Entry - *312610 - RETINITIS PIGMENTOSA GTPase REGULATOR; RPGR - OMIM
 
* 312610

RETINITIS PIGMENTOSA GTPase REGULATOR; RPGR


HGNC Approved Gene Symbol: RPGR

Cytogenetic location: Xp11.4     Genomic coordinates (GRCh38): X:38,269,163-38,327,509 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xp11.4 Cone-rod dystrophy, X-linked, 1 304020 XLR 3
Macular degeneration, X-linked atrophic 300834 XLR 3
Retinitis pigmentosa 3 300029 XL 3
Retinitis pigmentosa, X-linked, and sinorespiratory infections, with or without deafness 300455 XL 3

TEXT

Cloning and Expression

Meindl et al. (1996) isolated and sequenced cosmids from the region of the microdeletions in patients with retinitis pigmentosa-3 (300029) and used these cosmids to make exon predictions. They thus identified a gene, provisionally named RPGR (retinitis pigmentosa GTPase regulator), which gives rise to a ubiquitously expressed 29-kb transcript. The predicted 90-kD RPGR protein contains in its N-terminal half a tandem repeat structure highly similar to the regulator of chromosome condensation (RCC1; 179710), which regulates the GTPase RAN (601179). Meindl et al. (1996) identified 8 potential asparagine-linked glycosylation sites along the N-terminal two-thirds of the predicted RPGR protein. The C terminus of the protein contains a cluster of basic residues followed by a consensus isoprenylation site. The authors noted that confirmation of the isoprenylation of this site would establish a novel means of membrane anchorage for a GTPase regulator.

Roepman et al. (1996) used a cosmid spanning a microdeletion in an RP3 patient to screen cDNA libraries. They then isolated additional cosmids that flanked the microdeletion region. Shotgun cosmid sequencing enabled them to sequence 32,895 bp of DNA. Computer-assisted analysis of this sequence predicted numerous additional exons which were confirmed by cDNA cloning. Sequence comparisons revealed that the deduced product of the RPGR gene showed strong similarity with RCC1.

Kirschner et al. (1999) studied the expression of the RPGR gene by Northern blot hybridization, cDNA library screening, and RT-PCR in various organs of mouse and human and identified at least 12 alternatively spliced isoforms. Some of the transcripts are tissue-specific and contain novel exons, which elongate or truncate the previously reported open reading frame of the mouse and human RPGR gene. Kirschner et al. (1999) identified a new exon, designated exon 15A by them, which is expressed exclusively in human retina and mouse eye and contains a premature stop codon. The deduced polypeptide lacked 169 amino acids from the C terminus of the ubiquitously expressed variant, including an isoprenylation site.

Kirschner et al. (2001) compared the genomic sequence of the human and mouse RPGR genes. Each spans a region of nearly 59 kb, and all previously identified exons are conserved between the 2 species. A sequence comparison identified 28 conserved sequence elements in introns, upstream of exon 1, within the promoter region, and downstream of the most 3-prime exon. Some of the intronic conserved sequence elements flank tissue-specific exons and therefore may represent important regulatory elements for alternative splicing. Comparative Northern blot hybridization of ubiquitous and tissue-specific RPGR probes identified high molecular mass transcripts with similar expression patterns in both human and mouse. These transcripts range from 6 to 15 kb and suggest the presence of additional transcribed sequences within RPGR.

Vervoort et al. (2000) sequenced a 172-kb region containing the entire RPGR gene. Analysis of the sequence disclosed a novel 3-prime terminal exon that was mutated in 60% of XLRP patients examined. This exon encodes 567 amino acids, with a repetitive domain rich in glutamic acid residues. The sequence is conserved in the mouse, bovine, and Fugu rubripes genes. It is preferentially expressed in mouse and bovine retina, further supporting its importance for retinal function. In addition to the 19 exons previously reported, Vervoort et al. (2000) found 5 additional exons: 15b1, 15b2, 15a, ORF14, and ORF15. Exons 15b1 and 15b2 are 2 overlapping exons within intron 15 that use alternative acceptor splice sites and the same donor site. Their inclusion is predicted to result in premature termination of translation. Exon 15a, which is a third internal exon within intron 15 and also encodes a premature stop codon, is identical with exon 15a reported previously (Kirschner et al., 1999). Exon ORF14 corresponds to the mouse exon 14-14a-15 (Kirschner et al., 1999); its inclusion does not disrupt the reading frame. Exon ORF15 is a large 3-prime terminal exon consisting of exon 15 and extending into part of intron 15. The predicted exon ORF15 protein domain contains an unusual region of low sequence complexity with high glutamic acid and glycine content (Plaid domain).

By RT-PCR analysis of human retina cDNA, Neidhardt et al. (2007) identified a novel exon in intron 9, designated exon 9A, of the RPGR gene. The novel exon starts 418 bp downstream of the splice donor site of exon 9 and contains 136 nucleotides. Exon 9A is present in a novel RPGR splice variant that truncates the RCC1 homologous protein domain of RPGR by 48 amino acids, yielding a 353-residue protein with a molecular mass of about 45 kD. There was strong expression of the 9A variant predominantly in the inner segment of human cones with a weaker signal in rods. Immunoprecipitation studies showed that the 9A variant binds distinct RPGRIP1 (605446) variants, suggesting functional relevance. The expression of mRNA containing exon 9A was quantified to approximately 4% of RPGR in normal individuals.

Ghosh et al. (2010) noted that there are 2 major isoforms of RPGR detected in the retina: RPGR(1-19), which has 19 exons and 815 amino acids, and RPGR(1-ORF15), which has 15 exons plus part of intron 15 and 1,152 amino acids. Both isoforms share exons 1 through 15 and N-terminal RCC1-like domain (RLD), whereas RPGR(1-ORF15) has a glu-gly-rich C-terminal domain.

Using immunogold electron microscopy with mouse and human retina, Khanna et al. (2005) found that, in addition to axoneme, RPGR-ORF15 localized to basal bodies of photoreceptor connecting cilium. It also colocalized with acetylated alpha-tubulin (see 602529) at the tip and tail axoneme of mouse sperm flagella and at primary cilia of MDCK canine kidney cells.


Gene Function

Boylan and Wright (2000) and Roepman et al. (2000) identified the RPGRIP1 gene (605446) and found that it encodes several alternatively spliced gene products that interact specifically with RPGR. Roepman et al. (2000) determined that RPGR and RPGRIP1 colocalized in the outer segment of rod photoreceptors, which is in agreement with the retinitis pigmentosa phenotype observed in RP3 patients.

Mavlyutov et al. (2002) used isoform-specific antibodies to demonstrate that RPGR and RPGRIP isoforms are distributed and colocalized at restricted foci throughout the outer segments of human and bovine (but not murine) rod photoreceptors. In humans, these proteins are also localized in cone outer segments, and RPGRIP is expressed in other neurons such as amacrine cells. The authors proposed the existence of species-specific subcellular processes governing the function and/or organization of the photoreceptor outer segment as reflected by the species-specific localization of RPGR and RPGRIP protein isoforms in this compartment. They contended that this may provide a rationale for the disparity of phenotypes among species and among various human mutations.

Using immunofluorescence and serial retinal sectioning, Hong et al. (2003) established the subcellular localization of RPGR in mice and other mammalian species; they found RPGR in connecting cilia of rods and cones and in a homologous structure, the transitional zone of motile cilia in airway epithelia. There was no evidence for species-specific variation of RPGR localization.

By immunohistochemistry, Iannaccone et al. (2003) found that the RPGR gene was expressed throughout the outer segments of human rod and cone photoreceptors, within the epithelial lining of human bronchial and sinus tissues, and in human fetal cochlea, including the stria vascularis, the suprastrial cells, and the apical portion of the spiral limbus. The findings broadened the possible functional roles of RPGR and provided evidence for a broad phenotype in patients with RPGR mutations.

Hong et al. (2004) created an RPGR transgene transcript using alternative splicing involving the purine-rich region of the ORF15 exon, generating a shortened mRNA and a premature stop codon. This truncation mutant caused more rapid photoreceptor degeneration than that in the RPGR null mutant. The disease course was similar, whether the transgene was coexpressed with wildtype RPGR or expressed alone on the RPGR null background. The authors concluded that certain truncated forms of RPGR can behave as a dominant, gain-of-function mutant. These data suggested that human RPGR mutations were not necessarily null and some might also act as dominant alleles, leading to a more severe phenotype than a null mutant.

RPGR, which is essential for the maintenance of photoreceptor viability, is expressed as constitutive and ORF15 variants because of alternative splicing. Hong et al. (2005) examined whether the retina-specific ORF15 variant alone could substantially substitute for RPGR function and tested whether the highly repetitive purine-rich region of ORF15 could be abbreviated without ablating the function, so as to accommodate RPGR replacement genes in adeno-associated virus (AAV) vectors. A cDNA representing mouse Rpgr-ORF15 but shortened by 654 bp in the repetitive region was placed under the control of a chicken beta-actin (CBA) hybrid promoter and used to create transgenic chimeras that were crossed with Rpgr knockout mice. In mice expressing the transgene but null for endogenous Rpgr, transgenic Rpgr-ORF15 was found in the connecting cilia of rod and cone photoreceptors at approximately 20% of the wildtype level. Photoreceptor morphology, cone opsin localization, expression of GFAP (137780) (a marker of retinal degeneration), and ERGs were consistent with the transgene exerting substantial rescue of retinal degeneration due to loss of endogenous Rpgr. Hong et al. (2005) concluded that RPGR-ORF15 is the functionally significant variant in photoreceptors and that the length of its repetitive region can be reduced while preserving its function.

In cultured mammalian cells, Shu et al. (2005) showed that both RPGR-ORF15 and RPGRIP1 (605446) localized to centrioles. By multiple methods, the authors showed that the C2 domain of RPGR-ORF15 interacted with the shuttling protein nucleophosmin (NPM1; 164040), which is a protein chaperone that shuttles between the nucleoli and the cytoplasm and has been associated with licensing of centrosomal division.

By immunoprecipitation of bovine retinal axoneme-enriched fraction, followed by mass spectrometric analysis of proteins separated by SDS-PAGE, Khanna et al. (2005) found that Rpgr-Orf15 bound the ATP-binding proteins Smc1 (SMC1A; 300040) and Smc3 (606062). Protein pull-down experiments revealed that the N-terminal RLD domain of human RPGR-ORF15 bound endogenous bovine Smc1 and Smc3. Bovine Rpgr-Orf15 also associated with Ift88 (600595) and the microtubule motor proteins Kif3a (604683) and Kap3 (KIFAP3; 601836) in an axoneme multiprotein complex. Khanna et al. (2005) concluded that RPGR-ORF15 may be involved in microtubule organization or regulation of transport in primary cilia.

By coimmunoprecipitation of bovine retinal extract, Murga-Zamalloa et al. (2010) found that Rpgr interacted with the small GTPase Rab8a (165040), which has a role in cilia biogenesis and maintenance. Human RPGR interacted predominantly with the GDP-bound form of human RAB8A and stimulated GDP/GTP exchange. Disease-causing mutations in RPGR diminished its interaction with RAB8A and/or reduced its GDP/GTP exchange activity. Depletion of RPGR in human retinal pigment epithelial cells disrupted association of RAB8A with cilia and resulted in shortened primary cilia.


Molecular Genetics

Shu et al. (2007) stated that a total of 240 different mutations in the RPGR gene had been reported, including 24 novel mutations identified by the authors. Of the 240 mutations, 95% are associated with XLRP, 3% with cone, cone-rod dystrophy or atrophic macular atrophy, and 2% with syndromal retinal dystrophies with ciliary dyskinesia and hearing loss.

Branham et al. (2012) screened 214 male patients with simplex retinal degenerative disease, 185 with RP and 29 with cone/cone-rod dystrophy (COD/CORD), for mutations in the RPGR and RP2 (300757) genes. They identified pathogenic mutations in 32 (15%) of the patients. Four patients with COD/CORD had a mutation in the ORF15 mutation hotspot of the RPGR gene. Of the RP patients, 3 had mutations in RP2 and 25 had mutations in RPGR (including 23 in the ORF15 region). Branham et al. (2012) concluded that their results demonstrated a substantial contribution of RPGR mutations to retinal degenerations, and in particular to simplex RP. They suggested that RPGR should be considered as a first tier gene for screening isolated males with retinal degeneration.

Retinitis Pigmentosa 3

Meindl et al. (1996) provided evidence that loss-of-function mutations within RPGR are responsible for X-linked retinitis pigmentosa-3 by identifying 2 small intragenic deletions and 2 nonsense and 3 missense mutations in highly conserved residues in unrelated patients with X-linked RP.

Roepman et al. (1996) screened the Xp21.1-p11.4 RP3 locus interval for microdeletions using a novel technique they called YAC representation hybridization (YRH). Application of this technique led to the generation of a defined amplifiable subset of restriction fragments representing the insert of a YAC spanning the region of interest. The mixture of PCR products was used to study Southern blots of restriction-digested genomic DNA. In 1 of 30 patients with X-linked retinitis pigmentosa, they detected a 6.4-kb microdeletion.

Roepman et al. (1996) detected mutations in the RPGR gene in RP patients and not in controls. Mutation screening was carried out in 28 patients by means of SSCP analysis. They designed intron primers for PCR amplification of 10 exons and detected 5 bandshifts in patients. The corresponding PCR fragments were sequenced and 3 different nucleotide exchanges (312610.0001, 312610.0002, and 312610.0003), and one 4-bp deletion (312610.0004) were identified. None of these changes were detected in 84 male controls. The 6 most 3-prime exons showed no mutations but did reveal several polymorphisms. The 3-prime end of the gene is, however, disrupted by the 6.4-kb deletion which is present in a patient with X-linked retinitis pigmentosa. Roepman et al. (1996) noted that the 5-prime end of the gene and the promoter region had not yet been cloned.

To characterize RPGR mutations in a systematic way, Fujita et al. (1997) identified 11 RP3 families by haplotype analysis. Sequence analysis of the PCR-amplified genomic DNA from patients representing these RP3 families showed no causative mutation in RPGR exons 2 to 19, spanning more than 98% of the coding region. In patients from 2 families, however, they identified transition mutations in the intron region near splice sites (IVS10+3; 312610.0005 and IVS13-8). RNA analysis showed that both splice site mutations resulted in the generation of aberrant RPGR transcripts. The results supported the hypothesis that mutations in the RPGR gene are not a common defect in the RP3 subtype of X-linked RP and that the majority of causative mutations may reside either in as yet unidentified RPGR exons or in another nearby gene at Xp21.1.

Buraczynska et al. (1997) examined the RPGR gene in a cohort of 80 affected males from apparently unrelated X-linked RP families by direct sequencing of the PCR-amplified products from genomic DNA. Fifteen different putative disease-causing mutations were identified in 17 of the 80 families: 4 nonsense mutations, 1 missense mutation, 6 microdeletions, and 4 intronic-sequence substitutions resulting in splice defects. In their Figure 2, they mapped the location of 12 mutations reported by Meindl et al. (1996) and Roepman et al. (1996) and the 15 different mutations identified in this study. Most of the mutations were detected in the conserved N-terminal region of the RPGR protein, containing tandem repeats homologous to those present in the RCC1 protein. In agreement with previous studies, they were able to demonstrate RPGR mutations in only about 20% of the examined X-linked RP patients. On the other hand, the RP3 subtype consistently accounts for 60 to 90% of families localized by linkage and haplotype genotyping. Buraczynska et al. (1997) raised the possibility that the RPGR gene contains unidentified mutation hotspots in sequences that have not been screened, such as the promoter region or intronic sequences and exon 1. The authors could not rule out the alternative possibility of another gene located in proximity to RPGR at Xp21.1 that also causes RP when mutated.

Souied et al. (1997) described 9 families that showed an X-linked pattern of inheritance with a total of 28 affected males and 34 affected females. The females in these families met criteria for the diagnosis of retinitis pigmentosa. The males had a delayed onset of disease, in the second decade, with central vision being preserved until 40 to 45 years of age. Linkage to the RP3 locus was demonstrated, but SSCP and sequence analysis of the RPGR gene demonstrated no mutations. Souied et al. (1997) suggested that these families demonstrated an X-linked dominant form of RP and that the negative mutation results may be explained either by allelic heterogeneity at the RP3 locus or involvement of a distinct locus mapping close to RP3.

