERGs were performed on mice at 1, 2, 3

and 6 months of age to assess rod and cone photoreceptor function (66). Mice

were dark adapted overnight, then approximately 20 minutes before procedure

they were anesthetized by intraperitoneal injection of ketamine and xylazine

diluted in sterile saline, and the eyes were dilated by topical application of 1%

tropicamide. ERG was recorded simultaneously from both eyes in response to 4-

ms broadband stimuli with the use of gold ring electrodes (Diagnosys, Lowell,

MA) and the ColorDome Ganzfeld system (Diagnosys), as reported before (67).

In dark-adapted mice, rod-driven ERGs were recorded in response to a 0.01

cd·s/m2 light stimulus (10 flashes at 0.2 Hz), and mixed rod/cone ERGs were

recorded in response to a 10 cd·s/m2 light stimulus (three flashes at 0.03 Hz).

Next, mice were light-adapted by exposure to a steady, rod-suppressing

background light (30 cd/m2) for 10 minutes. This background light remained

during the acquisition of cone-driven ERGs that were recorded in response to a

20 cd·s/m2 light stimulus (20 flashes at 0.5 Hz). The magnitude of the ERG b-

wave was measured as the absolute voltage change from the trough of the a-

wave (or from the voltage measured at the expected implicit time of that trough,

should the a-wave be undetectable) to the b-wave peak (67).

16 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Optical Coherence Tomography (OCT): OCT was performed on mice at 1, 2,

and 3 months of age. After anesthesia and pupil dilation (as above), cross-

sectional retinal images were acquired with the InVivoVue OCT system

(Bioptogen, Morrisville, NC). A rectangular and radial OCT volume centered on

the optic nerve head was captured for both eyes. A B-scan located approximately

200 µm temporal and nasal of the ventral optic nerve head was selected for

measurement. Retinal thickness was measured as the distance between the

nerve fiber layer and the hyporeflective boundary between the retinal pigment

epithelium (RPE) and choroid.

Histology: The mice at 1, 2, 3 and 6 months of age were sacrificed by

transcardial perfusion (4% paraformaldehyde, approximately 30ml per mouse).

After perfusion, each eye was enucleated and placed in 4% PFA in PBS

overnight for post-fixation. After dehydration with graded ethanol solutions, the

eyes were embedded in glycol methacrylate (GMA) (Technovit 7100, Heraeas

Kulzer GmBH, Wehrheim, Germany) and 3µm thick sections were cut at the level

of the optic nerve head (LKB Historange microtome). The sections were stained

with hematoxylin and eosin and imaged by bright field microscopy (Eclipse Ti,

Nikon, Tokyo, Japan).

Immunofluorescence: The pups at ages PN18 to PN31 were sacrificed by CO2

asphyxiation and the eyes were enucleated. Since many of the cilia antibodies

are incompatible with tissue fixation, one eye was fresh frozen and one fixed. For

17 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

the non-fixed tissue, the eye was immediately embedded (OCT reagent, Tissue-

Tek, Fisher Scientific, USA) within molds and snap-frozen using 200 proof

ethanol over dry ice. For the fixed tissues, the cornea and the lens were

dissected and the eyes were placed in 4% paraformaldehyde (PFA) for 1 hour

post-fixation, and the eyecups were cryopreserved (30% sucrose in phosphate

buffered saline buffer (PBS), overnight at 4°C) (Sigma-Aldrich, USA) and

embedded (OCT reagent). The eyes were cryosectioned into 10 µm slices and

then processed using the following procedure: blocking and permeabilization

(0.05% Triton X100, 1% Bovine Serium Albumin (BSA) in PBS, 1 hour at RT);

primary antibody staining (dilutions listed below, 0.3% Triton X100 in PBS,

overnight at 4°C); incubation of the secondary antibodies conjugated with a

fluorophore (Alexa Fluor 488, AlexaFluor 555, and AlexaFluor 647)

(ThermoFisher, Waltham, MA, USA) (1:500 dilution, 0.3% Triton X100 in PBS, 2

hours at RT). The nuclei were counterstained with Hoechst after the secondary

antibody incubation (1:1000 dilution in PBS for 15 minutes at RT). In between

the steps the retina sections were washed four times with PBS. The slides were

mounted with coverslips with Fluoromount medium (Electron Microscopy

Sciences, Hatfield, PA, USA). The images were acquired in a sequential manner,

with pictures taken every 0.5µm in the z-plane using confocal microscopy (SP5,

Leica Microsystems, USA). Figure 5 images were taken with a fluorescent

microscope (Eclipse Ti, Nikon, Tokyo, Japan).

