Molecular Vision 2010; 16:1373-1381 <http://www.molvis.org/molvis/v16/a151>
Received 24 May 2010 | Accepted 13 July 2010 | Published 17 July 2010

Interaction of ciliary disease protein retinitis pigmentosa GTPase regulator with nephronophthisis-associated proteins in mammalian retinas

Carlos A. Murga-Zamalloa,1 Nimit J. Desai,1 Friedhelm Hildebrandt,2 Hemant Khanna1

1Department of Ophthalmology and Visual Sciences and Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI; 2Department of Pediatrics and Human Genetics, University of Michigan, Ann Arbor, MI

Correspondence to: Hemant Khanna, University of Massachusetts Medical School, Department of Ophthalmology, Biotech 5 Suite 250, 381 Plantation St, Worcester, MA, 01605; Phone: (508) 856-8991; FAX: (508) 856-1552; email: Hemant.Khanna@umassmed.edu

Abstract

Purpose: Retinitis pigmentosa GTPase regulator (RPGR) is a cilia-centrosomal protein that frequently mutates in X-linked retinal degeneration and associated disorders. RPGR interacts with multiple ciliary proteins in the retina. Perturbations in the assembly of RPGR complexes are associated with retinal degeneration. This study was undertaken to delineate the composition and dissection of RPGR complexes in mammalian retinas.

Methods: Immunoprecipitation of RPGR from ciliary fraction of bovine retina was performed, followed by mass spectrometry analysis. The glutathione S-transferase pull-down assay was performed to validate the interaction. Immunodepletion experiments were performed to dissect the partitioning of RPGR in different protein complexes in mammalian retinas.

Results: We found that RPGR associates with a ciliary protein nephrocystin-4 (nephroretinin; NPHP4) that is mutated in nephronophthisis (NPH) and RP (Senior-Løken syndrome). This association is abolished in the Rpgr-knockout mouse retina. The RCC1-like domain of RPGR interacts with the N-terminal 316 amino acids of NPHP4. In the retina, RPGR also associates with NPHP1, an NPHP4-interacting protein; RPGR interacts directly with amino acids 243–586 of NPHP1. We further show that, in the retina, RPGR associates with and is partitioned in at least two different complexes with NPHP-associated proteins, (i) NPHP1, NPHP2, and NPHP5, and (ii) NPHP4, NPHP6, and NPHP8.

Conclusions: RPGR may regulate some complexes with NPHP proteins in the mammalian retina. The disruption of these complexes may contribute to the pathogenesis of retinal degeneration in X-linked RP and associated ciliary diseases.

Introduction

Primary or sensory cilia are microtubule-based extensions of the plasma membrane that are found in almost all cell types [1,2]. They regulate diverse cellular processes, including signal transduction, sensory perception, and protein trafficking [3]. Assembly of cilia occurs by a conserved process called intraflagellar transport (IFT) [4]. During IFT, precursor moieties are assembled at the base of the cilia (basal bodies) and transported in anterograde and retrograde directions by the kinesin-II and dynein motor subunits. The transport is facilitated by the multiple IFT polypeptides. Defects in cilia assembly or function are associated with multi-systemic disorders, including Senior-Løken syndrome (SLSN), Bardet-Biedl syndrome, Joubert syndrome (JBTS), and Meckel-Gruber syndrome [5,6].

Retinitis pigmentosa (RP; OMIM 268000), a degenerative disease of the photoreceptors, is frequently associated with ciliary dysfunction. RP is a clinically and genetically heterogeneous group of disorders characterized by severe vision loss and blindness [7]. X-linked RP (XLRP) is a common form of RP, accounting for 10%–15% of all RP cases [8,9]. Clinical manifestations of XLRP usually include night blindness due to rod photoreceptor dysfunction, followed by loss of cone function and blindness by the fifth decade of life [8,9]. Some female carriers also exhibit severe retinal defects.

RP3, a major locus for XLRP, encodes for the retinitis pigmentosa GTPase regulator (RPGR) gene [10,11]. Mutations in RPGR account for 70%–80% of XLRP and ~20% of simplex RP cases [12,13]. Some RPGR patients exhibit extra-retinal phenotypes, including hearing dysfunction, sperm defects, respiratory infections, and primary cilia dyskinesia [1416]. The RPGR gene undergoes extensive alternative splicing and expresses multiple protein isoforms in the retina [1720]. Most RPGR isoforms contain a common N-terminal domain encoded by exons 1–15, which encompass an RCC1-like domain (RLD; encoded by exons 2–11). The originally described constitutive isoforms of RPGR are encoded by exons 1–19 and account for ~20% of XLRP patients with no known mutations in exons 16–19. Later studies revealed another isoform of RPGR that contains an alternative terminal exon ORF15 (encompasses part of intron 15). Mutations in exon ORF15 account for additional 50%–60% of XLRP patients.

The RLD of RPGR is thought to be the functional domain based on its homology to RCC1 and its involvement in interaction with other proteins. RPGR and RPGR-RLD predominantly localize to primary cilia and photoreceptor connecting cilium (CC) [19,21], which is a conduit for trafficking of proteins from the inner segment to the photosensitive outer segment [22]. An Rpgr-knockout (Rpgr-ko; deletion of exons 4–6 in RLD) mouse mutant [23], which was later shown to carry a hypomorphic allele of Rpgr [19], exhibits delayed onset photoreceptor degeneration and mistrafficking of cone opsins. In addition, two canine models of RPGR mutation have been reported [24]. These animal models exhibit disparate phenotypes depending upon the type of mutation. Despite extensive efforts, it is still not clear how RPGR regulates photoreceptor function or how mutations in RPGR cause retinal degenerative disease.