Kirschner et al. (1999) demonstrated that the novel RPGR exon 15A that they identified was deleted in a family with X-linked RP (312610.0008). Kirschner et al. (1999) concluded that their results indicate tissue-dependent regulation of alternative splicing of the RPGR gene and that the presence of the retina-specific transcript may explain why phenotypic aberrations in RP3 are confined to the eye.

Miano et al. (1999) found a total of 29 different RPGR mutations identified in northern European and United States patients. They performed mutation analysis of the RPGR gene in a cohort of 49 southern European males with XLRP. By multiplex SSCA and direct sequencing of all 19 RPGR exons, 7 different mutations, all novel, were identified in 8 of the 49 families; these included 3 splice site mutations, 2 microdeletions, and 2 missense mutations. RNA analysis showed that the 3 splice site defects resulted in the generation of aberrant RPGR transcripts. Six of these mutations were detected in the conserved N-terminal region of RPGR protein, containing tandem repeats homologous to repeats within the RCC1 protein (179710). Strikingly, none of the RPGR mutations reported in other populations were identified in this series.

Because mutations in the RPGR gene were found in fewer than the 70 to 75% of XLRP patients predicted from linkage studies, Vervoort et al. (2000) hypothesized that mutations in the remaining XLRP patients may have resided in undiscovered exons of RPGR and sequenced a 172-kb region containing the entire gene. Analysis of the sequence disclosed a new 3-prime terminal exon, ORF15, that was mutated in 60% of XLRP patients examined. Vervoort et al. (2000) concluded that mutations in RPGR are the only cause of RP3 type XLRP and account for the disease in over 70% of XLRP patients and an estimated 11% of all RP patients. Vervoort et al. (2000) found 28 XLRP patients with mutations in exon ORF15, each of which leads to premature termination of translation. Most were small nucleotide deletions, and 5 of them were substitutions leading to nonsense mutation. None of the mutations were detected in 150 control chromosomes. The high frequency of mutations within the terminal exon ORF15 (17 different mutations in 1 kb) compared with other parts of the same RPGR transcript (6 mutations in 1.6 kb), suggested that it is a mutation hotspot. Vervoort et al. (2000) found each of 5 different mutations on at least 2 different haplotypes, indicating recurrent mutation.

In each of 3 unrelated Japanese families segregating X-linked retinitis pigmentosa, Yokoyama et al. (2001) identified a novel mutation in exon ORF15. The mutations were of insertion/deletion type and were predicted to lead to a frameshift, resulting in a truncated protein. The findings supported the previous hypothesis that exon ORF15 is a mutation hotspot. Affected males had typical retinitis pigmentosa, whereas the obligate carrier females showed a wide clinical spectrum, ranging from minor symptoms to severe visual disability. Some carrier females showed typical RP, and most carriers manifested high myopia and astigmatism, with insufficiently corrected visual acuity.

In 4 of the 9 families with XLRP reported by Souied et al. (1997), Rozet et al. (2002) identified mutations in exon ORF15 of the RPGR gene. Rozet et al. (2002) also reported 5 additional affected families with mutations in ORF15. All 7 of the identified mutations were predicted to result in a truncated protein. Rozet et al. (2002) noted that the age at onset in affected females was delayed compared to affected males (20 to 40 years vs 10 to 20 years, respectively).

Vervoort and Wright (2002) reviewed mutations in RPGR in X-linked retinitis pigmentosa (RP3). They commented on the fact that exon ORF15 is a hotspot for mutation, at least in the British population, in which it harbors 80% of the mutations found within a sample of 47 X-linked retinitis pigmentosa patients.

In a North American cohort of 234 families with RP, Breuer et al. (2002) conducted a comprehensive screen of the RP2 (300757) and RPGR (including ORF15) genes and their 5-prime upstream regions. Of these families, 91 (39%) showed definitive X-linked inheritance, an additional 88 (38%) revealed a pattern consistent with X-linked disease, and the remaining 55 (23%) were simplex male patients with RP who had an early onset and/or severe disease. In agreement with previous studies, they showed that mutations in the RP2 gene and in the original 19 RPGR exons are detected in less than 10% and approximately 20% of XLRP probands, respectively. Their studies revealed RPGR ORF15 mutations in an additional 30% of 91 well-documented families with X-linked recessive inheritance and in 22% of the total 234 probands analyzed. They suggested that mutations in an uncharacterized RPGR exon(s), intronic changes, or another gene in the region may be responsible for the disease in the remainder of this North American cohort.

Bader et al. (2003) screened 58 German XLRP families and found RP2 mutations in 8% and RPGR mutations in 71%. They also reported a detailed strategy for analyzing the RPGR ORF15 mutation hot spot, which could not be screened by standard procedures.

Sharon et al. (2003) determined the mutation spectrum of the RP2 and RPGR genes in patients with X-linked retinitis pigmentosa. They screened 187 unrelated male patients and found 10 mutations in RP2, 2 of which were novel, and 80 mutations in RPGR, 41 of which were novel. Patients with RP2 mutations had, on average, lower visual acuity but similar visual field area, final dark-adapted threshold, and 30-Hz ERG amplitude compared with those with RPGR mutations. Among the 66% of patients with RPGR mutations in ORF, regression analysis showed that the final dark-adapted threshold became lower (i.e., closer to normal) and that the 30-Hz ERG amplitude increased as the length of the wildtype ORF15 amino acid sequence increased. Furthermore, as the length of the abnormal amino acid sequence following ORF15 frameshift mutations increased, the severity of disease increased. In summary, these cross-sectional analyses suggested that, at a given age, patients with RP2 mutations retained less visual acuity than do patients with RPGR mutations and that, among patients with RPGR mutations, those with ORF15 mutations have milder disease than do patients with mutations in exons 1 to 14.

Demirci et al. (2006) reported a 16-year-old boy with RP (300029) and bilateral Coats-like vasculopathy (see 300216) in whom they identified a novel nonsense mutation in ORF15 of the RPGR gene (312610.0024).

Pelletier et al. (2007) reported the screening of the RP2 and RPGR genes in a cohort of 127 French families comprising 93 familial cases of retinitis pigmentosa suggesting X-linked inheritance, including 48 of 93 families; 7 male sibships of RP; 25 sporadic male cases of RP; and 2 cone dystrophies (COD). They identified a total of 14 RP2 mutations, 12 of which were novel, in 14 of 88 familial cases of RP and 1 of 25 sporadic male cases (4%). In 13 of 14 of the familial cases, no expression of the disease was noted in females, while in 1 of 14 families 1 woman developed retinitis pigmentosa in the third decade. A total of 42 RPGR mutations, 26 of which were novel, were identified in 80 families, including 69 of 88 familial cases (78.4%); 2 of 7 male sibship cases (28.6%); 8 of 25 sporadic male cases (32%); and 1 of 2 COD. No expression of the disease was noted in females in 41 of 69 familial cases (59.4%), while at least 1 severely affected woman was recognized in 28 of 69 families (40.6%). The frequency of RP2 and RPGR mutations in familial cases of retinitis pigmentosa suggestive of X-linked transmission was in accordance with that reported elsewhere (RP2: 15.9% vs 6-20%; RPGR: 78.4% vs 55-90%). About 30% of male sporadic cases and 30% of male sibships of RP carried RP2 or RPGR mutations, confirming the pertinence of the genetic screening of XLRP genes in male patients affected with RP commencing in the first decade and leading to profound visual impairment before the age of 30 years.

Sandberg et al. (2007) measured the rates of visual acuity, visual field, and electroretinogram (ERG) loss in 2 large cohorts, one of patients with XLRP due to mutations in the RPGR gene and the other of patients with autosomal dominant RP due to mutations in the RHO gene (see 180380). Patients with RPGR mutations lost Snellen visual acuity at more than twice the mean rate of patients with RHO mutations. The median age of legal blindness was 32 years younger in patients with RPGR mutation than in patients with RHO mutations. Legal blindness was due primarily to loss of visual acuity in RPGR patients and to loss of visual field in RHO patients. Loss of acuity in RPGR patients appeared to be associated with foveal thinning.

In affected individuals from an Israeli family with 'semi-dominant' X-linked retinitis pigmentosa, in which obligatory female carriers manifested high myopia, low visual acuity, constricted visual fields, and severely reduced electroretinogram amplitudes, Banin et al. (2007) identified the G275S mutation in the RPGR gene (312610.0003). The authors stated that obligate carriers from 2 unrelated Danish families in which Roepman et al. (1996) previously identified this mutation had no visual complaints and normal to slightly reduced retinal function. The disease-related RPGR haplotype of the Israeli family was found to be different from that of 2 Danish families, indicating that the G275S mutation arose twice independently on different X-chromosome backgrounds. Genetic analysis excluded skewed X-inactivation patterns, chromosomal abnormalities, distorted RPGR expression levels, and mutations in 3 candidate genes as the cause for the differences in disease severity of female carriers. Banin et al. (2007) suggested that an additional gene or genes linked to RPGR modulate disease expression in severely affected carriers.

Nishiguchi et al. (2013) identified a Japanese male RP patient who was heterozygous for a frameshift mutation in the ciliary gene NEK2 (604043.0001), but who also carried a frameshift mutation in RPGR (312610.0026). Studies in zebrafish suggested that the RPGR allele interacts in trans with the NEK2 locus to exacerbate photoreceptor defects.

Among female carriers from 45 families with RP3, Comander et al. (2015) found that those with RPGR ORF15 mutations tended to have worse visual function than those with RPGR exon 1 through 14 mutations.

Cone-Rod Degeneration

Mears et al. (2000) restudied a family with what was labeled 'X-linked dominant cone-rod degeneration' (300029) and thought to map to Xp22.13-p22.11, and remapped the disorder to Xp22.1-p11.4. This new interval overlapped the RP3 (RPGR) and the CORDX1 (304020) genes. They identified a de novo insertion (312610.0013) in exon ORF15 of the RPGR gene. The identification of an RPGR mutation in a family with a severe form of cone-rod degeneration suggested that RPGR mutations may encompass a broader phenotypic spectrum than had previously been recognized in 'typical' retinitis pigmentosa.

Retinitis Pigmentosa and Sinopulmonary Infections with or without Deafness

In the family with X-linked retinitis pigmentosa with recurrent respiratory infections (300455) described by van Dorp et al. (1992), Dry et al. (1999) identified an IVS5+1G-T splice site mutation in the RPGR gene (312610.0016).

In a family in which affected males in an X-linked recessive pedigree pattern had retinitis pigmentosa associated with impaired hearing and sinorespiratory infections, Zito et al. (2003) identified A 2-bp deletion, 845delTG, in exon 8 of the RPGR gene (312610.0019).

In a family in which a mother had retinitis pigmentosa and her 2 sons had retinitis pigmentosa, multiple respiratory infections, and primary ciliary dyskinesia, Moore et al. (2006) identified a 57-bp deletion in the RPGR gene (312610.0023).

Atrophic Macular Degeneration

Ayyagari et al. (2002) described a family in which 10 males had primarily macular atrophy causing progressive loss of visual acuity with minimal peripheral visual impairment (300834). One additional male showed extensive macular degeneration plus peripheral loss of retinal pigment epithelium and choriocapillaries. Full-field electroretinograms showed normal cone and rod responses in some affected males despite advanced macular degeneration. In affected members of this family, Ayyagari et al. (2002) identified a G-to-T transversion at nucleotide 1164 of intron 15 (312610.0017) that cosegregated with the disease and may create a donor splice site. Thus the phenotypic range associated with this gene was expanded.


Animal Model

Hong et al. (2000) created an RPGR-deficient murine model of RP3 by gene knockout. In the mutant mice, cone photoreceptors exhibited ectopic localization of cone opsins in the cell body and synapses, and rod photoreceptors had a reduced level of rhodopsin. Subsequently, both rod and cone photoreceptors degenerated. RPGR was found normally localized to the connecting cilia of rod and cone photoreceptors. The data pointed to a role for RPGR in maintaining the polarized protein distribution across the connecting cilium by facilitating directional transport or restricting redistribution. The function of RPGR is essential for the long-term maintenance of photoreceptor viability.

X-linked progressive retinal atrophy (XLPRA) in the Siberian Husky dog closely resembles XLRP in humans. Zeiss et al. (2000) established a linkage map of the canine X chromosome, and determined that XLPRA was tightly linked to an intragenic RPGR polymorphism (lod = 11.7, zero recombination), thus confirming locus homology with RP3. They cloned the full-length canine RPGR cDNA and 3 additional splice variants. No disease-causing mutation was found in the RPGR-coding sequence of the 4 splice variants characterized, a finding similar to approximately 80% of human XLRP patients whose disease maps to the RP3 locus.

Zhang et al. (2002) found different mutations in exon ORF15 of the RPGR gene in 2 distinct mutant dog strains: XLPRA1 and XLPRA2. Microdeletions resulting in a premature stop or a frameshift mutation produced very different retinal phenotypes, which were allele-specific and consistent for each mutation. The phenotype associated with a frameshift mutation in XLPRA2 was very severe and manifested during retinal development; the phenotype resulting from a nonsense mutation in XLPRA1 was expressed only after normal photoreceptor morphogenesis. The frameshift mutation dramatically alters the deduced amino acid sequence, and the protein aggregates in the endoplasmic reticulum of transfected COS-7 cells.

Beltran et al. (2006) described the course of retinal disease in canine XLPRA2 caused by a 2-nucleotide microdeletion in RPGR ORF15. The disorder is characterized by abnormal photoreceptor maturation followed by progressive rod-cone degeneration and early inner retina remodeling. Abnormal development of photoreceptors was recognizable as early as 3.9 weeks of age. Outer segment (OS) misalignment was followed by their disorganization and fragmentation. Reduction in length and broadening of rod and cone inner segments (IS) was observed next, followed by the focal loss of rod and cone IS later. The proportion of dying photoreceptors peaked at approximately 6 to 7 weeks of age and was significantly reduced after 12 weeks. In addition to rod and cone opsin mislocalization, there was early rod neurite sprouting, retraction of rod bipolar cell dendrites, and increased Mueller cell reactivity. Later in the course of the disease, changes were also noted in horizontal cells and amacrine cells.

Beltran et al. (2007) noted that ciliary neurotrophic factor (CNTF; 118945) had been found to rescue photoreceptors in several animal models. They evaluated treatment with CNTF in XLPRA2 dogs. All CNTF-treated eyes showed early clinical signs of corneal epitheliopathy, subcapsular cataracts, and uveitis. No statistically significant difference in outer nuclear layer thickness was seen between CNTF-treated and control eyes. Prominent retinal remodeling that consisted of an abnormal increase in the number of rods, and in misplacement of some rods, cones, and bipolar and Muller cells, was observed in the peripheral retina of CNTF-treated eyes. In XLPRA2 dogs, intravitreal injection of CNTF failed to prevent photoreceptors from undergoing cell death in the central and midperipheral retina. CNTF also caused ocular side effects and morphologic alterations in the periphery that were consistent with cell dedifferentiation and proliferation. Beltran et al. (2007) concluded that some inherited forms of retinal degeneration may not respond to the neuroprotective effects of CNTF.

Ghosh et al. (2010) showed that rpgr was expressed predominantly in the retina, brain, and gut of zebrafish. In zebrafish retina, rpgr primarily localized to the sensory cilium of photoreceptors. Antisense morpholino-mediated knockdown of rpgr function in zebrafish resulted in reduced length of Kupffer vesicle cilia and was associated with ciliary anomalies including shortened body axis, kinked tail, hydrocephaly, and edema, but did not affect retinal development. These phenotypes could be rescued by wildtype human RPGR. Several RPGR mutants (see, e.g., 312610.0006 and 312610.0020) could also reverse the morpholino-induced phenotype, suggesting their potential hypomorphic function. Selected RPGR mutations observed in XLRP (see, e.g. 312610.0009; E589X) or syndromic retinitis pigmentosa (312610.0016; 312610.0019; 312610.0023) did not completely rescue the rpgr-morpholino phenotype, indicating a more deleterious effect of the mutation on the function of RPGR. Ghosh et al. (2010) proposed that RPGR may be involved in cilia-dependent cascades during development in zebrafish.