18 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Primary antibodies used on 4% PFA fixed tissues were: mouse-anti-Cre (1:500,

Millipore, MAB3120); mouse-anti-Rhodopsin (1:1000, Millipore, MAB5356);

mouse-anti-Transducin (1:100, BD BioSciences, 610589). Primary antibodies

used on nonfixed tissue after brief postfixation on the slide (1% PFA in PBS for

15 minutes prior to staining) were: guinea pig-anti-GLI2 (1:1000, kind gift from Dr.

Eggenschwiler); rabbit-anti-GLI3 (1:200, Thermo Fisher, PS5-28029); mouse-

anti-acetylated tubulin (1:200, Sigma-Aldrich, T6793-100uL); rabbit-anti-IFT139

(1:500, custom). Primary antibodies used with nonfixed tissue were rabbit-anti-

IFT43 (1:100, Proteintech, 24338-1-AP); mouse-anti-PRPH2 (1:15, kind gift from

Dr. Molday); rabbit-anti-GLI1 (1:200, abcam, ab92611); rabbit-anti-SMO (1:1000,

kind gift from Dr. Anderson); rabbit-anti-BBS9 (1:500, Sigma-Aldrich,

HPA021289-100UL); mouse-anti-Gc1 (1:1000, kind gift from Dr. Baer); chicken-

anti-RP1 (1:1000, custom); rabbit-anti-IFT172 (1:1000, custom).

Ift172 Antibody Development: A custom affinity-purified rabbit anti-IFT172

polyclonal antibody was generated through a comercial provider (SC1676

PolyExpress Premium Service, GenScript USA Piscataway, NJ, USA). For rabbit

immunizations a 488 amino acid peptide from the C terminus of the human

IFT172 protein was chosen:

EEYEREATKKGARGVEGFVEQARHWEQAGEYSRAVDCYLKVRDSGNSGLAE

KCWMKAAELSIKFLPPQRNMEVVLAVGPQLIGIGKHSAAAELYLNLDLVKEAIDA

FIEGEEWNKAKRVAKELDPRYEDYVDQHYKEFLKNQGKVDSLVGVDVIAALDL

YVEQGQWDKCIETATKQNYKILHKYVALYATHLIREGSSAQALALYVQHGAPAN

19 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

PQNFNIYKRIFTDMVSSPGTNCAEAYHSWADLRDVLFNLCENLVKSSEANSPA

HEEFKTMLLIAHYYATRSAAQSVKQLETVAARLSVSLLRHTQLLPVDKAFYEAGI

AAKAVGWDNMAFIFLNRFLDLTDAIEEGTLDGLDHSDFQDTDIPFEVPLPAKQH

VPEAEREEVRDWVLTVSMDQRLEQVLPRDERGAYEASLVAASTGVRALPCLIT

GYPILRNKIEFKRPGKAANKDNWNKFLMAIKTSHSPVCQDVLKFISQWCGGLPS

TSFSFQ

Transmission Electron Microscopy (TEM) Materials & Methods: Mice were

sacrificed by transcardial perfusion with half strength Karnovsky’s fixative (2%

formaldehyde + 2.5% glutaraldehyde, in 0.1 M sodium cacodylate buffer, pH 7.4;

Electron Microscopy Sciences, Hatfield, Pennsylvania) at room temperature.

The eyes were enucleated and after cornea and lens dissection they were placed

back into the half strength Karnovsky’s fixative for a minimum of 24 hours under

refrigeration. After fixation, samples were rinsed with 0.1M sodium cacodylate

buffer, post-fixed with 2% osmium tetroxide in 0.1M sodium cacodylate buffer for

1.5 hours, en bloc stained with 2% aqueous uranyl acetate for 30 minutes, then

dehydrated with graded ethyl alcohol solutions, transitioned with propylene oxide

and resin infiltrated in tEPON-812 epoxy resin (Tousimis, Rockville, Maryland)

utilizing an automated EMS Lynx 2 EM tissue processor (Electron Microscopy

Sciences, Hatfield, Pennsylvania.) The processed samples were oriented into

tEPON-812 epoxy resin inside flat molds and polymerized in a 60oC oven.