Identification of RPGR-interacting proteins has played a key role in understanding its function. RPGR interacts with several ciliary and transport proteins in the retina, including intraflagellar transport protein IFT88/Polaris and RPGR-interacting protein 1 (RPGRIP1) [19,25]. In addition, RPGR associates with NPHP proteins mutated in renal retinal syndromes, including SLSN and JBTS [2629]. For example, RPGR exists in complex with NPHP5 (or IQ domain containing calmodulin binding protein [IQCB1]; SLSN), centrosomal protein of 290 kDa (CEP290)/NPHP6 (Leber congenital amaurosis, SLSN, JBTS), and NPHP8/RPGRIP1-like (RPGRIP1L; mutated in JBTS and Meckel-Gruber syndrome) in the retina [3032]. Notably, hypomorphic mutations in NPHP6 and NPHP8, which are associated with relatively early-onset photoreceptor degeneration [2629,3335], disrupt their association with RPGR [30,31]. Based on these observations, we hypothesize that RPGR-containing multiprotein complexes play a key role in facilitating photoreceptor protein trafficking.

To elucidate the precise role of RPGR in regulating ciliary transport, it is important to identify and characterize the components of the RPGR-interaction network in the retina. Using co-immunoprecipitation (IP) and mass spectrometry analysis, here we report that RPGR binds to NPHP1 and NPHP4. Using serial immunodepletion, we also found that the RPGR-NPHP interaction network can be divided into at least two distinct complexes: the first complex constitutes NPHP1, NPHP2, and NPHP5, while the second complex consists of NPHP4, NPHP6, and NPHP8.

Methods

Animals

Animal experiments were performed in accordance with the guidelines of the Institute for Laboratory Animal Research (Guide for the Care and Use of Laboratory Animals). Prior approval for animal studies was obtained from the University of Michigan Animal Care and Use Committee. ARVO's guidelines were followed for the care and use of animals. The animals were fed ad libitum and were kept on a 12 h light and 12 h dark cycle. The background of the mice is C57BL6/J.

Antibodies and reagents

RPGR, NPHP5, and CEP290/NPHP6 antibodies have been previously described [19,30,32]. Anti-NPHP1 antibody was purchased from Abcam. Mouse anti-NPHP2/Inversin was procured from Novus Biologicals (Littleton, CO) and mouse anti-NPHP4 was obtained from Abnova (Taipei City, Taiwan).

Immunoprecipitation

Co-IP experiments were performed as described [19]. Briefly, bovine (Detroit, MI) or mouse retinal extracts were prepared in phosphate buffered saline (KH2PO4: 1.76 mM; Na2HPO4: 10 mM; KCl: 2.7 mM; NaCl: 138 mM; pH: 7.4) followed by incubation with the primary antibody or normal IgG (pre-immune bleed of rabbits) overnight at 4 °C. The protein-antibody complexes were then incubated with protein A or protein G agarose beads for 30 min at room temperature with gentle shaking. The beads were then washed with phosphate buffered saline with 1% (V/V) Triton X-100 precipitated proteins and subjected to tandem mass spectrometry. Dissection of complexes by immunodepletion (ID) was performed as described [36,37]. In brief, retina protein extract was subjected to immunoprecipitation using first primary antibody to deplete the associated complexes. The supernatant of the immunoprecipitation was then incubated with the second primary antibody and the precipitated proteins were analyzed by immunoblotting.

In vitro transcription/translation and glutathione S-transferase pull down

Proteins were synthesized in vitro using the Promega (Madison, WI) TnT Quick kit in the presence or absence of 35S-labeled methionine. Glutathione S-transferase (GST) or GST-RLD proteins were purified from E. coli and used in the GST pull-down assay, as described [19]. The 35S-labeled protein signal was analyzed by autoradiography with STORM 840 (GE Healthcare, Piscataway, NJ). RPGR-RLD was cloned into pGEX4T-2 (GE Healthcare, Piscataway, NJ). cDNA encoding NPHP1, NPHP4 and deleted domains were cloned into plasmid pcDNA3.1 (Invitrogen, Carlsbad, CA) [19].

Results

Retinitis pigmentosa GTPase regulator associates with nephrocystin-4

To identify RPGR-interacting proteins in mammalian retinas, we performed IP using anti-RPGR antibody followed by mass spectrometry analysis of the precipitated proteins. Our analysis revealed nephroretinin (NPHP4) as a strong interactor of RPGR. NPHP4 is a cilia-centrosomal protein mutated in nephronophthisis and SLSN [38,39]. Given that NPHP4 interacts with RPGRIP1 and NPHP8, and that it localizes to photoreceptor cilia [26,40], we focused on further analyzing the physiologic relevance of RPGR-NPHP4 interaction. Co-IP using anti-RPGR antibody followed by detection of the NPHP4 immunoreactive band further confirmed that RPGR and NPHP4 exist in the same complex in the retina (Figure 1A). Reverse IP using the anti-NPHP4 antibody also revealed RPGR-immunoreactive bands (data not shown). IP using IgG did not show any immunoreactivity. To examine whether RPGR-NPHP4 interaction is relevant to disease, we assessed the association of RPGR and NPHP4 in the Rpgr-ko mouse retina. As we have shown before, a deleted variant of RPGR is still expressed in the Rpgr-ko mouse retina and can be detected using the RPGRORF15 antibody [19]. Using co-IP, we showed that the mutant RPGR protein cannot associate with NPHP4 in the retina. (Figure 1B). These results suggest that the RLD of RPGR is involved in its interaction with NPHP4.