Shu et al. (2010) identified 2 genes resembling human RPGR in zebrafish (Zfrpgr1 and Zfrpgr2), both of which are expressed within the nascent and adult eye as well as more widely during development. Zfrpgr2 appears to be functionally orthologous to human RPGR, because it encodes similar protein isoforms (Zfrpgr2(Orf15) and Zfrpgr2(ex1-17)) and, similar to other ciliary proteins, translation suppression causes developmental defects affecting gastrulation and tail and head development. These defects are consistent with a ciliary function and were rescued by human RPGR but not by RPGR mutants causing retinal dystrophy. Unlike in mammals, RPGR knockdown in zebrafish resulted in both abnormal development and increased cell death in the dysplastic retina. Developmental abnormalities in the eye included lamination defects, failure to develop photoreceptor outer segments, and a small eye phenotype, associated with increased cell death throughout the retina. These defects could be rescued by expression of wildtype but not mutant forms of human RPGR. Zfrpgr2 knockdown also resulted in an intracellular transport defect affecting retrograde but not anterograde transport of organelles. Shu et al. (2010) concluded that Zfrpgr2 is necessary both for the normal differentiation and lamination of the retina and to prevent apoptotic retinal cell death, which may relate to its proposed role in dynein-based retrograde transport processes.


ALLELIC VARIANTS ( 26 Selected Examples):

.0001 RETINITIS PIGMENTOSA 3

RPGR, PHE130CYS
  
RCV000010575...

In a patient with retinitis pigmentosa-3 (300029), Roepman et al. (1996) identified a T-to-G transversion at nucleotide 420 of their RP3 cDNA sequence, resulting in a phe130-to-cys substitution (F130C). The authors did not denote the amino acid residue position in their publication.

Murga-Zamalloa et al. (2010) showed that the F130C mutation in RPGR did not alter interaction of RPGR with the small GTPase RAB8A (165040) in vitro, but it reduced the ability of RPGR to induce GDP/GTP exchange on RAB8A.


.0002 RETINITIS PIGMENTOSA 3

RPGR, PRO235SER
  
RCV000010576...

In a patient with retinitis pigmentosa-3 (300029), Roepman et al. (1996) identified a C-to-T transition at nucleotide 734 of their RP3 cDNA sequence, resulting in a pro235-to-ser substitution (P235S). The authors did not denote the amino acid residue position in their publication.


.0003 RETINITIS PIGMENTOSA 3

RPGR, GLY275SER
  
RCV000010577...

In 2 patients with retinitis pigmentosa-3 (300029), Roepman et al. (1996) identified a G-to-A transition at nucleotide 854 of their RP3 cDNA sequence, resulting in a gly275-to-ser substitution (G275S). The authors did not denote the amino acid residue position in their publication.

In affected individuals from an Israeli family with 'semi-dominant' X-linked retinitis pigmentosa, in which obligatory female carriers manifested high myopia, low visual acuity, constricted visual fields, and severely reduced electroretinogram amplitudes, Banin et al. (2007) identified the G275S mutation in the RPGR gene. The disease-related RPGR haplotype of the Israeli family was found to be different from that of the 2 families previously studied by Roepman et al. (1996) in which obligate carriers of G275S had no visual complaints, indicating that the G275S mutation arose twice independently on different X-chromosome backgrounds. Genetic analysis excluded skewed X-inactivation patterns, chromosomal abnormalities, distorted RPGR expression levels, and mutations in 3 candidate genes as the cause for the differences in disease severity of female carriers. Banin et al. (2007) suggested that an additional gene or genes linked to RPGR modulate disease expression in severely affected carriers.


.0004 RETINITIS PIGMENTOSA 3

RPGR, 4-BP DEL, NT1433
  
RCV000085055...

Roepman et al. (1996) found a 4-bp deletion of nucleotides 1433-1436 of their RP3 cDNA sequence in a patient with retinitis pigmentosa-3 (300029) that resulted in a truncated RP3 protein with 6 abnormal C-terminal amino acids.


.0005 RETINITIS PIGMENTOSA 3

RPGR, IVS10DS, A-G, +3
  
RCV000010579...

In a patient with retinitis pigmentosa-3 (300029), Fujita et al. (1997) found an A-to-G transition in the RPGR gene at the third basepair downstream of exon 10 nucleotide 1304 (sequence designation is according to Meindl et al., 1996) in the splice-donor region of intron 10. The mutation resulted in incorrect splicing of the exon 10-11 junction.


.0006 RETINITIS PIGMENTOSA 3

RPGR, GLY60VAL
  
RCV000010580...

One of the 15 novel mutations identified by Buraczynska et al. (1997) was a G-to-T transversion at nucleotide 238 in exon 3, predicted to lead to a gly60-to-val (G60V) amino acid substitution. Fishman et al. (1998) reported in detail on 2 families with retinitis pigmentosa-3 (300029) with the G60V mutation, one of which was the family earlier reported by Buraczynska et al. (1997). The mutation was associated with a severe clinical phenotype in male patients and a patchy retinopathy without a tapetal-like reflex in carrier females. Psychophysical and electrophysiologic testing on carriers indicated that cone and rod functions were impaired equivalently. When present in carriers, visual field restriction was most apparent in, or limited to, the superotemporal quadrant, which corresponded to the retinal pigmentary changes that tended to occur in the inferonasal retina.


.0007 RETINITIS PIGMENTOSA 3

RPGR, 2-BP DEL, NT1571
  
RCV000085061...

In a black family with retinitis pigmentosa-3 (300029), Fishman et al. (1998) identified a 2-basepair deletion in the RP3 gene. The deletion occurred in exon 13 and created a frameshift and premature stop codon, resulting in a protein truncation. The deletion involved nucleotides 1571 and 1572, a CA dinucleotide. The clinical findings were those characteristic of X-linked retinitis pigmentosa. In 2 obligate carriers, a tapetal-like reflex was not clinically apparent.


.0008 RETINITIS PIGMENTOSA 3

RPGR, EX15ADEL
   RCV000010583

In a family with retinitis pigmentosa-3 (300029), Kirschner et al. (1999) identified deletion of exon 15A of the RPGR gene. The splice variant containing exon 15A is expressed only in the retina. The phenotype was of retinal dystrophy with relatively late onset of night blindness, around the age of 10 years. From 30 years of age, very rapid deterioration took place and led to blindness by the age of 37 years. Electrophysiologically, the retinopathy appeared as a cone-rod dystrophy.


.0009 RETINITIS PIGMENTOSA 3

RPGR, THR99ASN
  
RCV000010584...

One of the unique mutations found by Miano et al. (1999) in south European patients with retinitis pigmentosa-3 (300029) was a C-to-A transversion of nucleotide 355 of the RP3 gene resulting in a thr99-to-asn (T99N) amino acid substitution. The patient was Spanish.


.0010 RETINITIS PIGMENTOSA 3

RPGR, 2-BP DEL, 673AG
  
RCV000162095...

Vervoort et al. (2000) identified a novel exon, ORF15, of the RPGR gene. One mutation detected in this region, found in 4 families with retinitis pigmentosa-3 (300029), was a 2-nucleotide deletion of AG at nucleotide 673 resulting in frameshift. The high frequency of mutations found in this exon suggested to Vervoort et al. (2000) that ORF15 is a mutation hotspot, with its high mutability being due to nucleotide composition (purine-rich) or repetitive nature of the sequence.


.0011 RETINITIS PIGMENTOSA 3

RPGR, 2-BP DEL, 652AG
  
RCV000076907...

In 2 families with retinitis pigmentosa-3 (300029), Vervoort et al. (2000) identified a mutation in ORF15, a 2-bp deletion of AG at nucleotide 652.


.0012 RETINITIS PIGMENTOSA 3

RPGR, GLU299TER
  
RCV000010586...

In a family with retinitis pigmentosa-3 (300029), Vervoort et al. (2000) identified a G-to-T transversion in ORF15, an alternatively spliced 3-prime terminal exon of the RPGR gene. This mutation resulted in a glutamic acid-to-stop substitution at residue 299 of ORF15 (E299X).


.0013 RETINITIS PIGMENTOSA 3

RPGR, 1-BP INS, 173A
  
RCV001251559...

In a family with X-linked cone-rod degeneration, previously reported by McGuire et al. (1995) as RP15 (300029), Mears et al. (2000) identified a 1-bp insertion, an adenine (173_174insA), causing a frameshift with insertion of 9 novel amino acids and truncation of the protein product 501 amino acids premature of the ORF15 stop codon. In this family, affected males and 'carrier' females presented with early cone involvement, which differs from the typical rod-predominant manifestation of X-linked RP.


.0014 CONE-ROD DYSTROPHY, X-LINKED, 1

CONE DYSTROPHY, X-LINKED, 1, INCLUDED
RPGR, 2-BP DEL, 1343GG
  
RCV000010588...

In 2 of 3 families with X-linked cone-rod dystrophy (CORDX1; 304020), Demirci et al. (2002) demonstrated a 2-bp deletion, delGG, in ORF15 of the RPGR gene, resulting in a frameshift leading to altered amino acid structure and early termination.

Yang et al. (2002) mapped 2 Caucasian families with X-linked cone dystrophy (COD1; see 304020) to the CORDX1 locus on Xp and identified 2 distinct mutations in ORF15 of the RPGR gene. One was ORF15+1343-1344delGG and the other ORF15+694-708del15 (312610.0018).


.0015 CONE-ROD DYSTROPHY, X-LINKED, 1

RPGR, 2-BP DEL, 1339AG
  
RCV000010589...

In a family with X-linked cone-rod dystrophy (CORDX1; 304020), Demirci et al. (2002) found a 2-bp deletion, delAG, in exon 15 (ORF15) of the RPGR gene, resulting in a frameshift leading to altered amino acid structure and early termination.


.0016 RETINITIS PIGMENTOSA AND SINORESPIRATORY INFECTIONS WITH OR WITHOUT DEAFNESS

RPGR, IVS5, G-T, +1
  
RCV000010590...

Dry et al. (1999) demonstrated a splice site mutation in the RPGR gene in a family with X-linked retinitis pigmentosa with recurrent respiratory infections (300455) in which van Dorp et al. (1992) had, by electron microscopy, shown nasal ciliary abnormalities in some affected males, consisting of deficient inner dynein arms, incomplete microtubules, and disorientation of cilia, associated with recurrent respiratory infections indistinguishable from immotile cilia syndrome. Van Dorp et al. (1992) did not note whether hearing was impaired in affected individuals.


.0017 MACULAR DEGENERATION, ATROPHIC, X-LINKED

RPGR, IVS15, G-T, +1164
  
RCV000010591...

In a family with X-linked recessive atrophic macular degeneration (300834), Ayyagari et al. (2002) found that affected males had an ORF15+1164G-T mutation thought to create a novel donor splice site in the RPGR gene.


.0018 CONE DYSTROPHY, X-LINKED, 1

RPGR, 15-BP DEL, NT694
  
RCV000475821...

Yang et al. (2002) mapped 2 Caucasian families with X-linked cone dystrophy (see 304020) to the CORDX1 locus on Xp and identified 2 distinct mutations in ORF15 of the RPGR gene. One was ORF15+1343-1344delGG (312610.0014) and the other ORF15+694-708del15. The latter mutation was predicted to delete 5 amino acids from the C-terminal portion of the protein product.


.0019 RETINITIS PIGMENTOSA, SINORESPIRATORY INFECTIONS, AND DEAFNESS

RPGR, 2-BP DEL, 845TG
  
RCV003151714

In a family in which affected males in an X-linked recessive pedigree pattern had retinitis pigmentosa associated with impaired hearing and sinorespiratory infections (300455), Zito et al. (2003) identified a 2-bp deletion, 845delTG, in exon 8 of the RPGR gene. This frameshift mutation at residue 262 was predicted to introduce 19 new amino acids and a premature stop codon, resulting in a truncated protein of 280 residues. Carrier females and affected males in this kindred were myopic but female carriers were asymptomatic, had normal fields to confrontation, and showed sparse peripheral intraretinal pigmentation. However, additional systemic symptoms were observed in both hemizygous males and heterozygous females. One of the most striking and obvious additional features was the requirement of hearing aids by both affected males and female carriers. Both had severe recurrent ear infections from early childhood continuing into adulthood. Because of hearing loss in the high frequencies, the audiogram was considered consistent with sensorineural hearing loss, although a conductive hearing component may have contributed. Affected males and carrier females suffered from severe recurrent sinus infections. Three affected males had chronic recurrent chest infections starting in early childhood, with episodes of bronchitis, which continued into adulthood. The possibility of renal failure being part of the clinical picture was suggested by its occurrence in 1 affected male. The phenotype overlapped those described for primary ciliary dyskinesia (244400) and Usher syndrome (276900) and provided support for an essential ciliary function for RPGR in the retina and other tissues.


.0020 RETINITIS PIGMENTOSA, SINORESPIRATORY INFECTIONS, AND DEAFNESS

RPGR, GLY173ARG
  
RCV003128227...

In 2 brothers with X-linked retinitis pigmentosa with impaired hearing and recurrent respiratory infections (300455), Iannaccone et al. (2003) identified a 576G-C transversion in a conserved region of the RPGR gene, resulting in a gly173-to-arg (G173R) substitution. The mutation was also identified in the asymptomatic mother and the symptomatic grandmother. The G173R change was not identified in more than 200 control chromosomes.


.0021 CONE-ROD DYSTROPHY, X-LINKED, 1

RPGR, GLU364ASP, GLU365TER
   RCV000010595

In a family with X-linked cone-rod dystrophy (304020), Ebenezer et al. (2005) found a 2 consecutive nucleotide substitutions in the ORF15 exon of the RPGR gene, 1094A-C and 1095G-T, that resulted in glu364-to-asp and glu365-to-ter amino acid substitutions, respectively (E364D/E365X).


.0022 CONE-ROD DYSTROPHY, X-LINKED, 1

RPGR, GLY392TER
  
RCV000010596

In a family with X-linked cone-rod dystrophy (304020), Ebenezer et al. (2005) identified a G-to-T transversion at nucleotide 1176 of the ORF15 exon of the RPGR gene that resulted in a gly392-to-ter (G392X) substitution.


.0023 RETINITIS PIGMENTOSA AND SINORESPIRATORY INFECTIONS WITHOUT DEAFNESS

RPGR, 57-BP DEL, NT631
   RCV003151716

In a family in which a mother with retinitis pigmentosa had 2 boys with multiple ear, sinus, and respiratory infections who were diagnosed with primary ciliary dyskinesia and retinitis pigmentosa (300455), Moore et al. (2006) identified a 57-bp deletion involving the last 48 bp of exon 6 plus the following 9 bp of the adjacent intron (631-IVS6+9del) in the RPGR gene. The boys had axonemal abnormalities on electron microscopy leading to the diagnosis of primary ciliary dyskinesia; ophthalmologic examination confirmed the diagnosis of retinitis pigmentosa. Moore et al. (2006) stated that this was the first clear demonstration of X-linked transmission of primary ciliary dyskinesia.


.0024 RETINITIS PIGMENTOSA 3

RPGR, 912G-T
  
RCV001073958...

Demirci et al. (2006) reported a 16-year-old boy with RP (300029) and bilateral Coats-like vasculopathy (see 300216) in whom they identified a 912G-T transversion in ORF15 of the RPGR gene, predicted to result in a truncated protein missing 264 amino acids at the carboxyl end. The mutation segregated with RP in the family; clinical findings in other family members, including 2 affected male patients and 3 obligate carrier females, were consistent with typical X-linked recessive RP. Because the proband was the only family member who had Coats-like exudative vasculopathy, Demirci et al. (2006) suggested that other genetic and/or environmental factors might be involved.


.0025 RETINITIS PIGMENTOSA 3

RPGR, IVS9AS, G-A, -55
  
RCV000010599...

In a Swiss man with mild retinitis pigmentosa-3 (300029), Neidhardt et al. (2007) identified a hemizygous G-to-A transition located 363 bp downstream of exon 9 and 55 bp upstream from the exon splice acceptor site of exon 9A (g.26652G-A), resulting in increased levels of RPGR mRNA transcripts containing exon 9A. The man had night blindness since childhood and presented in his late thirties with peripheral visual impairment. The level of exon 9A expression in the patient's unaffected mother, who was heterozygous for the mutation, was normalized to 100%; the patient was found to have 180% expression levels.


.0026 RETINITIS PIGMENTOSA 3

RPGR, 2-BP DEL, 2405AG
   RCV000076907...