Semi-thin sections were cut at 1 µm thickness through the mid-equatorial plane

traversing the optic nerve in the posterior eyecup samples and stained with 1%

20 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

toluidine blue in 1% sodium tetraborate aqueous solution for assessment by light

microscopy. Ultrathin sections (80 nm) were cut from each sample block using a

Leica EM UC7 ultramicrotome (Leica Microsystems, Buffalo Grove, IL, USA) and

a diamond knife, then collected using a loop tool onto either 2x1 mm, single slot

formvar-carbon coated or 200 mesh uncoated copper grids and air-dried. The

thin sections on grids were stained with aqueous 2.5% aqueous gadolinium

triacetate hydrate and Sato’s lead citrate stains using a modified Hiraoka grid

staining system. Grids were imaged using a FEI Tecnai G2 Spirit transmission

electron microscope (FEI, Hillsboro, Oregon) at 8o kV interfaced with an AMT

XR41 digital CCD camera (Advanced Microscopy Techniques, Woburn,

Massachusetts) for digital TIFF file image acquisition. TEM imaging of retinas

were assessed and digital images captured at 2kx2k pixel, 16-bit resolution.

Transducin Translocation experiment

In this experiment, the mice were distributed in 2 different groups. For each

group, three Ift172fl/fliCre mice and littermate Ift172fl/fl control mice were used.

Group 1: The mice were dark-adapted overnight, euthanized in darkness with

CO2 and their eyes were enucleated under dark conditions, fixed in (4% PFA in

PBS) and prepared for cryosectioning as described in previous section. Group 2:

The pupils of mice were dilated as described above in ERG section, and mice

were exposed to saturating light conditions (1400lux) for 20 min. CO2 was used

to euthanize the mice, and their eyes were enucleated and prepared from

dryosectioning as before. The eyes from different conditions were sectioned in

one block and they were immonolabeled with mouse-anti-Transducin antibody

21 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

(1:100, BD BioSciences, 610589) on the same slide. The images were taken with

confocal microscopy (SP5, Leica Microsystems, USA). Image J software was

used to analyse signal intensities, where signal form the OS was divided by the

total signal form the whole retina. The analysis conditions were kept exact for test

and control mice. Two-way ANOVA and a post-hoc multiple t-test with

Bonferroni-Sidak correction were used for statistical analysis (Graphpad, Prism 7

software).

Acknowledgments

This work was supported by grants from the Fight for Sight (2016 International

Retinal Research Foundation Grant-In-Aid Award to KMB), Research to Prevent

Blindness Medical Student Research Fellowship (PRG), National Eye Institute

[EY012910 (EAP) and P30EY014104 (MEEI core support), P30EYE003790

(SERI core support)], the Foundation Fighting Blindness (USA, EAP).