Retinitis pigmentosa GTPase regulator directly interacts with nephrocystin-4

To test whether RPGR-RLD interacts directly with NPHP4, we performed a GST pull-down assay using recombinant GST-RLD of RPGR and in vitro translated NPHP4 (full length and different domains [Figure 1C]). Using GST-RLD protein purified from E. coli and 35S-NPHP4, we found that the RLD directly interacts with NPHP4 (Figure 1D). No interaction was detected when GST alone was used in this experiment. We then sought to identify the domain of NPHP4 that interacts with RPGR-RLD. To this end, we synthesized different 35S-labeled domains of NPHP4 (Figure 1C) and used in the GST pull-down assay. As shown in Figure 1E, GST-RLD can interact directly with N-terminal 316 amino acids (N316). An interaction is also detected with the N1000- and N1186 domains of NPHP4.

Retinitis pigmentosa GTPase interacts with nephrocystin-1

It has been shown that NPHP4 interacts with NPHP1 (nephrocystin) [41], which is a ciliary protein mutated in nephronophthisis type 1 [42,43] and which localizes to photoreceptor CC [44]. As NPHP4’s RPGR-binding domain coincides with its NPHP1-binding domain [41], we tested whether RPGR also interacts with NPHP1. Using bovine retinal extract, we found that the anti-RPGR antibody could immunoprecipitate NPHP1 (Figure 2A). Reverse IP using anti-NPHP1 antibody also revealed RPGR-immunoreactive bands (Figure 2B). We then examined the interaction of RPGR with NPHP1 in vitro. Using 35S-labeled NPHP1 (full-length and different domains [Figure 2C]) and GST-RLD fusion protein, we showed that full-length NPHP1 directly binds to RLD. No interaction is detected with GST alone (Figure 2D). We also synthesized 35S-labeled NPHP1 domains (Figure 2E) and used them in the GST-pull down assay. We found that amino acids 243–586 of NPHP1 were sufficient bind to RPGR-RLD (Figure 2F).

Retinitis pigmentosa GTPase regulator forms distinct complexes with nephrocystin proteins

All NPHP proteins identified to date localize to cilia and centrosomes. In fact, NPHP4 and NPHP1, identified in this study as binding partners of RPGR, have also been localized to photoreceptor CC [40,44,45]. As RPGR is a cilia-centrosomal protein and associates with the majority of NPHP proteins in the retina (NPHP1, NPHP2, NPHP4, NPHP5, NPHP6, NPHP8, and NPHP11) [46], we sought to determine how RPGR might be partitioned in the NPHP complexes. We first performed serial ID of NPHP4 or NPHP1 to immunodeplete the fraction of RPGR that is in complex with these proteins. The remaining RPGR in the supernatant was immunoprecipitated using anti-RPGR antibody and both precipitates were tested for the presence or absence of RPGR or other NPHP proteins. This strategy has been described previously [36,37] and is schematically represented in Figure 3A. We first tested that the anti-NPHP4 antibody is efficient in immunodepleting NPHP4. Using anti-NPHP4 antibody, we found that a majority (>90%) of NPHP4 is immunodepleted from the bovine retinal extracts (Figure 3B; sup represents supernatant). Using the NPHP4-immunodepleted supernatant (NPHP4-ID sup), we performed IP using anti-RPGR antibody and analyzed the precipitate for the presence of other NPHP proteins. We found that the remaining RPGR still associated with NPHP1, NPHP2, and NPHP5. However, NPHP6 and NPHP8 were no longer detected in complex with RPGR in the sample depleted of NPHP4 complexes (Figure 3C). Similar results were obtained when NPHP2 and NPHP5 were immunodepleted from bovine retinal extracts (data not shown). On the other hand, after ID of NPHP1 (Figure 4A), RPGR still associated with NPHP4, NPHP6, and NPHP8, but not with NPHP2 or NPHP5 (Figure 4B). Taken together, our data indicate that there are at least two different RPGR-NPHP complexes: (i) RPGR-NPHP1-NPHP2-NPHP5 and (ii) RPGR-NPHP4-NPHP6-NPHP8.

Retinitis pigmentosa GTPase regulator-nephrocystin-4 complex is distinct from nephrocystin-1-nephrocystin-4 complex

We have found that the previously reported NPHP1-binding domain of NPHP4 coincides with its RPGR-binding domain. Hence, we hypothesized that RPGR-NPHP4 complexes are distinct from NPHP4-NPHP1 complexes. As shown above, after serial ID of NPHP1, NPHP4 can still associate with RPGR. We then examined whether ID of RPGR abolishes the NPHP1-NPHP4 complexes. Our analysis revealed that even when the majority of RPGR was depleted, NPHP4 still associated with NPHP1 (Figure 5A-C). These results indicate that there are potentially RPGR-independent NPHP1-NPHP4 complexes in the retina.