In a Japanese male patient with retinitis pigmentosa (RP3; 300029), Nishiguchi et al. (2013) identified a 2-bp deletion (c.2405_2406delAG) in the RPGR gene, causing a frameshift (Glu802fs); the authors stated that this mutation had previously been described as a sufficient cause of X-linked RP by Vervoort et al. (2000). The Japanese patient also carried a heterozygous frameshift mutation in another RP-associated ciliary gene, NEK2 (604043.0001); studies in zebrafish suggested that the RPGR allele interacts in trans with the NEK2 locus to exacerbate photoreceptor defects.


REFERENCES

  1. Ayyagari, R., Demirci, F. Y., Liu, J., Bingham, E. L., Stringham, H., Kakuk, L. E., Boehnke, M., Gorin, M. B., Richards, J. E., Sieving, P. A. X-linked recessive atrophic macular degeneration from RPGR mutation. Genomics 80: 166-171, 2002. [PubMed: 12160730, related citations] [Full Text]

  2. Bader, I., Brandau, O., Achatz, H., Apfelstedt-Sylla, E., Hergersberg, M., Lorenz, B., Wissinger, B., Wittwer, B., Rudolph, G., Meindl, A., Meitinger, T. X-linked retinitis pigmentosa: RPGR mutations in most families with definite X linkage and clustering of mutations in a short sequence stretch of exon ORF15. Invest. Ophthal. Vis. Sci. 44: 1458-1463, 2003. [PubMed: 12657579, related citations] [Full Text]

  3. Banin, E., Mizrahi-Meissonnier, L., Neis, R., Silverstein, S., Magyar, I., Abeliovich, D., Roepman, R., Berger, W., Rosenberg, T., Sharon, D. A non-ancestral RPGR missense mutation in families with either recessive or semi-dominant X-linked retinitis pigmentosa. Am. J. Med. Genet. 143A: 1150-1158, 2007. [PubMed: 17480003, related citations] [Full Text]

  4. Beltran, W. A., Hammond, P., Acland, G. M., Aguirre, G. D. A frameshift mutation in RPGR exon ORF15 causes photoreceptor degeneration and inner retina remodeling in a model of X-linked retinitis pigmentosa. Invest. Ophthal. Vis. Sci. 47: 1669-1681, 2006. [PubMed: 16565408, related citations] [Full Text]

  5. Beltran, W. A., Wen, R., Acland, G. M., Aguirre, G. D. Intravitreal injection of ciliary neurotrophic factor (CNTF) causes peripheral remodeling and does not prevent photoreceptor loss in canine RPGR mutant retina. Exp. Eye Res. 84: 753-771, 2007. [PubMed: 17320077, images, related citations] [Full Text]

  6. Boylan, J. P., Wright, A. F. Identification of a novel protein interacting with RPGR. Hum. Molec. Genet. 9: 2085-2093, 2000. [PubMed: 10958647, related citations] [Full Text]

  7. Branham, K., Othman, M., Brumm, M., Karoukis, A. J., Atmaca-Sonmez, P., Yashar, B. M., Schwartz, S. B., Stover, N. B., Trzupek, K., Wheaton, D., Jennings, B., Ciccarelli, M. L., and 13 others. Mutations in RPGR and RP2 account for 15% of males with simplex retinal degenerative disease. Invest. Ophthal. Vis. Sci. 53: 8232-8237, 2012. [PubMed: 23150612, related citations] [Full Text]

  8. Breuer, D. K., Yashar, B. M., Filippova, E., Hiriyanna, S., Lyons, R. H., Mears, A. J., Asaye, B., Acar, C., Vervoort, R., Wright, A. F., Musarella, M. A., Wheeler, P., and 9 others. A comprehensive mutation analysis of RP2 and RPG2 in a North American cohort of families with X-linked retinitis pigmentosa. Am. J. Hum. Genet. 70: 1545-1554, 2002. Note: Erratum: Am. J. Hum. Genet. 71: 1258 only, 2002. [PubMed: 11992260, images, related citations] [Full Text]

  9. Buraczynska, M., Wu, W., Fujita, R., Buraczynska, K., Phelps, E., Andreasson, S., Bennett, J., Birch, D. G., Fishman, G. A., Hoffman, D. R., Inana, G., Jacobson, S. G., Musarella, M. A., Sieving, P. A., Swaroop, A. Spectrum of mutations in the RPGR gene that are identified in 20% of families with X-linked retinitis pigmentosa. Am. J. Hum. Genet. 61: 1287-1292, 1997. [PubMed: 9399904, related citations] [Full Text]

  10. Comander, J., Weigel-DiFranco, C., Sandberg, M. A., Berson, E. L. Visual function in carriers of X-linked retinitis pigmentosa. Ophthalmology 122: 1899-1906, 2015. [PubMed: 26143542, images, related citations] [Full Text]

  11. Demirci, F. Y. K., Rigatti, B. W., Mah, T. S., Gorin, M. B. A novel RPGR exon ORF15 mutation in a family with X-linked retinitis pigmentosa and Coats'-like exudative vasculopathy. Am. J. Ophthal. 141: 208-210, 2006. [PubMed: 16387007, related citations] [Full Text]

  12. Demirci, F. Y. K., Rigatti, B. W., Wen, G., Radak, A. L., Mah, T. S., Baic, C. L., Traboulsi, E. I., Alitalo, T., Ramser, J., Gorin, M. B. X-linked cone-rod dystrophy (locus COD1): identification of mutations in RPGR exon ORF15. Am. J. Hum. Genet. 70: 1049-1053, 2002. [PubMed: 11857109, related citations] [Full Text]

  13. Dry, K. L., Manson, F. D. C., Lennon, A., Bergen, A. A. B., Van Dorp, D. B., Wright, A. F. Identification of a 5-prime splice site mutation in the RPGR gene in a family with X-linked retinitis pigmentosa (RP3). Hum. Mutat. 13: 141-145, 1999. [PubMed: 10094550, related citations] [Full Text]

  14. Ebenezer, N. D., Michaelides, M., Jenkins, S. A., Audo, I., Webster, A. R., Cheetham, M. E., Stockman, A., Maher, E. R., Ainsworth, J. R., Yates, J. R., Bradshaw, K., Holder, G. E., Moore, A. T., Hardcastle, A. J. Identification of novel RPGR ORF15 mutations in X-linked progressive cone-rod dystrophy. Invest. Ophthal. Vis. Sci. 46: 1891-1898, 2005. [PubMed: 15914600, related citations] [Full Text]

  15. Fishman, G. A., Grover, S., Buraczynska, M., Wu, W., Swaroop, A. A new 2-base pair deletion in the RPGR gene in a black family with X-linked retinitis pigmentosa. Arch. Ophthal. 116: 213-218, 1998. [PubMed: 9488274, related citations] [Full Text]

  16. Fishman, G. A., Grover, S., Jacobson, S. G., Alexander, K. R., Derlacki, D. J., Wu, W., Buraczynska, M., Swaroop, A. X-linked retinitis pigmentosa in two families with a missense mutation in the RPGR gene and putative change of glycine to valine at codon 60. Ophthalmology 105: 2286-2296, 1998. [PubMed: 9855162, related citations] [Full Text]

  17. Fujita, R., Buraczynska, M., Gieser, L., Wu, W., Forsythe, P., Abrahamson, M., Jacobson, S. G., Sieving, P. A., Andreasson, S., Swaroop, A. Analysis of the RPGR gene in 11 pedigrees with the retinitis pigmentosa type 3 genotype: paucity of mutations in the coding region but splice defects in two families. Am. J. Hum. Genet. 61: 571-580, 1997. [PubMed: 9326322, related citations] [Full Text]

  18. Ghosh, A. K., Murga-Zamalloa, C. A., Chan, L., Hitchcock, P. F., Swaroop, A., Khanna, H. Human retinopathy-associated ciliary protein retinitis pigmentosa GTPase regulator mediates cilia-dependent vertebrate development. Hum. Molec. Genet. 19: 90-98, 2010. [PubMed: 19815619, images, related citations] [Full Text]

  19. Hong, D.-H., Pawlyk, B. S., Adamian, M., Li, T. Dominant, gain-of-function mutant produced by truncation of RPGR. Invest. Ophthal. Vis. Sci. 45: 36-41, 2004. [PubMed: 14691151, related citations] [Full Text]

  20. Hong, D.-H., Pawlyk, B. S., Adamian, M., Sandberg, M. A., Li, T. A single, abbreviated RPGR-ORF15 variant reconstitutes RPGR function in vivo. Invest. Ophthal. Vis. Sci. 46: 435-441, 2005. [PubMed: 15671266, related citations] [Full Text]

  21. Hong, D.-H., Pawlyk, B. S., Shang, J., Sandberg, M. A., Berson, E. L., Li, T. A retinitis pigmentosa GTPase regulator (RPGR)-deficient mouse model for X-linked retinitis pigmentosa (RP3). Proc. Nat. Acad. Sci. 97: 3649-3654, 2000. [PubMed: 10725384, images, related citations] [Full Text]

  22. Hong, D-H., Pawlyk, B., Sokolov, M., Strissel, K. J., Yang, Y., Tulloch, B., Wright, A. F., Arshavsky, V. Y., Li, T. RPGR isoforms in photoreceptor connecting cilia and the transitional zone of motile cilia. Invest. Ophthal. Vis. Sci. 44: 2413-2421, 2003. [PubMed: 12766038, related citations] [Full Text]

  23. Iannaccone, A., Breuer, D. K., Wang, X. F., Kuo, S. F., Normando, E. M., Filippova, E., Baldi, A., Hiriyanna, S., MacDonald, C. B., Baldi, F., Cosgrove, D., Morton, C. C., Swaroop, A., Jablonski, M. M. Clinical and immunohistochemical evidence for an X linked retinitis pigmentosa syndrome with recurrent infections and hearing loss in association with an RPGR mutation. (Letter) J. Med. Genet. 40: e118, 2003. Note: Electronic Article. [PubMed: 14627685, related citations] [Full Text]

  24. Khanna, H., Hurd, T. W., Lillo, C., Shu, X., Parapuram, S. K., He, S., Akimoto, M., Wright, A. F., Margolis, B., Williams, D. S., Swaroop, A. RPGR-ORF15, which is mutated in retinitis pigmentosa, associates with SMC1, SMC3, and microtubule transport proteins. J. Biol. Chem. 280: 33580-33587, 2005. [PubMed: 16043481, images, related citations] [Full Text]

  25. Kirschner, R., Erturk, D., Zeitz, C., Sahin, S., Ramser, J., Cremers, F. P. M., Ropers, H.-H., Berger, W. DNA sequence comparison of human and mouse retinitis pigmentosa GTPase regulator (RPGR) identifies tissue-specific exons and putative regulatory elements. Hum. Genet. 109: 271-278, 2001. [PubMed: 11702207, related citations] [Full Text]

  26. Kirschner, R., Rosenberg, T., Schultz-Heienbrok, R., Lenzner, S., Feil, S., Roepman, R., Cremers, F. P. M., Ropers, H.-H., 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. Molec. Genet. 8: 1571-1578, 1999. [PubMed: 10401007, related citations] [Full Text]

  27. Mavlyutov, T. A., Zhao, H., Ferreira, P. A. Species-specific subcellular localization of RPGR and RPGRIP isoforms: implications for the phenotypic variability of congenital retinopathies among species. Hum. Molec. Genet. 11: 1899-1907, 2002. [PubMed: 12140192, related citations] [Full Text]

  28. McGuire, R. E., Sullivan, L. S., Blanton, S. H., Church, M. W., Heckenlively, J. R., Daiger, S. P. X-linked dominant cone-rod degeneration: linkage mapping of a new locus for retinitis pigmentosa (RP15) to Xp22.13-p22.11. Am. J. Hum. Genet. 57: 87-94, 1995. [PubMed: 7611300, related citations]

  29. Mears, A. J., Hiriyanna, S., Vervoort, R., Yashar, B., Gieser, L., Fahrner, S., Daiger, S. P., Heckenlively, J. R., Sieving, P. A., Wright, A. F., Swaroop, A. Remapping of the RP15 locus for X-linked cone-rod degeneration to Xp11.4-p21.1, and identification of a de novo insertion in the RPGR exon ORF15. Am. J. Hum. Genet. 67: 1000-1003, 2000. [PubMed: 10970770, images, related citations] [Full Text]

  30. Meindl, A., Dry, K., Herrmann, K., Manson, F., Ciccodicola, A., Edgar, A., Carvalho, M. R. S., 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). Nature Genet. 13: 35-42, 1996. [PubMed: 8673101, related citations] [Full Text]

  31. Miano, M. G., Testa, F., Strazzullo, M., Trujillo, M., De Bernardo, C., Grammatico, B., Simonelli, F., Mangino, M., Torrente, I., Ruberto, G., Beneyto, M., Antinolo, G., Rinaldi, E., Danesino, C., Ventruto, V., D'Urso, M., Ayuso, C., Baiget, M., Ciccodicola, A. Mutation analysis of the RPGR gene reveals novel mutations in south European patients with X-linked retinitis pigmentosa. Europ. J. Hum. Genet. 7: 687-694, 1999. [PubMed: 10482958, related citations] [Full Text]

  32. Moore, A., Escudier, E., Roger, G., Tamalet, A., Pelosse, B., Marlin, S., Clement, A., Geremek, M., Delaisi, B., Bridoux, A.-M., 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. 43: 326-333, 2006. [PubMed: 16055928, images, related citations] [Full Text]

  33. Murga-Zamalloa, C. A., Atkins, S. J., Peranen, J., Swaroop, A., Khanna, H. Interaction of retinitis pigmentosa GTPase regulator (RPGR) with RAB8A GTPase: implications for cilia dysfunction and photoreceptor degeneration. Hum. Molec. Genet. 19: 3591-3598, 2010. [PubMed: 20631154, images, related citations] [Full Text]

  34. Neidhardt, J., Glaus, E., Barthelmes, D., Zeitz, C., Fleischhauer, J., Berger, W. Identification and characterization of a novel RPGR isoform in human retina. Hum. Mutat. 28: 797-807, 2007. [PubMed: 17405150, related citations] [Full Text]

  35. Nishiguchi, K. M., Tearle, R. G., Liu, Y. P., Oh, E. C., Miyake, N., Benaglio, P., Harper, S., Koskiniemi-Kuendig, H., Venturini, G., Sharon, D., Koenekoop, R. K., Nakamura, M., and 10 others. Whole genome sequencing in patients with retinitis pigmentosa reveals pathogenic DNA structural changes and NEK2 as a new disease gene. Proc. Nat. Acad. Sci. 110: 16139-16144, 2013. [PubMed: 24043777, images, related citations] [Full Text]

  36. Pelletier, V., Jambou, M., Delphin, N., Zinovieva, E., Stum, M., Gigarel, N., Dollfus, H., Hamel, C., Toutain, A., Dufier, J.-L., Roche, O., Munnich, A., Bonnefont, J.-P., Kaplan J., Rozet, J.-M. Comprehensive study of mutations in RP2 and RPGR in patients affected with distinct retinal dystrophies: genotype-phenotype correlations and impact on genetic counseling. Hum. Mutat. 28: 81-91, 2007. [PubMed: 16969763, related citations] [Full Text]

  37. Roepman, R., Bernoud-Hubac, N., Schick, D. E., Maugeri, A., Berger, W., Ropers, H.-H., Cremers, F. P. M., Ferreira, P. A. The retinitis pigmentosa GTPase regulator (RPGR) interacts with novel transport-like proteins in the outer segments of rod photoreceptors. Hum. Molec. Genet. 9: 2095-2105, 2000. [PubMed: 10958648, related citations] [Full Text]

  38. Roepman, R., van Duijnhoven, G., Rosenberg, T., Pinckers, A. J. L. G., Bleeker-Wagemakers, L. M., Bergen, A. A. B., Post, J., Beck, A., Reinhardt, R., Ropers, H.-H., Cremers, F. P. M., Berger, W. Positional cloning of the gene for X-linked retinitis pigmentosa 3: homology with the guanine-nucleotide-exchange factor RCC1. Hum. Molec. Genet. 5: 1035-1041, 1996. [PubMed: 8817343, related citations] [Full Text]

  39. Rozet, J.-M., Perrault, I., Gigarel, N., Souied, E., Ghazi, I., Gerber, S., Dufier, J.-L., Munnich, A., Kaplan, J. Dominant X linked retinitis pigmentosa is frequently accounted for by truncating mutations in exon ORF15 of the RPGR gene. J. Med. Genet. 39: 284-285, 2002. [PubMed: 11950860, related citations] [Full Text]