Conflict of Interest Statement: The authors declare no conflict of interest

22 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

REFERENCES

1. Goetz,S.C. and Anderson,K. V (2010) The primary cilium: a signalling centre during vertebrate development. Nat. Rev. Genet., 11, 331–344. 2. Mockel,A., Perdomo,Y., Stutzmann,F., Letsch,J., Marion,V. and Dollfus,H. (2011) Retinal dystrophy in Bardet-Biedl syndrome and related syndromic ciliopathies. Prog. Retin. Eye Res., 30, 258–274. 3. Cortés,C.R., Metzis,V. and Wicking,C. (2015) Unmasking the ciliopathies: craniofacial defects and the primary cilium. Wiley Interdiscip. Rev. Dev. Biol., 10.1002/wdev.199. 4. Waters,A.M. and Beales,P.L. (2011) Ciliopathies: an expanding disease spectrum. Pediatr. Nephrol., 26, 1039–1056. 5. Ramamurthy,V. and Cayouette,M. (2009) Development and disease of the photoreceptor cilium. Clin. Genet., 76, 137–145. 6. Rachel,R.A., Li,T. and Swaroop,A. (2012) Photoreceptor sensory cilia and ciliopathies: focus on CEP290, RPGR and their interacting proteins. Cilia, 1, 22. 7. Bujakowska,K.M., Liu,Q. and Pierce,E.A. (2017) Photoreceptor Cilia and Retinal Ciliopathies. Cold Spring Harb. Perspect. Biol., 10.1101/cshperspect.a028274. 8. Pedersen,L.B. and Rosenbaum,J.L. (2008) Intraflagellar transport (IFT) role in ciliary assembly, resorption and signalling. Curr. Top. Dev. Biol., 85, 23–61. 9. Williamson,S.M., Silva,D. a, Richey,E. and Qin,H. (2012) Probing the role of IFT particle complex A and B in flagellar entry and exit of IFT- in Chlamydomonas. Protoplasma, 249, 851–856. 10. Halbritter,J., Bizet,A.A., Schmidts,M., Porath,J.D., Braun,D.A., Gee,H.Y., McInerney-Leo,A.M., Krug,P., Filhol,E., Davis,E.E., et al. (2013) Defects in the IFT-B component IFT172 cause Jeune and Mainzer-Saldino syndromes in humans. Am. J. Hum. Genet., 93, 1–11. 11. Bujakowska,K.M., Zhang,Q., Siemiatkowska,A.M., Liu,Q., Place,E., Falk,M.J., Consugar,M., Lancelot,M.-E., Antonio,A., Lonjou,C., et al. (2015) Mutations in IFT172 cause isolated retinal degeneration and Bardet-Biedl syndrome. Hum. Mol. Genet., 24, 230–242. 12. Davis,E.E., Zhang,Q., Liu,Q., Diplas,B.H., Davey,L.M., Hartley,J., Stoetzel,C., Szymanska,K., Ramaswami,G., Logan,C. V, et al. (2011) TTC21B contributes both causal and modifying alleles across the ciliopathy spectrum. Nat. Genet., 43, 189–196. 13. Coussa,R.G., Otto,E.A., Gee,H.-Y., Arthurs,P., Ren,H., Lopez,I., Keser,V., Fu,Q., Faingold,R., Khan,A., et al. (2013) WDR19: an ancient, retrograde, intraflagellar ciliary protein is mutated in autosomal recessive and in Senior-Loken syndrome. Clin. Genet., 84, 150–9. 14. Arts,H.H., Bongers,E.M.H.F., Mans,D.A., van Beersum,S.E.C., Oud,M.M., Bolat,E., Spruijt,L., Cornelissen,E.A.M., Schuurs-Hoeijmakers,J.H.M., de Leeuw,N., et al. (2011) C14ORF179 encoding IFT43 is mutated in Sensenbrenner syndrome. J. Med. Genet., 48, 390–395. 15. Bredrup,C., Saunier,S., Oud,M.M., Fiskerstrand,T., Hoischen,A.,

23 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Brackman,D., Leh,S.M., Midtbø,M., Filhol,E., Bole-Feysot,C., et al. (2011) Ciliopathies with skeletal anomalies and renal insufficiency due to mutations in the IFT-A gene WDR19. Am. J. Hum. Genet., 89, 634–643. 16. Huber,C. and Cormier-Daire,V. (2012) Ciliary disorder of the skeleton. Am. J. Med. Genet. Part C Semin. Med. Genet., 160 C, 165–174. 17. Aldahmesh,M.A., Li,Y., Alhashem,A., Anazi,S., Alkuraya,H., Hashem,M., Awaji,A.A., Sogaty,S., Alkharashi,A., Alzahrani,S., et al. (2014) IFT27, encoding a small GTPase component of IFT particles, is mutated in a consanguineous family with Bardet-Biedl syndrome. Hum. Mol. Genet., 23, 3307–15. 18. Girisha,K.M., Shukla,A., Trujillano,D., Bhavani,G.S., Hebbar,M., Kadavigere,R. and Rolfs,A. (2016) A homozygous nonsense variant in IFT52 is associated with a human skeletal ciliopathy. Clin. Genet., 10.1111/cge.12762. 19. Walczak-Sztulpa,J., Eggenschwiler,J., Osborn,D., Brown,D.A., Emma,F., Klingenberg,C., Hennekam,R.C., Torre,G., Garshasbi,M., Tzschach,A., et al. (2010) Cranioectodermal Dysplasia, Sensenbrenner syndrome, is a ciliopathy caused by mutations in the IFT122 gene. Am. J. Hum. Genet., 86, 949–956. 20. Bizet,A.A., Becker-Heck,A., Ryan,R., Weber,K., Filhol,E., Krug,P., Halbritter,J., Delous,M., Lasbennes,M.-C., Linghu,B., et al. (2015) Mutations in TRAF3IP1/IFT54 reveal a new role for IFT proteins in microtubule stabilization. Nat. Commun., 6, 8666. 21. Schaefer,E., Stoetzel,C., Scheidecker,S., Geoffroy,V., Prasad,M.K., Redin,C., Missotte,I., Lacombe,D., Mandel,J.-L., Muller,J., et al. (2016) Identification of a novel mutation confirms the implication of IFT172 (BBS20) in Bardet–Biedl syndrome. J. Hum. Genet., 10.1038/jhg.2015.162. 22. Lucker,B.F., Behal,R.H., Qin,H., Siron,L.C., Taggart,W.D., Rosenbaum,J.L. and Cole,D.G. (2005) Characterization of the intraflagellar transport complex B core: direct interaction of the IFT81 and IFT74/72 subunits. J. Biol. Chem., 280, 27688–27696. 23. Taschner,M., Bhogaraju,S. and Lorentzen,E. (2012) Architecture and function of IFT complex proteins in ciliogenesis. Differentiation., 83, S12–S22. 24. Pedersen,L.B., Miller,M.S., Geimer,S., Leitch,J.M., Rosenbaum,J.L. and Cole,D.G. (2005) Chlamydomonas IFT172 is encoded by FLA11 , interacts with CrEB1, and regulates IFT at the flagellar tip. Curr. Biol., 15, 262–266. 25. Iomini,C., Babaev-Khaimov,V., Sassaroli,M. and Piperno,G. (2001) Protein particles in Chlamydomonas flagella undergo a transport cycle consisting of four phases. J. Cell Biol., 153, 13–24. 26. Huangfu,D., Liu,A., Rakeman,A.S., Murcia,N.S., Niswander,L. and Anderson,K. V (2003) Hedgehog signalling in the mouse requires intraflagellar transport proteins. Nature, 426, 83–87. 27. Friedland-Little,J.M., Hoffmann,A.D., Ocbina,P.J.R., Peterson,M.A., Bosman,J.D., Chen,Y., Cheng,S.Y., Anderson,K. V and Moskowitz,I.P. (2011) A novel murine allele of Intraflagellar Transport Protein 172 causes a syndrome including VACTERL-like features with hydrocephalus. Hum. Mol.