Discussion

Ciliopathies are accompanied by overlapping as well as somewhat distinct phenotypes, although they arise from a common defect in ciliary functions [47]. Nephronophthisis, a ciliopathy characterized by fibrocystic renal disease, is the leading genetic cause of end stage renal disease in children and young adults [6]. The syndromic disorder SLSN is characterized by nephronophthisis and retinal degeneration. To date, 10 NPHP genes have been identified as causing NPHP or SLSN of variable severities (NPHP1–9 and NPHP11) [26,27,29,32,38,42,43,4852]. All NPHP proteins localize to primary cilia and photoreceptor sensory cilia [5,6,53]. Degeneration of photoreceptors is a commonly observed phenotype in ciliopathies. However, the mechanism of the progression and pathogenesis of photoreceptor degeneration due to mutations in ciliary proteins is not completely understood. Here, we describe the network of ciliary complexes of RPGR and NPHP proteins in mammalian retina that may be altered in disease condition. Our work has revealed important findings. First, we described NPHP4 and NPHP1 as novel RPGR-interacting proteins in the retina. Second, we showed that RPGR and NPHP proteins form distinct complexes in the retina. Third, we demonstrated that the RLD of RPGR might mediate RPGR-NPHP interactions. Significantly, our results also demonstrated that RPGR-NPHP interactions are altered in the mouse mutant of Rpgr. As RPGR and its interacting proteins have been shown to play a key role in regulating protein trafficking in photoreceptors, our data point to a role of RPGR-NPHP protein complexes in cilia-dependent transport and associated disease.

It has been shown that NPHP1 and NPHP4 interact with each other in renal epithelial cells [41]. Although RPGR interacts with both NPHP1 and NPHP4, our data suggest that RPGR-NPHP4 and RPGR-NPHP1 complexes may be distinct from the NPHP1-NPHP4 complexes in the retina. We have recently shown that knockdown of RPGR results in shorter cilia in the Kupffer’s vesicles of zebrafish [54]. Interestingly, Mollet et al. [41] reported that knockdown of NPHP1 or NPHP4 also results in shorter cilia. Cumulatively, these studies provide further evidence for a functional overlap between RPGR and NPHP proteins in regulating photoreceptor function.

What is the function of RPGR-NPHP complexes? As photoreceptors rely almost completely on the primary cilium for microtubule-based intersegmental transport of proteins involved in phototransduction and outer segment renewal [22,5558], slight perturbations in protein trafficking can lead to photoreceptor degeneration and blindness [30,5961]. Our data suggest that distinct protein complexes may facilitate the binding, trafficking, and release of the cargo moieties at different steps of the protein transport pathways. A communication or “hand over” mechanism may exist between the distinct protein complexes for efficient cargo transfer and delivery. RPGR seems to play a key role in regulating a subset of these complexes in photoreceptors. The degree of disruption of individual complexes at particular steps of the protein transport pathways may determine the severity of associated disease. Further studies are required to assess the physiologic relevance and functionality of these complexes in protein trafficking in photoreceptors.

Ciliopathies exhibit clinical heterogeneity even within single families. The present studies suggest the involvement of genetic modifiers of the disease phenotype. We recently showed that mutations in NPHP8, which alter its interaction with RPGR, act as modifiers of the expressivity of the retinal degeneration phenotype in ciliopathy patients [31]. More recently, it has been reported that mutations in Abelson helper integration site 1 protein homolog (AHI1), which interacts with NPHP1 [62], act as modifier of retinal degeneration phenotype in nephronophthisis [63]. Taken together, the RPGR-interactome dissected in this study provides clues to further analyze the molecular mechanisms underlying the genetic and clinical heterogeneity associated with ciliopathies.

Acknowledgments

We thank Dr. Anand Swaroop, Dr. Edgar A. Otto, and Dr. Suresh Patil for critical discussions. FH is an investigator of the Howard Hughes Medical Institute, a Doris Duke Distinguished Clinical Scientist, and Frederick G.L. Huetwell Professor. This research is funded by grants from the National Institutes of Health (R01EY007961, R01DK1069274, R01DK1068306, R01DK064614), Foundation Fighting Blindness, Midwest Eye Banks and Transplantation Center, and Rare Disease Initiative, University of Michigan. We also acknowledge the Michigan Proteome Consortium, the Michigan Economic Development Corporation, and Michigan Technology Tri-Corridor for mass spectrometric analysis.