  40. Sandberg, M. A., Rosner, B., Weigel-DiFranco, C., Dryja, T. P., Berson, E. L. Disease course of patients with X-linked retinitis pigmentosa due to RPGR gene mutations. Invest. Ophthal. Vis. Sci. 48: 1298-1304, 2007. [PubMed: 17325176, related citations] [Full Text]

  41. Sharon, D., Sandberg, M. A., Rabe, V. W., Stillberger, M., Dryja, T. P., Berson, E. L. RP2 and RPGR mutations and clinical correlations in patients with X-linked retinitis pigmentosa. Am. J. Hum. Genet. 73: 1131-1146, 2003. [PubMed: 14564670, images, related citations] [Full Text]

  42. Shu, X., Black, G. C., Rice, J. M., Hart-Holden, N., Jones, A., O'Grady, A., Ramsden, S., Wright, A. F. RPGR mutation analysis and disease: an update. Hum. Mutat. 28: 322-328, 2007. [PubMed: 17195164, related citations] [Full Text]

  43. Shu, X., Fry, A. M., Tulloch, B., Manson, F. D. C., Crabb, J. W., Khanna, H., Faragher, A. J., Lennon, A., He, S., Trojan, P., Giessl, A., Wolfrum, U., Vervoort, R., Swaroop, A., Wright, A. F. RPGR ORF15 isoform co-localizes with RPGRIP1 at centrioles and basal bodies and interacts with nucleophosmin. Hum. Molec. Genet. 14: 1183-1197, 2005. [PubMed: 15772089, related citations] [Full Text]

  44. Shu, X., Zeng, Z., Gautier, P., Lennon, A., Gakovic, M., Patton, E. E., Wright, A. F. Zebrafish Rpgr is required for normal retinal development and plays a role in dynein-based retrograde transport processes. Hum. Molec. Genet. 19: 657-670, 2010. [PubMed: 19955120, related citations] [Full Text]

  45. Souied, E., Segues, B., Ghazi, I., Rozet, J.-M., Chatelin, S., Gerber, S., Perrault, I., Michel-Awad, A., Briard, M.-L., Plessis, G., Dufier, J.-L., Munnich, A., Kaplan, J. Severe manifestations in carrier females in X linked retinitis pigmentosa. J. Med. Genet. 34: 793-797, 1997. [PubMed: 9350809, related citations] [Full Text]

  46. van Dorp, D. B., Wright, A. F., Carothers, A. D., Bleeker-Wagemakers, E. M. A family with RP3 type of X-linked retinitis pigmentosa: an association with ciliary abnormalities. Hum. Genet. 88: 331-334, 1992. [PubMed: 1733835, related citations] [Full Text]

  47. Vervoort, R., Lennon, A., Bird, A. C., Tulloch, B., Axton, R., Miano, M. G., Meindl, A., Meitinger, T., Ciccodicola, A., Wright, A. F. Mutational hot spot within a new RPGR exon in X-linked retinitis pigmentosa. Nature Genet. 25: 462-466, 2000. [PubMed: 10932196, related citations] [Full Text]

  48. Vervoort, R., Wright, A. F. Mutations of RPGR in X-linked retinitis pigmentosa (RP3). Hum. Mutat. 19: 486-500, 2002. [PubMed: 11968081, related citations] [Full Text]

  49. Yang, Z., Peachey, N. S., Moshfeghi, D. M., Thirumalaichary, S., Chorich, L., Shugart, Y. Y., Fan, K., Zhang, K. Mutations in the RPGR gene cause X-linked cone dystrophy. Hum. Molec. Genet. 11: 605-611, 2002. [PubMed: 11875055, related citations] [Full Text]

  50. Yokoyama, A., Maruiwa, F., Hayakawa, M., Kanai, A., Vervoort, R., Wright, A. F., Yamada, K., Niikawa, N., Naoi, N. Three novel mutations of the RPGR gene exon ORF15 in three Japanese families with X-linked retinitis pigmentosa. Am. J. Med. Genet. 104: 232-238, 2001. [PubMed: 11754050, related citations]

  51. Zeiss, C. J., Ray, K., Acland, G. M., Aguirre, G. D. Mapping of X-linked progressive retinal atrophy (XLPRA), the canine homolog of retinitis pigmentosa 3 (RP3). Hum. Molec. Genet. 9: 531-537, 2000. [PubMed: 10699176, related citations] [Full Text]

  52. Zhang, Q., Acland, G. M., Wu, W. X., Johnson, J. L., Pearce-Kelling, S., Tulloch, B., Vervoort, R., Wright, A. F., Aguirre, G. D. Different RPGR exon ORF15 mutations in Canids provide insights into photoreceptor cell degeneration. Hum. Molec. Genet. 11: 993-1003, 2002. [PubMed: 11978759, related citations] [Full Text]

  53. Zito, I., Downes, S. M., Patel, R. J., Cheetham, M. E., Ebenezer, N. D., Jenkins, S. A., Bhattacharya, S. S., Webster, A. R., Holder, G. E., Bird, A. C., Bamiou, D. E., Hardcastle, A. J. RPGR mutation associated with retinitis pigmentosa, impaired hearing, and sinorespiratory infections. J. Med. Genet. 40: 609-615, 2003. [PubMed: 12920075, related citations] [Full Text]


Jane Kelly - updated : 4/7/2016
Marla J. F. O'Neill - updated : 12/13/2013
Jane Kelly - updated : 5/22/2013
Patricia A. Hartz - updated : 4/26/2012
Patricia A. Hartz - updated : 8/31/2011
George E. Tiller - updated : 2/8/2011
George E. Tiller - updated : 11/12/2010
George E. Tiller - updated : 5/19/2008
Jane Kelly - updated : 4/16/2008
Marla J. F. O'Neill - updated : 2/1/2008
Jane Kelly - updated : 12/5/2007
Jane Kelly - updated : 11/20/2007
Cassandra L. Kniffin - updated : 10/9/2007
Cassandra L. Kniffin - updated : 5/29/2007
Victor A. McKusick - updated : 3/28/2007
Jane Kelly - updated : 9/13/2006
Marla J. F. O'Neill - updated : 7/6/2006
Jane Kelly - updated : 11/18/2005
Jane Kelly - updated : 6/2/2005
Cassandra L. Kniffin - updated : 5/13/2005
Jane Kelly - updated : 6/14/2004
Cassandra L. Kniffin - updated : 1/16/2004
Victor A. McKusick - updated : 12/12/2003
Jane Kelly - updated : 10/27/2003
Victor A. McKusick - updated : 10/1/2003
Jane Kelly - updated : 8/22/2003
George E. Tiller - updated : 7/9/2003
George E. Tiller - updated : 12/12/2002
George E. Tiller - updated : 10/9/2002
Victor A. McKusick - updated : 10/1/2002
Victor A. McKusick - updated : 6/11/2002
Victor A. McKusick - updated : 5/14/2002
Victor A. McKusick - updated : 4/12/2002
Victor A. McKusick - updated : 12/4/2001
Victor A. McKusick - updated : 10/12/2001
George E. Tiller - updated : 12/4/2000
Victor A. McKusick - updated : 10/19/2000
Victor A. McKusick - updated : 10/19/2000
Ada Hamosh - updated : 8/1/2000
Jane Kelly - updated : 6/28/2000
Victor A. McKusick - updated : 4/20/2000
George E. Tiller - updated : 4/14/2000
Victor A. McKusick - updated : 11/8/1999
Ada Hamosh - updated : 8/18/1999
Victor A. McKusick - updated : 2/25/1999
Michael J. Wright - updated : 6/5/1998
Victor A. McKusick - updated : 5/12/1998
Victor A. McKusick - updated : 2/25/1998
Victor A. McKusick - updated : 10/7/1997
Moyra Smith - updated : 8/21/1996
Iosif W. Lurie - updated : 7/4/1996
Moyra Smith - updated : 4/26/1996
Creation Date:
Victor A. McKusick : 10/12/1987
carol : 01/13/2023
alopez : 03/15/2022
carol : 01/08/2019
carol : 04/07/2016
carol : 11/19/2014
joanna : 1/22/2014
carol : 12/17/2013
mcolton : 12/13/2013
tpirozzi : 7/3/2013
carol : 5/22/2013
mgross : 5/2/2012
mgross : 5/2/2012
terry : 4/26/2012
mgross : 9/9/2011
terry : 8/31/2011
wwang : 3/14/2011
carol : 2/15/2011
terry : 2/8/2011
wwang : 11/19/2010
terry : 11/12/2010
alopez : 10/14/2010
joanna : 7/27/2010
alopez : 7/17/2009
alopez : 7/15/2009
alopez : 7/15/2009
alopez : 7/14/2009
carol : 3/9/2009
alopez : 2/16/2009
wwang : 10/30/2008
wwang : 5/22/2008
terry : 5/19/2008
carol : 4/16/2008
wwang : 2/5/2008
terry : 2/1/2008
carol : 12/5/2007
carol : 11/20/2007
wwang : 10/17/2007
ckniffin : 10/9/2007
carol : 7/18/2007
wwang : 6/11/2007
ckniffin : 5/29/2007
alopez : 4/3/2007
terry : 3/28/2007
carol : 2/28/2007
carol : 9/13/2006
wwang : 7/6/2006
wwang : 2/24/2006
alopez : 11/18/2005
tkritzer : 6/2/2005
tkritzer : 5/31/2005
ckniffin : 5/13/2005
terry : 7/19/2004
alopez : 6/14/2004
tkritzer : 2/18/2004
ckniffin : 1/16/2004
cwells : 12/19/2003
terry : 12/12/2003
ckniffin : 12/4/2003
tkritzer : 11/6/2003
tkritzer : 10/28/2003
tkritzer : 10/27/2003
tkritzer : 10/23/2003
tkritzer : 10/23/2003
tkritzer : 10/10/2003
tkritzer : 10/1/2003
carol : 8/22/2003
cwells : 7/9/2003
terry : 5/15/2003
tkritzer : 1/3/2003
tkritzer : 1/2/2003
cwells : 12/12/2002
cwells : 10/9/2002
carol : 10/2/2002
tkritzer : 10/1/2002
tkritzer : 10/1/2002
alopez : 6/18/2002
terry : 6/11/2002
alopez : 5/17/2002
terry : 5/14/2002
alopez : 4/19/2002
alopez : 4/17/2002
terry : 4/12/2002
carol : 1/2/2002
mcapotos : 12/10/2001
terry : 12/4/2001
mcapotos : 10/30/2001
mcapotos : 10/12/2001
carol : 12/4/2000
mcapotos : 11/28/2000
terry : 11/17/2000
carol : 11/6/2000
carol : 10/19/2000
terry : 10/19/2000
alopez : 8/2/2000
terry : 8/1/2000
alopez : 6/28/2000
mcapotos : 5/11/2000
mcapotos : 5/9/2000
terry : 4/20/2000
terry : 4/14/2000
carol : 3/13/2000
alopez : 11/12/1999
terry : 11/8/1999
alopez : 8/20/1999
terry : 8/18/1999
carol : 5/5/1999
carol : 3/9/1999
terry : 2/25/1999
dkim : 9/10/1998
alopez : 6/17/1998
terry : 6/5/1998
carol : 5/26/1998
alopez : 5/21/1998
terry : 5/12/1998
joanna : 5/7/1998
mark : 2/25/1998
mark : 2/25/1998
mark : 2/23/1998
terry : 12/11/1997
mark : 10/8/1997
terry : 10/7/1997
mark : 11/4/1996
jamie : 10/23/1996
jamie : 10/16/1996
mark : 8/21/1996
terry : 8/21/1996
mark : 8/20/1996
carol : 7/4/1996
carol : 6/27/1996
carol : 4/26/1996
mark : 1/10/1996
terry : 1/5/1996
carol : 11/18/1994
davew : 7/5/1994
mimadm : 2/28/1994
carol : 6/7/1993
carol : 1/27/1993
carol : 1/19/1993

* 312610

RETINITIS PIGMENTOSA GTPase REGULATOR; RPGR


HGNC Approved Gene Symbol: RPGR

Cytogenetic location: Xp11.4     Genomic coordinates (GRCh38): X:38,269,163-38,327,509 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xp11.4 Cone-rod dystrophy, X-linked, 1 304020 X-linked recessive 3
Macular degeneration, X-linked atrophic 300834 X-linked recessive 3
Retinitis pigmentosa 3 300029 X-linked 3
Retinitis pigmentosa, X-linked, and sinorespiratory infections, with or without deafness 300455 X-linked 3

TEXT

Cloning and Expression

Meindl et al. (1996) isolated and sequenced cosmids from the region of the microdeletions in patients with retinitis pigmentosa-3 (300029) and used these cosmids to make exon predictions. They thus identified a gene, provisionally named RPGR (retinitis pigmentosa GTPase regulator), which gives rise to a ubiquitously expressed 29-kb transcript. The predicted 90-kD RPGR protein contains in its N-terminal half a tandem repeat structure highly similar to the regulator of chromosome condensation (RCC1; 179710), which regulates the GTPase RAN (601179). Meindl et al. (1996) identified 8 potential asparagine-linked glycosylation sites along the N-terminal two-thirds of the predicted RPGR protein. The C terminus of the protein contains a cluster of basic residues followed by a consensus isoprenylation site. The authors noted that confirmation of the isoprenylation of this site would establish a novel means of membrane anchorage for a GTPase regulator.

Roepman et al. (1996) used a cosmid spanning a microdeletion in an RP3 patient to screen cDNA libraries. They then isolated additional cosmids that flanked the microdeletion region. Shotgun cosmid sequencing enabled them to sequence 32,895 bp of DNA. Computer-assisted analysis of this sequence predicted numerous additional exons which were confirmed by cDNA cloning. Sequence comparisons revealed that the deduced product of the RPGR gene showed strong similarity with RCC1.

Kirschner et al. (1999) studied the expression of the RPGR gene by Northern blot hybridization, cDNA library screening, and RT-PCR in various organs of mouse and human and identified at least 12 alternatively spliced isoforms. Some of the transcripts are tissue-specific and contain novel exons, which elongate or truncate the previously reported open reading frame of the mouse and human RPGR gene. Kirschner et al. (1999) identified a new exon, designated exon 15A by them, which is expressed exclusively in human retina and mouse eye and contains a premature stop codon. The deduced polypeptide lacked 169 amino acids from the C terminus of the ubiquitously expressed variant, including an isoprenylation site.

Kirschner et al. (2001) compared the genomic sequence of the human and mouse RPGR genes. Each spans a region of nearly 59 kb, and all previously identified exons are conserved between the 2 species. A sequence comparison identified 28 conserved sequence elements in introns, upstream of exon 1, within the promoter region, and downstream of the most 3-prime exon. Some of the intronic conserved sequence elements flank tissue-specific exons and therefore may represent important regulatory elements for alternative splicing. Comparative Northern blot hybridization of ubiquitous and tissue-specific RPGR probes identified high molecular mass transcripts with similar expression patterns in both human and mouse. These transcripts range from 6 to 15 kb and suggest the presence of additional transcribed sequences within RPGR.

Vervoort et al. (2000) sequenced a 172-kb region containing the entire RPGR gene. Analysis of the sequence disclosed a novel 3-prime terminal exon that was mutated in 60% of XLRP patients examined. This exon encodes 567 amino acids, with a repetitive domain rich in glutamic acid residues. The sequence is conserved in the mouse, bovine, and Fugu rubripes genes. It is preferentially expressed in mouse and bovine retina, further supporting its importance for retinal function. In addition to the 19 exons previously reported, Vervoort et al. (2000) found 5 additional exons: 15b1, 15b2, 15a, ORF14, and ORF15. Exons 15b1 and 15b2 are 2 overlapping exons within intron 15 that use alternative acceptor splice sites and the same donor site. Their inclusion is predicted to result in premature termination of translation. Exon 15a, which is a third internal exon within intron 15 and also encodes a premature stop codon, is identical with exon 15a reported previously (Kirschner et al., 1999). Exon ORF14 corresponds to the mouse exon 14-14a-15 (Kirschner et al., 1999); its inclusion does not disrupt the reading frame. Exon ORF15 is a large 3-prime terminal exon consisting of exon 15 and extending into part of intron 15. The predicted exon ORF15 protein domain contains an unusual region of low sequence complexity with high glutamic acid and glycine content (Plaid domain).