24 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Genet., 20, 3725–3737. 28. Gorivodsky,M., Mukhopadhyay,M., Wilsch-Braeuninger,M., Phillips,M., Teufel,A., Kim,C., Malik,N., Huttner,W. and Westphal,H. (2009) Intraflagellar transport protein 172 is essential for primary cilia formation and plays a vital role in patterning the mammalian brain. Dev. Biol., 325, 24–32. 29. Lunt,S.C., Haynes,T. and Perkins,B.D. (2009) Zebrafish ift57, ift88, and ift172 intraflagellar transport mutants disrupt cilia but do not affect hedgehog signaling. Dev. Dyn., 238, 1744–1759. 30. Sukumaran,S. and Perkins,B.D. (2009) Early defects in photoreceptor outer segment morphogenesis in zebrafish ift57, ift88 and ift172 Intraflagellar Transport mutants. Vision Res., 49, 479–489. 31. Howard,P., Howard,T. and Maurer,R. (2010) Generation of mice with a conditional allele for Ift172. Transgenic Res., 19, 121–126. 32. Li,S., Chen,D., Sauvé,Y., McCandless,J., Chen,Y.-J. and Chen,C.-K. (2005) Rhodopsin-iCre transgenic mouse line for Cre-mediated rod-specific gene targeting. Genesis, 41, 73–80. 33. Liu,Q., Lyubarsky,A., Skalet,J.H., Pugh,E.N. and Pierce,E.A. (2003) RP1 is required for the correct stacking of outer segment discs. Investig. Ophthalmol. Vis. Sci., 44, 4171–4183. 34. Marszalek,J.R., Liu,X., Roberts,E. a, Chui,D., Marth,J.D., Williams,D.S. and Goldstein,L.S. (2000) Genetic evidence for selective transport of opsin and arrestin by kinesin-II in mammalian photoreceptors. Cell, 102, 175–187. 35. Pazour,G.J., Baker,S. a, Deane,J. a, Cole,D.G., Dickert,B.L., Rosenbaum,J.L., Witman,G.B. and Besharse,J.C. (2002) The intraflagellar transport protein, IFT88, is essential for vertebrate photoreceptor assembly and maintenance. J. Cell Biol., 157, 103–13. 36. Insinna,C. and Besharse,J. (2008) Intraflagellar transport and the sensory outer segment of vertebrate photoreceptors. Dev. Dyn., 237, 1982–1992. 37. Lee,E.S., Burnside,B. and Flannery,J.G. (2006) Characterization of peripherin/rds and Rom-1 transport in rod photoreceptors of transgenic and knockout animals. Investig. Ophthalmol. Vis. Sci., 47, 2150–2160. 38. Calvert,P.D., Strissel,K.J., Schiesser,W.E., Pugh,E.N. and Arshavsky,V.Y. (2006) Light-driven translocation of signaling proteins in vertebrate photoreceptors. Trends Cell Biol., 16, 560–568. 39. Calvert,P.D., Schiesser,W.E. and Pugh,E.N. (2010) Diffusion of a soluble protein, photoactivatable GFP, through a sensory cilium. J. Gen. Physiol., 135, 173–96. 40. Broekhuyse,R.M., Tolhuizen,E.F., Janssen,A.P. and Winkens,H.J. (1985) Light induced shift and binding of S-antigen in retinal rods. Curr. Eye Res., 4, 613–618. 41. Philp,N.J., Chang,W. and Long,K. (1987) Light-stimulated protein movement in rod photoreceptor cells of the rat retina. FEBS Lett., 225, 127–32. 42. Pearring,J.N., Salinas,R.Y., Baker,S.A. and Arshavsky,V.Y. (2013) Protein sorting, targeting and trafficking in photoreceptor cells. Prog. Retin. Eye Res., 36, 24–51. 43. Slepak,V.Z. and Hurley,J.B. (2008) Mechanism of light-induced translocation