References

  1. Sorokin S. Centrioles and the formation of rudimentary cilia by fibroblasts and smooth muscle cells. J Cell Biol. 1962; 15:363-77. [PMID: 13978319]
  2. Sorokin SP. Centriole formation and ciliogenesis. Aspen Emphysema Conf. 1968; 11:213-6. [PMID: 5751848]
  3. Pazour GJ, Witman GB. The vertebrate primary cilium is a sensory organelle. Curr Opin Cell Biol. 2003; 15:105-10. [PMID: 12517711]
  4. Rosenbaum J. Intraflagellar transport. Curr Biol. 2002; 12:R125 [PMID: 11864582]
  5. Badano JL, Mitsuma N, Beales PL, Katsanis N. The ciliopathies: an emerging class of human genetic disorders. Annu Rev Genomics Hum Genet. 2006; 7:125-48. [PMID: 16722803]
  6. Hildebrandt F, Otto E. Cilia and centrosomes: a unifying pathogenic concept for cystic kidney disease? Nat Rev Genet. 2005; 6:928-40. [PMID: 16341073]
  7. Heckenlively JR, Yoser SL, Friedman LH, Oversier JJ. Clinical findings and common symptoms in retinitis pigmentosa. Am J Ophthalmol. 1988; 105:504-11. [PMID: 3259404]
  8. Bird AC. X-linked retinitis pigmentosa. Br J Ophthalmol. 1975; 59:177-99. [PMID: 1138842]
  9. Fishman GA, Farber MD, Derlacki DJ. X-linked retinitis pigmentosa. Profile of clinical findings. Arch Ophthalmol. 1988; 106:369-75. [PMID: 3257866]
  10. Meindl A, Dry K, Herrmann K, Manson F, Ciccodicola A, Edgar A, Carvalho MR, Achatz H, Hellebrand H, Lennon A, Migliaccio C, Porter K, Zrenner E, Bird A, Jay M, Lorenz B, Wittwer B, D'Urso M, Meitinger T, Wright A. A gene (RPGR) with homology to the RCC1 guanine nucleotide exchange factor is mutated in X-linked retinitis pigmentosa (RP3). Nat Genet. 1996; 13:35-42. [PMID: 8673101]
  11. Roepman R, van Duijnhoven G, Rosenberg T, Pinckers AJ, Bleeker-Wagemakers LM, Bergen AA, Post J, Beck A, Reinhardt R, Ropers HH, Cremers FP, Berger W. Positional cloning of the gene for X-linked retinitis pigmentosa 3: homology with the guanine-nucleotide-exchange factor RCC1. Hum Mol Genet. 1996; 5:1035-41. [PMID: 8817343]
  12. Breuer DK, Yashar BM, Filippova E, Hiriyanna S, Lyons RH, Mears AJ, Asaye B, Acar C, Vervoort R, Wright AF, Musarella MA, Wheeler P, MacDonald I, Iannaccone A, Birch D, Hoffman DR, Fishman GA, Heckenlively JR, Jacobson SG, Sieving PA, Swaroop A. A comprehensive mutation analysis of RP2 and RPGR in a North American cohort of families with X-linked retinitis pigmentosa. Am J Hum Genet. 2002; 70:1545-54. [PMID: 11992260]
  13. Shu X, Black GC, Rice JM, Hart-Holden N, Jones A, O'Grady A, Ramsden S, Wright AF. RPGR mutation analysis and disease: an update. Hum Mutat. 2007; 28:322-8. [PMID: 17195164]
  14. Iannaccone A, Wang X, Jablonski MM, Kuo SF, Baldi A, Cosgrove D, Morton CC, Swaroop A. Increasing evidence for syndromic phenotypes associated with RPGR mutations. Am J Ophthalmol. 2004; 137:785-6. [PMID: 15059739]
  15. Moore A, Escudier E, Roger G, Tamalet A, Pelosse B, Marlin S, Clément A, Geremek M, Delaisi B, Bridoux AM, Coste A, Witt M, Duriez B, Amselem S. RPGR is mutated in patients with a complex X linked phenotype combining primary ciliary dyskinesia and retinitis pigmentosa. J Med Genet. 2006; 43:326-33. [PMID: 16055928]
  16. van Dorp DB, Wright AF, Carothers AD, Bleeker-Wagemakers EM. A family with RP3 type of X-linked retinitis pigmentosa: an association with ciliary abnormalities. Hum Genet. 1992; 88:331-4. [PMID: 1733835]
  17. He S, Parapuram SK, Hurd TW, Behnam B, Margolis B, Swaroop A, Khanna H. Retinitis Pigmentosa GTPase Regulator (RPGR) protein isoforms in mammalian retina: insights into X-linked Retinitis Pigmentosa and associated ciliopathies. Vision Res. 2008; 48:366-76. [PMID: 17904189]
  18. Hong DH, Li T. Complex expression pattern of RPGR reveals a role for purine-rich exonic splicing enhancers. Invest Ophthalmol Vis Sci. 2002; 43:3373-82. [PMID: 12407146]
  19. Khanna H, Hurd TW, Lillo C, Shu X, Parapuram SK, He S, Akimoto M, Wright AF, Margolis B, Williams DS, Swaroop A. RPGR-ORF15, which is mutated in retinitis pigmentosa, associates with SMC1, SMC3, and microtubule transport proteins. J Biol Chem. 2005; 280:33580-7. [PMID: 16043481]
  20. Kirschner R, Rosenberg T, Schultz-Heienbrok R, Lenzner S, Feil S, Roepman R, Cremers FP, Ropers HH, Berger W. RPGR transcription studies in mouse and human tissues reveal a retina-specific isoform that is disrupted in a patient with X-linked retinitis pigmentosa. Hum Mol Genet. 1999; 8:1571-8. [PMID: 10401007]