By RT-PCR analysis of human retina cDNA, Neidhardt et al. (2007) identified a novel exon in intron 9, designated exon 9A, of the RPGR gene. The novel exon starts 418 bp downstream of the splice donor site of exon 9 and contains 136 nucleotides. Exon 9A is present in a novel RPGR splice variant that truncates the RCC1 homologous protein domain of RPGR by 48 amino acids, yielding a 353-residue protein with a molecular mass of about 45 kD. There was strong expression of the 9A variant predominantly in the inner segment of human cones with a weaker signal in rods. Immunoprecipitation studies showed that the 9A variant binds distinct RPGRIP1 (605446) variants, suggesting functional relevance. The expression of mRNA containing exon 9A was quantified to approximately 4% of RPGR in normal individuals.

Ghosh et al. (2010) noted that there are 2 major isoforms of RPGR detected in the retina: RPGR(1-19), which has 19 exons and 815 amino acids, and RPGR(1-ORF15), which has 15 exons plus part of intron 15 and 1,152 amino acids. Both isoforms share exons 1 through 15 and N-terminal RCC1-like domain (RLD), whereas RPGR(1-ORF15) has a glu-gly-rich C-terminal domain.

Using immunogold electron microscopy with mouse and human retina, Khanna et al. (2005) found that, in addition to axoneme, RPGR-ORF15 localized to basal bodies of photoreceptor connecting cilium. It also colocalized with acetylated alpha-tubulin (see 602529) at the tip and tail axoneme of mouse sperm flagella and at primary cilia of MDCK canine kidney cells.


Gene Function

Boylan and Wright (2000) and Roepman et al. (2000) identified the RPGRIP1 gene (605446) and found that it encodes several alternatively spliced gene products that interact specifically with RPGR. Roepman et al. (2000) determined that RPGR and RPGRIP1 colocalized in the outer segment of rod photoreceptors, which is in agreement with the retinitis pigmentosa phenotype observed in RP3 patients.

Mavlyutov et al. (2002) used isoform-specific antibodies to demonstrate that RPGR and RPGRIP isoforms are distributed and colocalized at restricted foci throughout the outer segments of human and bovine (but not murine) rod photoreceptors. In humans, these proteins are also localized in cone outer segments, and RPGRIP is expressed in other neurons such as amacrine cells. The authors proposed the existence of species-specific subcellular processes governing the function and/or organization of the photoreceptor outer segment as reflected by the species-specific localization of RPGR and RPGRIP protein isoforms in this compartment. They contended that this may provide a rationale for the disparity of phenotypes among species and among various human mutations.

Using immunofluorescence and serial retinal sectioning, Hong et al. (2003) established the subcellular localization of RPGR in mice and other mammalian species; they found RPGR in connecting cilia of rods and cones and in a homologous structure, the transitional zone of motile cilia in airway epithelia. There was no evidence for species-specific variation of RPGR localization.

By immunohistochemistry, Iannaccone et al. (2003) found that the RPGR gene was expressed throughout the outer segments of human rod and cone photoreceptors, within the epithelial lining of human bronchial and sinus tissues, and in human fetal cochlea, including the stria vascularis, the suprastrial cells, and the apical portion of the spiral limbus. The findings broadened the possible functional roles of RPGR and provided evidence for a broad phenotype in patients with RPGR mutations.

Hong et al. (2004) created an RPGR transgene transcript using alternative splicing involving the purine-rich region of the ORF15 exon, generating a shortened mRNA and a premature stop codon. This truncation mutant caused more rapid photoreceptor degeneration than that in the RPGR null mutant. The disease course was similar, whether the transgene was coexpressed with wildtype RPGR or expressed alone on the RPGR null background. The authors concluded that certain truncated forms of RPGR can behave as a dominant, gain-of-function mutant. These data suggested that human RPGR mutations were not necessarily null and some might also act as dominant alleles, leading to a more severe phenotype than a null mutant.

RPGR, which is essential for the maintenance of photoreceptor viability, is expressed as constitutive and ORF15 variants because of alternative splicing. Hong et al. (2005) examined whether the retina-specific ORF15 variant alone could substantially substitute for RPGR function and tested whether the highly repetitive purine-rich region of ORF15 could be abbreviated without ablating the function, so as to accommodate RPGR replacement genes in adeno-associated virus (AAV) vectors. A cDNA representing mouse Rpgr-ORF15 but shortened by 654 bp in the repetitive region was placed under the control of a chicken beta-actin (CBA) hybrid promoter and used to create transgenic chimeras that were crossed with Rpgr knockout mice. In mice expressing the transgene but null for endogenous Rpgr, transgenic Rpgr-ORF15 was found in the connecting cilia of rod and cone photoreceptors at approximately 20% of the wildtype level. Photoreceptor morphology, cone opsin localization, expression of GFAP (137780) (a marker of retinal degeneration), and ERGs were consistent with the transgene exerting substantial rescue of retinal degeneration due to loss of endogenous Rpgr. Hong et al. (2005) concluded that RPGR-ORF15 is the functionally significant variant in photoreceptors and that the length of its repetitive region can be reduced while preserving its function.

In cultured mammalian cells, Shu et al. (2005) showed that both RPGR-ORF15 and RPGRIP1 (605446) localized to centrioles. By multiple methods, the authors showed that the C2 domain of RPGR-ORF15 interacted with the shuttling protein nucleophosmin (NPM1; 164040), which is a protein chaperone that shuttles between the nucleoli and the cytoplasm and has been associated with licensing of centrosomal division.

By immunoprecipitation of bovine retinal axoneme-enriched fraction, followed by mass spectrometric analysis of proteins separated by SDS-PAGE, Khanna et al. (2005) found that Rpgr-Orf15 bound the ATP-binding proteins Smc1 (SMC1A; 300040) and Smc3 (606062). Protein pull-down experiments revealed that the N-terminal RLD domain of human RPGR-ORF15 bound endogenous bovine Smc1 and Smc3. Bovine Rpgr-Orf15 also associated with Ift88 (600595) and the microtubule motor proteins Kif3a (604683) and Kap3 (KIFAP3; 601836) in an axoneme multiprotein complex. Khanna et al. (2005) concluded that RPGR-ORF15 may be involved in microtubule organization or regulation of transport in primary cilia.

By coimmunoprecipitation of bovine retinal extract, Murga-Zamalloa et al. (2010) found that Rpgr interacted with the small GTPase Rab8a (165040), which has a role in cilia biogenesis and maintenance. Human RPGR interacted predominantly with the GDP-bound form of human RAB8A and stimulated GDP/GTP exchange. Disease-causing mutations in RPGR diminished its interaction with RAB8A and/or reduced its GDP/GTP exchange activity. Depletion of RPGR in human retinal pigment epithelial cells disrupted association of RAB8A with cilia and resulted in shortened primary cilia.


Molecular Genetics

Shu et al. (2007) stated that a total of 240 different mutations in the RPGR gene had been reported, including 24 novel mutations identified by the authors. Of the 240 mutations, 95% are associated with XLRP, 3% with cone, cone-rod dystrophy or atrophic macular atrophy, and 2% with syndromal retinal dystrophies with ciliary dyskinesia and hearing loss.

Branham et al. (2012) screened 214 male patients with simplex retinal degenerative disease, 185 with RP and 29 with cone/cone-rod dystrophy (COD/CORD), for mutations in the RPGR and RP2 (300757) genes. They identified pathogenic mutations in 32 (15%) of the patients. Four patients with COD/CORD had a mutation in the ORF15 mutation hotspot of the RPGR gene. Of the RP patients, 3 had mutations in RP2 and 25 had mutations in RPGR (including 23 in the ORF15 region). Branham et al. (2012) concluded that their results demonstrated a substantial contribution of RPGR mutations to retinal degenerations, and in particular to simplex RP. They suggested that RPGR should be considered as a first tier gene for screening isolated males with retinal degeneration.

Retinitis Pigmentosa 3

Meindl et al. (1996) provided evidence that loss-of-function mutations within RPGR are responsible for X-linked retinitis pigmentosa-3 by identifying 2 small intragenic deletions and 2 nonsense and 3 missense mutations in highly conserved residues in unrelated patients with X-linked RP.

Roepman et al. (1996) screened the Xp21.1-p11.4 RP3 locus interval for microdeletions using a novel technique they called YAC representation hybridization (YRH). Application of this technique led to the generation of a defined amplifiable subset of restriction fragments representing the insert of a YAC spanning the region of interest. The mixture of PCR products was used to study Southern blots of restriction-digested genomic DNA. In 1 of 30 patients with X-linked retinitis pigmentosa, they detected a 6.4-kb microdeletion.

Roepman et al. (1996) detected mutations in the RPGR gene in RP patients and not in controls. Mutation screening was carried out in 28 patients by means of SSCP analysis. They designed intron primers for PCR amplification of 10 exons and detected 5 bandshifts in patients. The corresponding PCR fragments were sequenced and 3 different nucleotide exchanges (312610.0001, 312610.0002, and 312610.0003), and one 4-bp deletion (312610.0004) were identified. None of these changes were detected in 84 male controls. The 6 most 3-prime exons showed no mutations but did reveal several polymorphisms. The 3-prime end of the gene is, however, disrupted by the 6.4-kb deletion which is present in a patient with X-linked retinitis pigmentosa. Roepman et al. (1996) noted that the 5-prime end of the gene and the promoter region had not yet been cloned.

To characterize RPGR mutations in a systematic way, Fujita et al. (1997) identified 11 RP3 families by haplotype analysis. Sequence analysis of the PCR-amplified genomic DNA from patients representing these RP3 families showed no causative mutation in RPGR exons 2 to 19, spanning more than 98% of the coding region. In patients from 2 families, however, they identified transition mutations in the intron region near splice sites (IVS10+3; 312610.0005 and IVS13-8). RNA analysis showed that both splice site mutations resulted in the generation of aberrant RPGR transcripts. The results supported the hypothesis that mutations in the RPGR gene are not a common defect in the RP3 subtype of X-linked RP and that the majority of causative mutations may reside either in as yet unidentified RPGR exons or in another nearby gene at Xp21.1.

Buraczynska et al. (1997) examined the RPGR gene in a cohort of 80 affected males from apparently unrelated X-linked RP families by direct sequencing of the PCR-amplified products from genomic DNA. Fifteen different putative disease-causing mutations were identified in 17 of the 80 families: 4 nonsense mutations, 1 missense mutation, 6 microdeletions, and 4 intronic-sequence substitutions resulting in splice defects. In their Figure 2, they mapped the location of 12 mutations reported by Meindl et al. (1996) and Roepman et al. (1996) and the 15 different mutations identified in this study. Most of the mutations were detected in the conserved N-terminal region of the RPGR protein, containing tandem repeats homologous to those present in the RCC1 protein. In agreement with previous studies, they were able to demonstrate RPGR mutations in only about 20% of the examined X-linked RP patients. On the other hand, the RP3 subtype consistently accounts for 60 to 90% of families localized by linkage and haplotype genotyping. Buraczynska et al. (1997) raised the possibility that the RPGR gene contains unidentified mutation hotspots in sequences that have not been screened, such as the promoter region or intronic sequences and exon 1. The authors could not rule out the alternative possibility of another gene located in proximity to RPGR at Xp21.1 that also causes RP when mutated.

Souied et al. (1997) described 9 families that showed an X-linked pattern of inheritance with a total of 28 affected males and 34 affected females. The females in these families met criteria for the diagnosis of retinitis pigmentosa. The males had a delayed onset of disease, in the second decade, with central vision being preserved until 40 to 45 years of age. Linkage to the RP3 locus was demonstrated, but SSCP and sequence analysis of the RPGR gene demonstrated no mutations. Souied et al. (1997) suggested that these families demonstrated an X-linked dominant form of RP and that the negative mutation results may be explained either by allelic heterogeneity at the RP3 locus or involvement of a distinct locus mapping close to RP3.

Kirschner et al. (1999) demonstrated that the novel RPGR exon 15A that they identified was deleted in a family with X-linked RP (312610.0008). Kirschner et al. (1999) concluded that their results indicate tissue-dependent regulation of alternative splicing of the RPGR gene and that the presence of the retina-specific transcript may explain why phenotypic aberrations in RP3 are confined to the eye.

Miano et al. (1999) found a total of 29 different RPGR mutations identified in northern European and United States patients. They performed mutation analysis of the RPGR gene in a cohort of 49 southern European males with XLRP. By multiplex SSCA and direct sequencing of all 19 RPGR exons, 7 different mutations, all novel, were identified in 8 of the 49 families; these included 3 splice site mutations, 2 microdeletions, and 2 missense mutations. RNA analysis showed that the 3 splice site defects resulted in the generation of aberrant RPGR transcripts. Six of these mutations were detected in the conserved N-terminal region of RPGR protein, containing tandem repeats homologous to repeats within the RCC1 protein (179710). Strikingly, none of the RPGR mutations reported in other populations were identified in this series.

Because mutations in the RPGR gene were found in fewer than the 70 to 75% of XLRP patients predicted from linkage studies, Vervoort et al. (2000) hypothesized that mutations in the remaining XLRP patients may have resided in undiscovered exons of RPGR and sequenced a 172-kb region containing the entire gene. Analysis of the sequence disclosed a new 3-prime terminal exon, ORF15, that was mutated in 60% of XLRP patients examined. Vervoort et al. (2000) concluded that mutations in RPGR are the only cause of RP3 type XLRP and account for the disease in over 70% of XLRP patients and an estimated 11% of all RP patients. Vervoort et al. (2000) found 28 XLRP patients with mutations in exon ORF15, each of which leads to premature termination of translation. Most were small nucleotide deletions, and 5 of them were substitutions leading to nonsense mutation. None of the mutations were detected in 150 control chromosomes. The high frequency of mutations within the terminal exon ORF15 (17 different mutations in 1 kb) compared with other parts of the same RPGR transcript (6 mutations in 1.6 kb), suggested that it is a mutation hotspot. Vervoort et al. (2000) found each of 5 different mutations on at least 2 different haplotypes, indicating recurrent mutation.

In each of 3 unrelated Japanese families segregating X-linked retinitis pigmentosa, Yokoyama et al. (2001) identified a novel mutation in exon ORF15. The mutations were of insertion/deletion type and were predicted to lead to a frameshift, resulting in a truncated protein. The findings supported the previous hypothesis that exon ORF15 is a mutation hotspot. Affected males had typical retinitis pigmentosa, whereas the obligate carrier females showed a wide clinical spectrum, ranging from minor symptoms to severe visual disability. Some carrier females showed typical RP, and most carriers manifested high myopia and astigmatism, with insufficiently corrected visual acuity.

In 4 of the 9 families with XLRP reported by Souied et al. (1997), Rozet et al. (2002) identified mutations in exon ORF15 of the RPGR gene. Rozet et al. (2002) also reported 5 additional affected families with mutations in ORF15. All 7 of the identified mutations were predicted to result in a truncated protein. Rozet et al. (2002) noted that the age at onset in affected females was delayed compared to affected males (20 to 40 years vs 10 to 20 years, respectively).

Vervoort and Wright (2002) reviewed mutations in RPGR in X-linked retinitis pigmentosa (RP3). They commented on the fact that exon ORF15 is a hotspot for mutation, at least in the British population, in which it harbors 80% of the mutations found within a sample of 47 X-linked retinitis pigmentosa patients.

In a North American cohort of 234 families with RP, Breuer et al. (2002) conducted a comprehensive screen of the RP2 (300757) and RPGR (including ORF15) genes and their 5-prime upstream regions. Of these families, 91 (39%) showed definitive X-linked inheritance, an additional 88 (38%) revealed a pattern consistent with X-linked disease, and the remaining 55 (23%) were simplex male patients with RP who had an early onset and/or severe disease. In agreement with previous studies, they showed that mutations in the RP2 gene and in the original 19 RPGR exons are detected in less than 10% and approximately 20% of XLRP probands, respectively. Their studies revealed RPGR ORF15 mutations in an additional 30% of 91 well-documented families with X-linked recessive inheritance and in 22% of the total 234 probands analyzed. They suggested that mutations in an uncharacterized RPGR exon(s), intronic changes, or another gene in the region may be responsible for the disease in the remainder of this North American cohort.

Bader et al. (2003) screened 58 German XLRP families and found RP2 mutations in 8% and RPGR mutations in 71%. They also reported a detailed strategy for analyzing the RPGR ORF15 mutation hot spot, which could not be screened by standard procedures.