25 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

of arrestin and transducin in photoreceptors: Interaction-restricted diffusion. IUBMB Life, 60, 2–9. 44. Nair,K.S., Hanson,S.M., Mendez,A., Gurevich,E. V., Kennedy,M.J., Shestopalov,V.I., Vishnivetskiy,S.A., Chen,J., Hurley,J.B., Gurevich,V. V., et al. (2005) Light-dependent redistribution of arrestin in vertebrate rods is an energy-independent process governed by protein-protein interactions. Neuron, 46, 555–567. 45. Wallace,V.A. (2008) Proliferative and cell fate effects of Hedgehog signaling in the vertebrate retina. Brain Res., 1192, 61–75. 46. Hui,C. and Angers,S. (2011) Gli Proteins in Development and Disease. Annu. Rev. Cell Dev. Biol., 27, 513–537. 47. Ocbina,P.J.R., Eggenschwiler,J.T., Moskowitz,I. and Anderson,K. V (2011) Complex interactions between genes controlling trafficking in primary cilia. Nat. Genet., 43, 547–553. 48. Montoliu,L., Chávez,S. and Vidal,M. (2000) Variegation associated with lacZ in transgenic animals: A warning note. Transgenic Res., 9, 237–239. 49. Li,Q., Emery,D.W., Han,H., Sun,J., Yu,M. and Stamatoyannopoulos,G. (2005) Differences of globin transgene expression in stably transfected cell lines and transgenic mice. Blood, 105, 3346–3352. 50. Vooijs,M., Jonkers,J. and Berns,A. (2001) A highly efficient ligand-regulated Cre recombinase mouse line shows that LoxP recombination is position dependent. EMBO Rep., 2, 292–297. 51. Young,R. (1983) The ninth Frederick H. Verhoeff lecture. The life history of retinal cells. Trans. Am. Ophthalmol. Soc., 81, 193–228. 52. Crouse,J.A., Lopes,V.S., Sanagustin,J.T., Keady,B.T., Williams,D.S. and Pazour,G.J. (2014) Distinct functions for IFT140 and IFT20 in opsin transport. Cytoskeleton, 71, 302–310. 53. Pearring,J.N., Spencer,W.J., Lieu,E.C. and Arshavsky,V.Y. (2015) Guanylate cyclase 1 relies on rhodopsin for intracellular stability and ciliary trafficking. Elife, 4, 1–13. 54. Reidel,B., Goldmann,T., Giessl,A. and Wolfrum,U. (2008) The translocation of signaling molecules in dark adapting mammalian rod photoreceptor cells is dependent on the cytoskeleton. Cell Motil. Cytoskeleton, 65, 785–800. 55. McGinnis,J.F., Matsumoto,B., Whelan,J.P. and Cao,W. (2002) Cytoskeleton participation in subcellular trafficking of signal transduction proteins in rod photoreceptor cells. J. Neurosci. Res., 67, 290–297. 56. Peterson,J.J., Orisme,W., Fellows,J., McDowell,J.H., Shelamer,C.L., Dugger,D.R. and Smith,W.C. (2005) A role for cytoskeletal elements in the light-driven translocation of proteins in rod photoreceptors. Investig. Ophthalmol. Vis. Sci., 46, 3988–3998. 57. Artemyev,N.O. (2008) Light-dependent compartmentalization of transducin in rod photoreceptors. Mol. Neurobiol., 37, 44–51. 58. Kerov,V. and Artemyev,N.O. (2011) Diffusion and light-dependent compartmentalization of transducin. Mol. Cell. Neurosci., 46, 340–346. 59. Weatherbee,S.D., Niswander,L.A. and Anderson,K. V. (2009) A mouse model for Meckel syndrome reveals Mks1 is required for ciliogenesis and