  21. Hong DH, Pawlyk B, Sokolov M, Strissel KJ, Yang J, Tulloch B, Wright AF, Arshavsky VY, Li T. RPGR isoforms in photoreceptor connecting cilia and the transitional zone of motile cilia. Invest Ophthalmol Vis Sci. 2003; 44:2413-21. [PMID: 12766038]
  22. Young RW. Passage of newly formed protein through the connecting cilium of retina rods in the frog. J Ultrastruct Res. 1968; 23:462-73. [PMID: 5692302]
  23. Hong DH, Pawlyk BS, Shang J, Sandberg MA, Berson EL, Li T. A retinitis pigmentosa GTPase regulator (RPGR)-deficient mouse model for X-linked retinitis pigmentosa (RP3). Proc Natl Acad Sci USA. 2000; 97:3649-54. [PMID: 10725384]
  24. Zhang Q, Acland GM, Wu WX, Johnson JL, Pearce-Kelling S, Tulloch B, Vervoort R, Wright AF, Aguirre GD. Different RPGR exon ORF15 mutations in Canids provide insights into photoreceptor cell degeneration. Hum Mol Genet. 2002; 11:993-1003. [PMID: 11978759]
  25. Hong DH, Yue G, Adamian M, Li T. Retinitis pigmentosa GTPase regulator (RPGRr)-interacting protein is stably associated with the photoreceptor ciliary axoneme and anchors RPGR to the connecting cilium. J Biol Chem. 2001; 276:12091-9. [PMID: 11104772]
  26. Arts HH, Doherty D, van Beersum SE, Parisi MA, Letteboer SJ, Gorden NT, Peters TA, Märker T, Voesenek K, Kartono A, Ozyurek H, Farin FM, Kroes HY, Wolfrum U, Brunner HG, Cremers FP, Glass IA, Knoers NV, Roepman R. Mutations in the gene encoding the basal body protein RPGRIP1L, a nephrocystin-4 interactor, cause Joubert syndrome. Nat Genet. 2007; 39:882-8. [PMID: 17558407]
  27. Delous M, Baala L, Salomon R, Laclef C, Vierkotten J, Tory K, Golzio C, Lacoste T, Besse L, Ozilou C, Moutkine I, Hellman NE, Anselme I, Silbermann F, Vesque C, Gerhardt C, Rattenberry E, Wolf MT, Gubler MC, Martinovic J, Encha-Razavi F, Boddaert N, Gonzales M, Macher MA, Nivet H, Champion G, Berthélémé JP, Niaudet P, McDonald F, Hildebrandt F, Johnson CA, Vekemans M, Antignac C, Rüther U, Schneider-Maunoury S, Attié-Bitach T, Saunier S. The ciliary gene RPGRIP1L is mutated in cerebello-oculo-renal syndrome (Joubert syndrome type B) and Meckel syndrome. Nat Genet. 2007; 39:875-81. [PMID: 17558409]
  28. den Hollander AI, Koenekoop RK, Yzer S, Lopez I, Arends ML, Voesenek KE, Zonneveld MN, Strom TM, Meitinger T, Brunner HG, Hoyng CB, van den Born LI, Rohrschneider K, Cremers FP. Mutations in the CEP290 (NPHP6) gene are a frequent cause of Leber congenital amaurosis. Am J Hum Genet. 2006; 79:556-61. [PMID: 16909394]
  29. Sayer JA, Otto EA, O'Toole JF, Nurnberg G, Kennedy MA, Becker C, Hennies HC, Helou J, Attanasio M, Fausett BV, Utsch B, Khanna H, Liu Y, Drummond I, Kawakami I, Kusakabe T, Tsuda M, Ma L, Lee H, Larson RG, Allen SJ, Wilkinson CJ, Nigg EA, Shou C, Lillo C, Williams DS, Hoppe B, Kemper MJ, Neuhaus T, Parisi MA, Glass IA, Petry M, Kispert A, Gloy J, Ganner A, Walz G, Zhu X, Goldman D, Nurnberg P, Swaroop A, Leroux MR, Hildebrandt F. The centrosomal protein nephrocystin-6 is mutated in Joubert syndrome and activates transcription factor ATF4. Nat Genet. 2006; 38:674-81. [PMID: 16682973]
  30. Chang B, Khanna H, Hawes N, Jimeno D, He S, Lillo C, Parapuram SK, Cheng H, Scott A, Hurd RE, Sayer JA, Otto EA, Attanasio M, O'Toole JF, Jin G, Shou C, Hildebrandt F, Williams DS, Heckenlively JR, Swaroop A. In-frame deletion in a novel centrosomal/ciliary protein CEP290/NPHP6 perturbs its interaction with RPGR and results in early-onset retinal degeneration in the rd16 mouse. Hum Mol Genet. 2006; 15:1847-57. [PMID: 16632484]
  31. Khanna H, Davis EE, Murga-Zamalloa CA, Estrada-Cuzcano A, Lopez I, den Hollander AI, Zonneveld MN, Othman MI, Waseem N, Chakarova CF, Maubaret C, Diaz-Font A, Macdonald I, Muzny DM, Wheeler DA, Morgan M, Lewis LR, Logan CV, Tan PL, Beer MA, Inglehearn CF, Lewis RA, Jacobson SG, Bergmann C, Beales PL, Attié-Bitach T, Johnson CA, Otto EA, Bhattacharya SS, Hildebrandt F, Gibbs RA, Koenekoop RK, Swaroop A, Katsanis N. A common allele in RPGRIP1L is a modifier of retinal degeneration in ciliopathies. Nat Genet. 2009; 41:739-45. [PMID: 19430481]