Sharon et al. (2003) determined the mutation spectrum of the RP2 and RPGR genes in patients with X-linked retinitis pigmentosa. They screened 187 unrelated male patients and found 10 mutations in RP2, 2 of which were novel, and 80 mutations in RPGR, 41 of which were novel. Patients with RP2 mutations had, on average, lower visual acuity but similar visual field area, final dark-adapted threshold, and 30-Hz ERG amplitude compared with those with RPGR mutations. Among the 66% of patients with RPGR mutations in ORF, regression analysis showed that the final dark-adapted threshold became lower (i.e., closer to normal) and that the 30-Hz ERG amplitude increased as the length of the wildtype ORF15 amino acid sequence increased. Furthermore, as the length of the abnormal amino acid sequence following ORF15 frameshift mutations increased, the severity of disease increased. In summary, these cross-sectional analyses suggested that, at a given age, patients with RP2 mutations retained less visual acuity than do patients with RPGR mutations and that, among patients with RPGR mutations, those with ORF15 mutations have milder disease than do patients with mutations in exons 1 to 14.

Demirci et al. (2006) reported a 16-year-old boy with RP (300029) and bilateral Coats-like vasculopathy (see 300216) in whom they identified a novel nonsense mutation in ORF15 of the RPGR gene (312610.0024).

Pelletier et al. (2007) reported the screening of the RP2 and RPGR genes in a cohort of 127 French families comprising 93 familial cases of retinitis pigmentosa suggesting X-linked inheritance, including 48 of 93 families; 7 male sibships of RP; 25 sporadic male cases of RP; and 2 cone dystrophies (COD). They identified a total of 14 RP2 mutations, 12 of which were novel, in 14 of 88 familial cases of RP and 1 of 25 sporadic male cases (4%). In 13 of 14 of the familial cases, no expression of the disease was noted in females, while in 1 of 14 families 1 woman developed retinitis pigmentosa in the third decade. A total of 42 RPGR mutations, 26 of which were novel, were identified in 80 families, including 69 of 88 familial cases (78.4%); 2 of 7 male sibship cases (28.6%); 8 of 25 sporadic male cases (32%); and 1 of 2 COD. No expression of the disease was noted in females in 41 of 69 familial cases (59.4%), while at least 1 severely affected woman was recognized in 28 of 69 families (40.6%). The frequency of RP2 and RPGR mutations in familial cases of retinitis pigmentosa suggestive of X-linked transmission was in accordance with that reported elsewhere (RP2: 15.9% vs 6-20%; RPGR: 78.4% vs 55-90%). About 30% of male sporadic cases and 30% of male sibships of RP carried RP2 or RPGR mutations, confirming the pertinence of the genetic screening of XLRP genes in male patients affected with RP commencing in the first decade and leading to profound visual impairment before the age of 30 years.

Sandberg et al. (2007) measured the rates of visual acuity, visual field, and electroretinogram (ERG) loss in 2 large cohorts, one of patients with XLRP due to mutations in the RPGR gene and the other of patients with autosomal dominant RP due to mutations in the RHO gene (see 180380). Patients with RPGR mutations lost Snellen visual acuity at more than twice the mean rate of patients with RHO mutations. The median age of legal blindness was 32 years younger in patients with RPGR mutation than in patients with RHO mutations. Legal blindness was due primarily to loss of visual acuity in RPGR patients and to loss of visual field in RHO patients. Loss of acuity in RPGR patients appeared to be associated with foveal thinning.

In affected individuals from an Israeli family with 'semi-dominant' X-linked retinitis pigmentosa, in which obligatory female carriers manifested high myopia, low visual acuity, constricted visual fields, and severely reduced electroretinogram amplitudes, Banin et al. (2007) identified the G275S mutation in the RPGR gene (312610.0003). The authors stated that obligate carriers from 2 unrelated Danish families in which Roepman et al. (1996) previously identified this mutation had no visual complaints and normal to slightly reduced retinal function. The disease-related RPGR haplotype of the Israeli family was found to be different from that of 2 Danish families, indicating that the G275S mutation arose twice independently on different X-chromosome backgrounds. Genetic analysis excluded skewed X-inactivation patterns, chromosomal abnormalities, distorted RPGR expression levels, and mutations in 3 candidate genes as the cause for the differences in disease severity of female carriers. Banin et al. (2007) suggested that an additional gene or genes linked to RPGR modulate disease expression in severely affected carriers.

Nishiguchi et al. (2013) identified a Japanese male RP patient who was heterozygous for a frameshift mutation in the ciliary gene NEK2 (604043.0001), but who also carried a frameshift mutation in RPGR (312610.0026). Studies in zebrafish suggested that the RPGR allele interacts in trans with the NEK2 locus to exacerbate photoreceptor defects.

Among female carriers from 45 families with RP3, Comander et al. (2015) found that those with RPGR ORF15 mutations tended to have worse visual function than those with RPGR exon 1 through 14 mutations.

Cone-Rod Degeneration

Mears et al. (2000) restudied a family with what was labeled 'X-linked dominant cone-rod degeneration' (300029) and thought to map to Xp22.13-p22.11, and remapped the disorder to Xp22.1-p11.4. This new interval overlapped the RP3 (RPGR) and the CORDX1 (304020) genes. They identified a de novo insertion (312610.0013) in exon ORF15 of the RPGR gene. The identification of an RPGR mutation in a family with a severe form of cone-rod degeneration suggested that RPGR mutations may encompass a broader phenotypic spectrum than had previously been recognized in 'typical' retinitis pigmentosa.

Retinitis Pigmentosa and Sinopulmonary Infections with or without Deafness

In the family with X-linked retinitis pigmentosa with recurrent respiratory infections (300455) described by van Dorp et al. (1992), Dry et al. (1999) identified an IVS5+1G-T splice site mutation in the RPGR gene (312610.0016).

In a family in which affected males in an X-linked recessive pedigree pattern had retinitis pigmentosa associated with impaired hearing and sinorespiratory infections, Zito et al. (2003) identified A 2-bp deletion, 845delTG, in exon 8 of the RPGR gene (312610.0019).

In a family in which a mother had retinitis pigmentosa and her 2 sons had retinitis pigmentosa, multiple respiratory infections, and primary ciliary dyskinesia, Moore et al. (2006) identified a 57-bp deletion in the RPGR gene (312610.0023).

Atrophic Macular Degeneration

Ayyagari et al. (2002) described a family in which 10 males had primarily macular atrophy causing progressive loss of visual acuity with minimal peripheral visual impairment (300834). One additional male showed extensive macular degeneration plus peripheral loss of retinal pigment epithelium and choriocapillaries. Full-field electroretinograms showed normal cone and rod responses in some affected males despite advanced macular degeneration. In affected members of this family, Ayyagari et al. (2002) identified a G-to-T transversion at nucleotide 1164 of intron 15 (312610.0017) that cosegregated with the disease and may create a donor splice site. Thus the phenotypic range associated with this gene was expanded.


Animal Model

Hong et al. (2000) created an RPGR-deficient murine model of RP3 by gene knockout. In the mutant mice, cone photoreceptors exhibited ectopic localization of cone opsins in the cell body and synapses, and rod photoreceptors had a reduced level of rhodopsin. Subsequently, both rod and cone photoreceptors degenerated. RPGR was found normally localized to the connecting cilia of rod and cone photoreceptors. The data pointed to a role for RPGR in maintaining the polarized protein distribution across the connecting cilium by facilitating directional transport or restricting redistribution. The function of RPGR is essential for the long-term maintenance of photoreceptor viability.

X-linked progressive retinal atrophy (XLPRA) in the Siberian Husky dog closely resembles XLRP in humans. Zeiss et al. (2000) established a linkage map of the canine X chromosome, and determined that XLPRA was tightly linked to an intragenic RPGR polymorphism (lod = 11.7, zero recombination), thus confirming locus homology with RP3. They cloned the full-length canine RPGR cDNA and 3 additional splice variants. No disease-causing mutation was found in the RPGR-coding sequence of the 4 splice variants characterized, a finding similar to approximately 80% of human XLRP patients whose disease maps to the RP3 locus.

Zhang et al. (2002) found different mutations in exon ORF15 of the RPGR gene in 2 distinct mutant dog strains: XLPRA1 and XLPRA2. Microdeletions resulting in a premature stop or a frameshift mutation produced very different retinal phenotypes, which were allele-specific and consistent for each mutation. The phenotype associated with a frameshift mutation in XLPRA2 was very severe and manifested during retinal development; the phenotype resulting from a nonsense mutation in XLPRA1 was expressed only after normal photoreceptor morphogenesis. The frameshift mutation dramatically alters the deduced amino acid sequence, and the protein aggregates in the endoplasmic reticulum of transfected COS-7 cells.

Beltran et al. (2006) described the course of retinal disease in canine XLPRA2 caused by a 2-nucleotide microdeletion in RPGR ORF15. The disorder is characterized by abnormal photoreceptor maturation followed by progressive rod-cone degeneration and early inner retina remodeling. Abnormal development of photoreceptors was recognizable as early as 3.9 weeks of age. Outer segment (OS) misalignment was followed by their disorganization and fragmentation. Reduction in length and broadening of rod and cone inner segments (IS) was observed next, followed by the focal loss of rod and cone IS later. The proportion of dying photoreceptors peaked at approximately 6 to 7 weeks of age and was significantly reduced after 12 weeks. In addition to rod and cone opsin mislocalization, there was early rod neurite sprouting, retraction of rod bipolar cell dendrites, and increased Mueller cell reactivity. Later in the course of the disease, changes were also noted in horizontal cells and amacrine cells.

Beltran et al. (2007) noted that ciliary neurotrophic factor (CNTF; 118945) had been found to rescue photoreceptors in several animal models. They evaluated treatment with CNTF in XLPRA2 dogs. All CNTF-treated eyes showed early clinical signs of corneal epitheliopathy, subcapsular cataracts, and uveitis. No statistically significant difference in outer nuclear layer thickness was seen between CNTF-treated and control eyes. Prominent retinal remodeling that consisted of an abnormal increase in the number of rods, and in misplacement of some rods, cones, and bipolar and Muller cells, was observed in the peripheral retina of CNTF-treated eyes. In XLPRA2 dogs, intravitreal injection of CNTF failed to prevent photoreceptors from undergoing cell death in the central and midperipheral retina. CNTF also caused ocular side effects and morphologic alterations in the periphery that were consistent with cell dedifferentiation and proliferation. Beltran et al. (2007) concluded that some inherited forms of retinal degeneration may not respond to the neuroprotective effects of CNTF.

Ghosh et al. (2010) showed that rpgr was expressed predominantly in the retina, brain, and gut of zebrafish. In zebrafish retina, rpgr primarily localized to the sensory cilium of photoreceptors. Antisense morpholino-mediated knockdown of rpgr function in zebrafish resulted in reduced length of Kupffer vesicle cilia and was associated with ciliary anomalies including shortened body axis, kinked tail, hydrocephaly, and edema, but did not affect retinal development. These phenotypes could be rescued by wildtype human RPGR. Several RPGR mutants (see, e.g., 312610.0006 and 312610.0020) could also reverse the morpholino-induced phenotype, suggesting their potential hypomorphic function. Selected RPGR mutations observed in XLRP (see, e.g. 312610.0009; E589X) or syndromic retinitis pigmentosa (312610.0016; 312610.0019; 312610.0023) did not completely rescue the rpgr-morpholino phenotype, indicating a more deleterious effect of the mutation on the function of RPGR. Ghosh et al. (2010) proposed that RPGR may be involved in cilia-dependent cascades during development in zebrafish.

Shu et al. (2010) identified 2 genes resembling human RPGR in zebrafish (Zfrpgr1 and Zfrpgr2), both of which are expressed within the nascent and adult eye as well as more widely during development. Zfrpgr2 appears to be functionally orthologous to human RPGR, because it encodes similar protein isoforms (Zfrpgr2(Orf15) and Zfrpgr2(ex1-17)) and, similar to other ciliary proteins, translation suppression causes developmental defects affecting gastrulation and tail and head development. These defects are consistent with a ciliary function and were rescued by human RPGR but not by RPGR mutants causing retinal dystrophy. Unlike in mammals, RPGR knockdown in zebrafish resulted in both abnormal development and increased cell death in the dysplastic retina. Developmental abnormalities in the eye included lamination defects, failure to develop photoreceptor outer segments, and a small eye phenotype, associated with increased cell death throughout the retina. These defects could be rescued by expression of wildtype but not mutant forms of human RPGR. Zfrpgr2 knockdown also resulted in an intracellular transport defect affecting retrograde but not anterograde transport of organelles. Shu et al. (2010) concluded that Zfrpgr2 is necessary both for the normal differentiation and lamination of the retina and to prevent apoptotic retinal cell death, which may relate to its proposed role in dynein-based retrograde transport processes.


ALLELIC VARIANTS 26 Selected Examples):

.0001   RETINITIS PIGMENTOSA 3

RPGR, PHE130CYS
SNP: rs62638644, ClinVar: RCV000010575, RCV000085105

In a patient with retinitis pigmentosa-3 (300029), Roepman et al. (1996) identified a T-to-G transversion at nucleotide 420 of their RP3 cDNA sequence, resulting in a phe130-to-cys substitution (F130C). The authors did not denote the amino acid residue position in their publication.

Murga-Zamalloa et al. (2010) showed that the F130C mutation in RPGR did not alter interaction of RPGR with the small GTPase RAB8A (165040) in vitro, but it reduced the ability of RPGR to induce GDP/GTP exchange on RAB8A.


.0002   RETINITIS PIGMENTOSA 3

RPGR, PRO235SER
SNP: rs62638651, ClinVar: RCV000010576, RCV000085116

In a patient with retinitis pigmentosa-3 (300029), Roepman et al. (1996) identified a C-to-T transition at nucleotide 734 of their RP3 cDNA sequence, resulting in a pro235-to-ser substitution (P235S). The authors did not denote the amino acid residue position in their publication.


.0003   RETINITIS PIGMENTOSA 3

RPGR, GLY275SER
SNP: rs62642057, ClinVar: RCV000010577, RCV000085125, RCV001003203, RCV002512961

In 2 patients with retinitis pigmentosa-3 (300029), Roepman et al. (1996) identified a G-to-A transition at nucleotide 854 of their RP3 cDNA sequence, resulting in a gly275-to-ser substitution (G275S). The authors did not denote the amino acid residue position in their publication.

In affected individuals from an Israeli family with 'semi-dominant' X-linked retinitis pigmentosa, in which obligatory female carriers manifested high myopia, low visual acuity, constricted visual fields, and severely reduced electroretinogram amplitudes, Banin et al. (2007) identified the G275S mutation in the RPGR gene. The disease-related RPGR haplotype of the Israeli family was found to be different from that of the 2 families previously studied by Roepman et al. (1996) in which obligate carriers of G275S had no visual complaints, indicating that the G275S mutation arose twice independently on different X-chromosome backgrounds. Genetic analysis excluded skewed X-inactivation patterns, chromosomal abnormalities, distorted RPGR expression levels, and mutations in 3 candidate genes as the cause for the differences in disease severity of female carriers. Banin et al. (2007) suggested that an additional gene or genes linked to RPGR modulate disease expression in severely affected carriers.


.0004   RETINITIS PIGMENTOSA 3

RPGR, 4-BP DEL, NT1433
SNP: rs62653030, ClinVar: RCV000085055, RCV002225285

Roepman et al. (1996) found a 4-bp deletion of nucleotides 1433-1436 of their RP3 cDNA sequence in a patient with retinitis pigmentosa-3 (300029) that resulted in a truncated RP3 protein with 6 abnormal C-terminal amino acids.


.0005   RETINITIS PIGMENTOSA 3

RPGR, IVS10DS, A-G, +3
SNP: rs62635002, ClinVar: RCV000010579, RCV000085047, RCV001073791, RCV002514520

In a patient with retinitis pigmentosa-3 (300029), Fujita et al. (1997) found an A-to-G transition in the RPGR gene at the third basepair downstream of exon 10 nucleotide 1304 (sequence designation is according to Meindl et al., 1996) in the splice-donor region of intron 10. The mutation resulted in incorrect splicing of the exon 10-11 junction.