26 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Hedgehog signaling. Hum. Mol. Genet., 18, 4565–4575. 60. Huangfu,D. and Anderson,K. V (2005) Cilia and Hedgehog responsiveness in the mouse. Proc. Natl. Acad. Sci. U. S. A., 102, 11325–11330. 61. He,M., Agbu,S. and Anderson,K. V. (2016) Microtubule Motors Drive Hedgehog Signaling in Primary Cilia. Trends Cell Biol., xx, 1–16. 62. Bangs,F. and Anderson,K. V. (2017) Primary cilia and Mammalian Hedgehog signaling. Cold Spring Harb. Perspect. Biol., 9, 1–22. 63. Eggenschwiler,J.T. and Anderson,K. V. (2007) Cilia and Developmental Signaling. Annu. Rev. Cell Dev. Biol., 23, 345–373. 64. Park,H.L., Bai,C., Platt,K.A., Matise,M.P., Beeghly,A., Hui,C. c, Nakashima,M. and Joyner,A.L. (2000) Mouse Gli1 mutants are viable but have defects in SHH signaling in combination with a Gli2 mutation. Development, 127, 1593–605. 65. Nolan-Stevaux,O., Lau,J., Truitt,M.L., Chu,G.C., Hebrok,M., Fernández- Zapico,M.E. and Hanahan,D. (2009) GLI1 is regulated through Smoothened- independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation. Genes Dev., 23, 24–36. 66. Marmor,M.F., Fulton,A.B., Holder,G.E., Miyake,Y., Brigell,M. and Bach,M. (2009) ISCEV Standard for full-field clinical electroretinography (2008 update). Doc. Ophthalmol., 118, 69–77. 67. Greenwald,S.H., Charette,J.R., Staniszewska,M., Shi,L.Y., Brown,S.D.M., Stone,L., Liu,Q., Hicks,W.L., Collin,G.B., Bowl,M.R., et al. (2016) Mouse Models of NMNAT1-Leber Congenital Amaurosis (LCA9) Recapitulate Key Features of the Human Disease. Am. J. Pathol., 186, 1925–1938.

27 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Legends to Figures

Figure 1. Generation of the Ift172fl/fliCre Mouse Model and of the IFT172

Antibody. A. Schematic representation of the targeted Ift172 allele (Ift172fl/fl)

and the knck-out allele (Ift172fl/fliCre). B. Expression of Cre recombinase (red) in

the ONL at postnatal days 18, 21 and 25. Nuclei are counterstained with Hoechst

(blue). C. Schematic representation of human IFT172 protein, containing

repetitive W40 and TPR motifs. 488 carboxyl terminal amino acid residues were

chosen for the rabbit polyclonal antibody generation. D. Staining for IFT172 (red)

and RP1 (green) showing at postnatal days 21, 25 and 28, showing depletion of

IFT172 from rods by P28 in Ift172fl/fliCre mice.

Figure 2. Ift172fl/fliCre mice display rapid retinal degeneration. A. OCT data

obtained at one, two, and three months of age in Ift172fl/fliCre and wild-type

control mice, showing diminishing ONL in the Ift172fl/fliCre mice. B. Graphical

representation of the neural retina and the ONL thickness measured from OCT

images in Ift172fl/fliCre and control genotypes. C. Representative histology

sections from Ift172fl/fliCre and control mice. D. b-wave amplitudes form dark-

adapted and light-adapted ERGs performed on Ift172fl/fliCre and control

genotypes.

Figure 3. Transmission Electron Microscopy Data Ultrastructure of

Ift172fl/fliCre and control mice showing OS shortening starting at PN25 and

28 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

cellular debris at PN28 and PN31 (black arrows). Intact transition zone

are visible at PN28 but not at PN31, where severe photoreceptor

degeneration occurs. Scale bar on the lower magniticatin picture represents 2µm

and on the higher magnification picture 500nm.