  32. Otto EA, Loeys B, Khanna H, Hellemans J, Sudbrak R, Fan S, Muerb U, O'Toole JF, Helou J, Attanasio M, Utsch B, Sayer JA, Lillo C, Jimeno D, Coucke P, De Paepe A, Reinhardt R, Klages S, Tsuda M, Kawakami I, Kusakabe T, Omran H, Imm A, Tippens M, Raymond PA, Hill J, Beales P, He S, Kispert A, Margolis B, Williams DS, Swaroop A, Hildebrandt F. Nephrocystin-5, a ciliary IQ domain protein, is mutated in Senior-Loken syndrome and interacts with RPGR and calmodulin. Nat Genet. 2005; 37:282-8. [PMID: 15723066]
  33. Dryja TP, Adams SM, Grimsby JL, McGee TL, Hong DH, Li T, Andréasson S, Berson EL. Null RPGRIP1 alleles in patients with Leber congenital amaurosis. Am J Hum Genet. 2001; 68:1295-8. [PMID: 11283794]
  34. Leitch CC, Zaghloul NA, Davis EE, Stoetzel C, Diaz-Font A, Rix S, Alfadhel M, Lewis RA, Eyaid W, Banin E, Dollfus H, Beales PL, Badano JL, Katsanis N. Hypomorphic mutations in syndromic encephalocele genes are associated with Bardet-Biedl syndrome. Nat Genet. 2008; 40:443-8. [PMID: 18327255]
  35. Valente EM, Silhavy JL, Brancati F, Barrano G, Krishnaswami SR, Castori M, Lancaster MA, Boltshauser E, Boccone L, Al-Gazali L, Fazzi E, Signorini S, Louie CM, Bellacchio E. International Joubert Syndrome Related Disorders Study Group, Bertini E, Dallapiccola B, Gleeson JG. Mutations in CEP290, which encodes a centrosomal protein, cause pleiotropic forms of Joubert syndrome. Nat Genet. 2006; 38:623-5. [PMID: 16682970]
  36. Murga-Zamalloa CA, Swaroop A, Khanna H. In: Andersen RE, Hollyfield, J.G., Lavail, M.M., editors. Retinal Degenerative Diseases: Laboratory and Therapeutic Investigations: Springer; 2010.
  37. Rodriguez P, Bonte E, Krijgsveld J, Kolodziej KE, Guyot B, Heck AJ, Vyas P, de Boer E, Grosveld F, Strouboulis J. GATA-1 forms distinct activating and repressive complexes in erythroid cells. EMBO J. 2005; 24:2354-66. [PMID: 15920471]
  38. Otto E, Hoefele J, Ruf R, Mueller AM, Hiller KS, Wolf MT, Schuermann MJ, Becker A, Birkenhäger R, Sudbrak R, Hennies HC, Nürnberg P, Hildebrandt F. A gene mutated in nephronophthisis and retinitis pigmentosa encodes a novel protein, nephroretinin, conserved in evolution. Am J Hum Genet. 2002; 71:1161-7. [PMID: 12205563]
  39. Mollet G, Salomon R, Gribouval O, Silbermann F, Bacq D, Landthaler G, Milford D, Nayir A, Rizzoni G, Antignac C, Saunier S. The gene mutated in juvenile nephronophthisis type 4 encodes a novel protein that interacts with nephrocystin. Nat Genet. 2002; 32:300-5. [PMID: 12244321]
  40. Roepman R, Letteboer SJ, Arts HH, van Beersum SE, Lu X, Krieger E, Ferreira PA, Cremers FP. Interaction of nephrocystin-4 and RPGRIP1 is disrupted by nephronophthisis or Leber congenital amaurosis-associated mutations. Proc Natl Acad Sci USA. 2005; 102:18520-5. [PMID: 16339905]
  41. Mollet G, Silbermann F, Delous M, Salomon R, Antignac C, Saunier S. Characterization of the nephrocystin/nephrocystin-4 complex and subcellular localization of nephrocystin-4 to primary cilia and centrosomes. Hum Mol Genet. 2005; 14:645-56. [PMID: 15661758]
  42. Hildebrandt F, Otto E, Rensing C, Nothwang HG, Vollmer M, Adolphs J, Hanusch H, Brandis M. A novel gene encoding an SH3 domain protein is mutated in nephronophthisis type 1. Nat Genet. 1997; 17:149-53. [PMID: 9326933]
  43. Saunier S, Calado J, Heilig R, Silbermann F, Benessy F, Morin G, Konrad M, Broyer M, Gubler MC, Weissenbach J, Antignac C. A novel gene that encodes a protein with a putative src homology 3 domain is a candidate gene for familial juvenile nephronophthisis. Hum Mol Genet. 1997; 6:2317-23. [PMID: 9361039]
  44. Fliegauf M, Horvath J, von Schnakenburg C, Olbrich H, Müller D, Thumfart J, Schermer B, Pazour GJ, Neumann HP, Zentgraf H, Benzing T, Omran H. Nephrocystin specifically localizes to the transition zone of renal and respiratory cilia and photoreceptor connecting cilia. J Am Soc Nephrol. 2006; 17:2424-33. [PMID: 16885411]
  45. Jiang ST, Chiou YY, Wang E, Chien YL, Ho HH, Tsai FJ, Lin CY, Tsai SP, Li H. Essential role of nephrocystin in photoreceptor intraflagellar transport in mouse. Hum Mol Genet. 2009; 18:1566-77. [PMID: 19208653]
  46. Murga-Zamalloa CA, Swaroop A, Khanna H. RPGR-containing protein complexes in syndromic and non-syndromic retinal degeneration due to ciliary dysfunction. J Genet. 2009; 88:399-407. [PMID: 20090203]
  47. Quinlan RJ, Tobin JL, Beales PL. Modeling ciliopathies: Primary cilia in development and disease. Curr Top Dev Biol. 2008; 84:249-310. [PMID: 19186246]