Figure 4. Cilia-associated Protein Localization. A. Ciliary axoneme staining

with RP1 (green) and acetylated tubulin (red), showing shortening of the Rp1

signal relative to acetylated tubulin in Ift172fl/fliCre mice compared to controls at

PN28. RP1 mislocalization to the inner segment is visible in Ift172fl/fliCre mice

starting at PN25. B. Staining of IFT139 (red) at the base of the ciliary axoneme,

makerd with Rp1 staining (green). C. IFT139 (red) and RP1 (green) both

mislocalize to the synaptic region in Ift172fl/fliCre mice, but not in controls, starting

at PN25. Nuclei in A, B and C are counterstained with Hoechst (blue).

Figure 5. Outer Segment Protein Localizations. A. Rhodopsin (red)

mislocalization to the ONL is present at PN21 and progresses over time in

Ift172fl/fliCre mice compared with Ift172fl/fl controls. B and C. Guanyl cyclase 1

(green) and Peripherin2 (red) localize appropriately to the outer segment at all

tested time points in both Ift172fl/fliCre mice and Ift172fl/fl controls.

29 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Figure 6. Transducin translocation kinetics are affected in the absence of

IFT172. A. Retina cryosections from dark-adapted mice at PN21 stained for

transducin-Gαt (green), DAPI (blue) and Cre (magenta). Transducin is

predominantly localized in rod OS region in Ift172fl/fliCre (bottom panel) and the

littermate Ift172fl/fl controls (top panel). B. Retina cryosections from light-adapted

mice at PN21 stained with transducin-Gαt (green), DAPI (blue) and Cre

(magenta). In both Ift172fl/fliCre (bottom panel) and littermate Ift172fl/fl controls

(top panel), transducin is translocated to rod IS and inner retinal region.

C. Retina cryosections from dark-adapted mice at PN25 showing predominant

transducin-Gαt localization to rod OS, in both genotypes. In addition, presence of

fl/fl transducin-Gαt in the IS of the Ift172 iCre mice but not the controls was-

observed. D. Retina cryosections from light-adapted mice at PN25, showing

fl/fl significantly higher transducin-Gαt staining in the IS of Ift172 iCre mice

compared to Ift172fl/fl littermate controls (white arrowhead). E. Relative

fluorescence intensity of the OS signal in relation to the total retina signal plotted

for Ift172fl/fliCre and Ift172fl/fl control in dark and light conditions at PN21 and

PN25.

Figure 7. GLI1 Localization. Photoreceptor localization of GLI1 (red) at PN21-

28, showing mislocalization in the photoreceptor inner segment at PN25.

RP1(green) marks the tranzision zone and nuclei are counterstained with

Hoechst (blue).

30 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Supplementary Figure 1. Additional Control Genotype OCT data. OCT data

of additional control genotypes (Ift172wt/wt Ift172fl/wtiCre, Ift172fl/fl ,) obtained at

one, two, and three months of age, with no signs of degeneration.

Supplementary Figure 2. Additional Cilia- associated Protein Localization

A. Immunostaining of Bbs9 (red), showing no differences in localization between

Ift172fl/fliCre and control mice. B. Immunostaining of IFT43 (red) ), showing no

differences in localization between Ift172fl/fliCre and control mice. Nuclei in A and

B are counterstained with Hoechst (blue) and RP1 is immunolabelled (green).

Supplementary Figure 3. Immunostaining of SMO, GLI2, or GLI3 (red), showing

no differences in localization between Ift172fl/fliCre and control mice. Nuclei in

are counterstained with Hoechst (blue) and RP1 is immunolabelled (green). Size

bar in the top panel represents 2µm and in the lower panel 30µm.

Abbreviations

Intraflagellar Transport (IFT), Outer segments (OS), Outer Nuclear Layer (ONL),

Inner Nuclear Layer (INL), Inner segments (IS), Retinal Pigment Epithelium

(RPE), Neural Retina (NR), Transmission Electron Microscopy (TEM), Light

Microscopy (LM), Peripherin 2 (Prph2), Guanyl-cyclase 1 (GC-1), Acetylated

Tubulin (AcTub), microliters (µL), millimolar (mM), micromolar (µM), Bovine

Serum Albumin (BSA), Paraformaldehyde (PFA), Phosphate Buffered Saline

(PBS), glioma-associated oncogene family member 1 (GLI1).

31 bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. bioRxiv preprint doi: https://doi.org/10.1101/210617; this version posted October 29, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.