  48. Attanasio M, Uhlenhaut NH, Sousa VH, O'Toole JF, Otto E, Anlag K, Klugmann C, Treier AC, Helou J, Sayer JA, Seelow D, Nürnberg G, Becker C, Chudley AE, Nürnberg P, Hildebrandt F, Treier M. Loss of GLIS2 causes nephronophthisis in humans and mice by increased apoptosis and fibrosis. Nat Genet. 2007; 39:1018-24. [PMID: 17618285]
  49. Olbrich H, Fliegauf M, Hoefele J, Kispert A, Otto E, Volz A, Wolf MT, Sasmaz G, Trauer U, Reinhardt R, Sudbrak R, Antignac C, Gretz N, Walz G, Schermer B, Benzing T, Hildebrandt F, Omran H. Mutations in a novel gene, NPHP3, cause adolescent nephronophthisis, tapeto-retinal degeneration and hepatic fibrosis. Nat Genet. 2003; 34:455-9. [PMID: 12872122]
  50. Otto EA, Schermer B, Obara T, O'Toole JF, Hiller KS, Mueller AM, Ruf RG, Hoefele J, Beekmann F, Landau D, Foreman JW, Goodship JA, Strachan T, Kispert A, Wolf MT, Gagnadoux MF, Nivet H, Antignac C, Walz G, Drummond IA, Benzing T, Hildebrandt F. Mutations in INVS encoding inversin cause nephronophthisis type 2, linking renal cystic disease to the function of primary cilia and left-right axis determination. Nat Genet. 2003; 34:413-20. [PMID: 12872123]
  51. Otto EA, Tory K, Attanasio M, Zhou W, Chaki M, Paruchuri Y, Wise EL, Wolf MT, Utsch B, Becker C, Nürnberg G, Nürnberg P, Nayir A, Saunier S, Antignac C, Hildebrandt F. Hypomorphic mutations in meckelin (MKS3/TMEM67) cause nephronophthisis with liver fibrosis (NPHP11). J Med Genet. 2009; 46:663-70. [PMID: 19508969]
  52. Otto EA, Trapp ML, Schultheiss UT, Helou J, Quarmby LM, Hildebrandt F. NEK8 mutations affect ciliary and centrosomal localization and may cause nephronophthisis. J Am Soc Nephrol. 2008; 19:587-92. [PMID: 18199800]
  53. Hildebrandt F, Attanasio M, Otto E. Nephronophthisis: disease mechanisms of a ciliopathy. J Am Soc Nephrol. 2009; 20:23-35. [PMID: 19118152]
  54. Ghosh AK, Murga-Zamalloa CA, Chan L, Hitchcock PF, Swaroop A, Khanna H. Human retinopathy-associated ciliary protein retinitis pigmentosa GTPase regulator mediates cilia-dependent vertebrate development. Hum Mol Genet. 2010; 19:90-8. [PMID: 19815619]
  55. Young RW, Droz B. The renewal of protein in retinal rods and cones. J Cell Biol. 1968; 39:169-84. [PMID: 5692679]
  56. Besharse JC, Baker SA, Luby-Phelps K, Pazour GJ. Photoreceptor intersegmental transport and retinal degeneration: a conserved pathway common to motile and sensory cilia. Adv Exp Med Biol. 2003; 533:157-64. [PMID: 15180260]
  57. Besharse JC. The Retina: A Model for Cell Biological Studies Part I. New York: Academic Press; 1986.
  58. Nair KS, Hanson SM, Kennedy MJ, Hurley JB, Gurevich VV, Slepak VZ. Direct binding of visual arrestin to microtubules determines the differential subcellular localization of its splice variants in rod photoreceptors. J Biol Chem. 2004; 279:41240-8. [PMID: 15272005]
  59. Deretic D, Schmerl S, Hargrave PA, Arendt A, McDowell JH. Regulation of sorting and post-Golgi trafficking of rhodopsin by its C-terminal sequence QVS(A)PA. Proc Natl Acad Sci USA. 1998; 95:10620-5. [PMID: 9724753]
  60. Deretic D, Williams AH, Ransom N, Morel V, Hargrave PA, Arendt A. Rhodopsin C-terminus, the site of mutations causing retinal disease, regulates trafficking by binding to ADP-ribosylation factor 4 (ARF4). Proc Natl Acad Sci USA. 2005; 102:3301-6. [PMID: 15728366]
  61. Marszalek JR, Liu X, Roberts EA, Chui D, Marth JD, Williams DS, Goldstein LS. Genetic evidence for selective transport of opsin and arrestin by kinesin-II in mammalian photoreceptors. Cell. 2000; 102:175-87. [PMID: 10943838]
  62. Eley L, Gabrielides C, Adams M, Johnson CA, Hildebrandt F, Sayer JA. Jouberin localizes to collecting ducts and interacts with nephrocystin-1. Kidney Int. 2008; 74:1139-49. [PMID: 18633336]
  63. Louie CM, Caridi G, Lopes VS, Brancati F, Kispert A, Lancaster MA, Schlossman AM, Otto EA, Leitges M, Gröne HJ, Lopez I, Gudiseva HV, O'Toole JF, Vallespin E, Ayyagari R, Ayuso C, Cremers FP, den Hollander AI, Koenekoop RK, Dallapiccola B, Ghiggeri GM, Hildebrandt F, Valente EM, Williams DS, Gleeson JG. AHI1 is required for photoreceptor outer segment development and is a modifier for retinal degeneration in nephronophthisis. Nat Genet. 2010; 42:175-80. [PMID: 20081859]