.0006   RETINITIS PIGMENTOSA 3

RPGR, GLY60VAL
SNP: rs62638634, ClinVar: RCV000010580, RCV000085072, RCV000822760, RCV001074755

One of the 15 novel mutations identified by Buraczynska et al. (1997) was a G-to-T transversion at nucleotide 238 in exon 3, predicted to lead to a gly60-to-val (G60V) amino acid substitution. Fishman et al. (1998) reported in detail on 2 families with retinitis pigmentosa-3 (300029) with the G60V mutation, one of which was the family earlier reported by Buraczynska et al. (1997). The mutation was associated with a severe clinical phenotype in male patients and a patchy retinopathy without a tapetal-like reflex in carrier females. Psychophysical and electrophysiologic testing on carriers indicated that cone and rod functions were impaired equivalently. When present in carriers, visual field restriction was most apparent in, or limited to, the superotemporal quadrant, which corresponded to the retinal pigmentary changes that tended to occur in the inferonasal retina.


.0007   RETINITIS PIGMENTOSA 3

RPGR, 2-BP DEL, NT1571
SNP: rs281865304, ClinVar: RCV000085061, RCV002225286, RCV002514522

In a black family with retinitis pigmentosa-3 (300029), Fishman et al. (1998) identified a 2-basepair deletion in the RP3 gene. The deletion occurred in exon 13 and created a frameshift and premature stop codon, resulting in a protein truncation. The deletion involved nucleotides 1571 and 1572, a CA dinucleotide. The clinical findings were those characteristic of X-linked retinitis pigmentosa. In 2 obligate carriers, a tapetal-like reflex was not clinically apparent.


.0008   RETINITIS PIGMENTOSA 3

RPGR, EX15ADEL
ClinVar: RCV000010583

In a family with retinitis pigmentosa-3 (300029), Kirschner et al. (1999) identified deletion of exon 15A of the RPGR gene. The splice variant containing exon 15A is expressed only in the retina. The phenotype was of retinal dystrophy with relatively late onset of night blindness, around the age of 10 years. From 30 years of age, very rapid deterioration took place and led to blindness by the age of 37 years. Electrophysiologically, the retinopathy appeared as a cone-rod dystrophy.


.0009   RETINITIS PIGMENTOSA 3

RPGR, THR99ASN
SNP: rs62638637, ClinVar: RCV000010584, RCV000085096

One of the unique mutations found by Miano et al. (1999) in south European patients with retinitis pigmentosa-3 (300029) was a C-to-A transversion of nucleotide 355 of the RP3 gene resulting in a thr99-to-asn (T99N) amino acid substitution. The patient was Spanish.


.0010   RETINITIS PIGMENTOSA 3

RPGR, 2-BP DEL, 673AG
SNP: rs730882261, ClinVar: RCV000162095, RCV001075867, RCV001091403, RCV001251604, RCV002516436

Vervoort et al. (2000) identified a novel exon, ORF15, of the RPGR gene. One mutation detected in this region, found in 4 families with retinitis pigmentosa-3 (300029), was a 2-nucleotide deletion of AG at nucleotide 673 resulting in frameshift. The high frequency of mutations found in this exon suggested to Vervoort et al. (2000) that ORF15 is a mutation hotspot, with its high mutability being due to nucleotide composition (purine-rich) or repetitive nature of the sequence.


.0011   RETINITIS PIGMENTOSA 3

RPGR, 2-BP DEL, 652AG
SNP: rs398122960, ClinVar: RCV000076907, RCV000622673, RCV001003195, RCV001008073, RCV001074955, RCV001807778, RCV001854345, RCV002286702, RCV002498372

In 2 families with retinitis pigmentosa-3 (300029), Vervoort et al. (2000) identified a mutation in ORF15, a 2-bp deletion of AG at nucleotide 652.


.0012   RETINITIS PIGMENTOSA 3

RPGR, GLU299TER
SNP: rs137852549, ClinVar: RCV000010586, RCV002464059, RCV003534308

In a family with retinitis pigmentosa-3 (300029), Vervoort et al. (2000) identified a G-to-T transversion in ORF15, an alternatively spliced 3-prime terminal exon of the RPGR gene. This mutation resulted in a glutamic acid-to-stop substitution at residue 299 of ORF15 (E299X).


.0013   RETINITIS PIGMENTOSA 3

RPGR, 1-BP INS, 173A
SNP: rs2067199347, ClinVar: RCV001251559, RCV002464426, RCV003230653

In a family with X-linked cone-rod degeneration, previously reported by McGuire et al. (1995) as RP15 (300029), Mears et al. (2000) identified a 1-bp insertion, an adenine (173_174insA), causing a frameshift with insertion of 9 novel amino acids and truncation of the protein product 501 amino acids premature of the ORF15 stop codon. In this family, affected males and 'carrier' females presented with early cone involvement, which differs from the typical rod-predominant manifestation of X-linked RP.


.0014   CONE-ROD DYSTROPHY, X-LINKED, 1

CONE DYSTROPHY, X-LINKED, 1, INCLUDED
RPGR, 2-BP DEL, 1343GG
SNP: rs606231180, ClinVar: RCV000010588, RCV001003190, RCV001074702, RCV001251568, RCV002512962, RCV003151713, RCV003225923, RCV003389748

In 2 of 3 families with X-linked cone-rod dystrophy (CORDX1; 304020), Demirci et al. (2002) demonstrated a 2-bp deletion, delGG, in ORF15 of the RPGR gene, resulting in a frameshift leading to altered amino acid structure and early termination.

Yang et al. (2002) mapped 2 Caucasian families with X-linked cone dystrophy (COD1; see 304020) to the CORDX1 locus on Xp and identified 2 distinct mutations in ORF15 of the RPGR gene. One was ORF15+1343-1344delGG and the other ORF15+694-708del15 (312610.0018).


.0015   CONE-ROD DYSTROPHY, X-LINKED, 1

RPGR, 2-BP DEL, 1339AG
SNP: rs606231181, ClinVar: RCV000010589, RCV001074839, RCV001251567, RCV002464061, RCV002512963, RCV003324498

In a family with X-linked cone-rod dystrophy (CORDX1; 304020), Demirci et al. (2002) found a 2-bp deletion, delAG, in exon 15 (ORF15) of the RPGR gene, resulting in a frameshift leading to altered amino acid structure and early termination.


.0016   RETINITIS PIGMENTOSA AND SINORESPIRATORY INFECTIONS WITH OR WITHOUT DEAFNESS

RPGR, IVS5, G-T, +1
SNP: rs62638646, ClinVar: RCV000010590, RCV001698940

Dry et al. (1999) demonstrated a splice site mutation in the RPGR gene in a family with X-linked retinitis pigmentosa with recurrent respiratory infections (300455) in which van Dorp et al. (1992) had, by electron microscopy, shown nasal ciliary abnormalities in some affected males, consisting of deficient inner dynein arms, incomplete microtubules, and disorientation of cilia, associated with recurrent respiratory infections indistinguishable from immotile cilia syndrome. Van Dorp et al. (1992) did not note whether hearing was impaired in affected individuals.


.0017   MACULAR DEGENERATION, ATROPHIC, X-LINKED

RPGR, IVS15, G-T, +1164
SNP: rs2067140471, ClinVar: RCV000010591, RCV001073899, RCV001251534, RCV002554689

In a family with X-linked recessive atrophic macular degeneration (300834), Ayyagari et al. (2002) found that affected males had an ORF15+1164G-T mutation thought to create a novel donor splice site in the RPGR gene.


.0018   CONE DYSTROPHY, X-LINKED, 1

RPGR, 15-BP DEL, NT694
SNP: rs777850798, gnomAD: rs777850798, ClinVar: RCV000475821, RCV002225624, RCV002464206

Yang et al. (2002) mapped 2 Caucasian families with X-linked cone dystrophy (see 304020) to the CORDX1 locus on Xp and identified 2 distinct mutations in ORF15 of the RPGR gene. One was ORF15+1343-1344delGG (312610.0014) and the other ORF15+694-708del15. The latter mutation was predicted to delete 5 amino acids from the C-terminal portion of the protein product.


.0019   RETINITIS PIGMENTOSA, SINORESPIRATORY INFECTIONS, AND DEAFNESS

RPGR, 2-BP DEL, 845TG
SNP: rs2147248070, ClinVar: RCV003151714

In a family in which affected males in an X-linked recessive pedigree pattern had retinitis pigmentosa associated with impaired hearing and sinorespiratory infections (300455), Zito et al. (2003) identified a 2-bp deletion, 845delTG, in exon 8 of the RPGR gene. This frameshift mutation at residue 262 was predicted to introduce 19 new amino acids and a premature stop codon, resulting in a truncated protein of 280 residues. Carrier females and affected males in this kindred were myopic but female carriers were asymptomatic, had normal fields to confrontation, and showed sparse peripheral intraretinal pigmentation. However, additional systemic symptoms were observed in both hemizygous males and heterozygous females. One of the most striking and obvious additional features was the requirement of hearing aids by both affected males and female carriers. Both had severe recurrent ear infections from early childhood continuing into adulthood. Because of hearing loss in the high frequencies, the audiogram was considered consistent with sensorineural hearing loss, although a conductive hearing component may have contributed. Affected males and carrier females suffered from severe recurrent sinus infections. Three affected males had chronic recurrent chest infections starting in early childhood, with episodes of bronchitis, which continued into adulthood. The possibility of renal failure being part of the clinical picture was suggested by its occurrence in 1 affected male. The phenotype overlapped those described for primary ciliary dyskinesia (244400) and Usher syndrome (276900) and provided support for an essential ciliary function for RPGR in the retina and other tissues.


.0020   RETINITIS PIGMENTOSA, SINORESPIRATORY INFECTIONS, AND DEAFNESS

RPGR, GLY173ARG
SNP: rs137852550, ClinVar: RCV003128227, RCV003151715

In 2 brothers with X-linked retinitis pigmentosa with impaired hearing and recurrent respiratory infections (300455), Iannaccone et al. (2003) identified a 576G-C transversion in a conserved region of the RPGR gene, resulting in a gly173-to-arg (G173R) substitution. The mutation was also identified in the asymptomatic mother and the symptomatic grandmother. The G173R change was not identified in more than 200 control chromosomes.


.0021   CONE-ROD DYSTROPHY, X-LINKED, 1

RPGR, GLU364ASP, GLU365TER
ClinVar: RCV000010595

In a family with X-linked cone-rod dystrophy (304020), Ebenezer et al. (2005) found a 2 consecutive nucleotide substitutions in the ORF15 exon of the RPGR gene, 1094A-C and 1095G-T, that resulted in glu364-to-asp and glu365-to-ter amino acid substitutions, respectively (E364D/E365X).


.0022   CONE-ROD DYSTROPHY, X-LINKED, 1

RPGR, GLY392TER
SNP: rs137852551, ClinVar: RCV000010596

In a family with X-linked cone-rod dystrophy (304020), Ebenezer et al. (2005) identified a G-to-T transversion at nucleotide 1176 of the ORF15 exon of the RPGR gene that resulted in a gly392-to-ter (G392X) substitution.


.0023   RETINITIS PIGMENTOSA AND SINORESPIRATORY INFECTIONS WITHOUT DEAFNESS

RPGR, 57-BP DEL, NT631
ClinVar: RCV003151716

In a family in which a mother with retinitis pigmentosa had 2 boys with multiple ear, sinus, and respiratory infections who were diagnosed with primary ciliary dyskinesia and retinitis pigmentosa (300455), Moore et al. (2006) identified a 57-bp deletion involving the last 48 bp of exon 6 plus the following 9 bp of the adjacent intron (631-IVS6+9del) in the RPGR gene. The boys had axonemal abnormalities on electron microscopy leading to the diagnosis of primary ciliary dyskinesia; ophthalmologic examination confirmed the diagnosis of retinitis pigmentosa. Moore et al. (2006) stated that this was the first clear demonstration of X-linked transmission of primary ciliary dyskinesia.


.0024   RETINITIS PIGMENTOSA 3

RPGR, 912G-T
SNP: rs2067163605, ClinVar: RCV001073958, RCV001251620, RCV002554691

Demirci et al. (2006) reported a 16-year-old boy with RP (300029) and bilateral Coats-like vasculopathy (see 300216) in whom they identified a 912G-T transversion in ORF15 of the RPGR gene, predicted to result in a truncated protein missing 264 amino acids at the carboxyl end. The mutation segregated with RP in the family; clinical findings in other family members, including 2 affected male patients and 3 obligate carrier females, were consistent with typical X-linked recessive RP. Because the proband was the only family member who had Coats-like exudative vasculopathy, Demirci et al. (2006) suggested that other genetic and/or environmental factors might be involved.


.0025   RETINITIS PIGMENTOSA 3

RPGR, IVS9AS, G-A, -55
SNP: rs1410177435, gnomAD: rs1410177435, ClinVar: RCV000010599, RCV002512964

In a Swiss man with mild retinitis pigmentosa-3 (300029), Neidhardt et al. (2007) identified a hemizygous G-to-A transition located 363 bp downstream of exon 9 and 55 bp upstream from the exon splice acceptor site of exon 9A (g.26652G-A), resulting in increased levels of RPGR mRNA transcripts containing exon 9A. The man had night blindness since childhood and presented in his late thirties with peripheral visual impairment. The level of exon 9A expression in the patient's unaffected mother, who was heterozygous for the mutation, was normalized to 100%; the patient was found to have 180% expression levels.


.0026   RETINITIS PIGMENTOSA 3

RPGR, 2-BP DEL, 2405AG
ClinVar: RCV000076907, RCV000622673, RCV001003195, RCV001008073, RCV001074955, RCV001807778, RCV001854345, RCV002286702, RCV002498372

In a Japanese male patient with retinitis pigmentosa (RP3; 300029), Nishiguchi et al. (2013) identified a 2-bp deletion (c.2405_2406delAG) in the RPGR gene, causing a frameshift (Glu802fs); the authors stated that this mutation had previously been described as a sufficient cause of X-linked RP by Vervoort et al. (2000). The Japanese patient also carried a heterozygous frameshift mutation in another RP-associated ciliary gene, NEK2 (604043.0001); studies in zebrafish suggested that the RPGR allele interacts in trans with the NEK2 locus to exacerbate photoreceptor defects.


REFERENCES

  1. Ayyagari, R., Demirci, F. Y., Liu, J., Bingham, E. L., Stringham, H., Kakuk, L. E., Boehnke, M., Gorin, M. B., Richards, J. E., Sieving, P. A. X-linked recessive atrophic macular degeneration from RPGR mutation. Genomics 80: 166-171, 2002. [PubMed: 12160730] [Full Text: https://doi.org/10.1006/geno.2002.6815]

  2. Bader, I., Brandau, O., Achatz, H., Apfelstedt-Sylla, E., Hergersberg, M., Lorenz, B., Wissinger, B., Wittwer, B., Rudolph, G., Meindl, A., Meitinger, T. X-linked retinitis pigmentosa: RPGR mutations in most families with definite X linkage and clustering of mutations in a short sequence stretch of exon ORF15. Invest. Ophthal. Vis. Sci. 44: 1458-1463, 2003. [PubMed: 12657579] [Full Text: https://doi.org/10.1167/iovs.02-0605]

  3. Banin, E., Mizrahi-Meissonnier, L., Neis, R., Silverstein, S., Magyar, I., Abeliovich, D., Roepman, R., Berger, W., Rosenberg, T., Sharon, D. A non-ancestral RPGR missense mutation in families with either recessive or semi-dominant X-linked retinitis pigmentosa. Am. J. Med. Genet. 143A: 1150-1158, 2007. [PubMed: 17480003] [Full Text: https://doi.org/10.1002/ajmg.a.31642]

  4. Beltran, W. A., Hammond, P., Acland, G. M., Aguirre, G. D. A frameshift mutation in RPGR exon ORF15 causes photoreceptor degeneration and inner retina remodeling in a model of X-linked retinitis pigmentosa. Invest. Ophthal. Vis. Sci. 47: 1669-1681, 2006. [PubMed: 16565408] [Full Text: https://doi.org/10.1167/iovs.05-0845]

  5. Beltran, W. A., Wen, R., Acland, G. M., Aguirre, G. D. Intravitreal injection of ciliary neurotrophic factor (CNTF) causes peripheral remodeling and does not prevent photoreceptor loss in canine RPGR mutant retina. Exp. Eye Res. 84: 753-771, 2007. [PubMed: 17320077] [Full Text: https://doi.org/10.1016/j.exer.2006.12.019]

  6. Boylan, J. P., Wright, A. F. Identification of a novel protein interacting with RPGR. Hum. Molec. Genet. 9: 2085-2093, 2000. [PubMed: 10958647] [Full Text: https://doi.org/10.1093/hmg/9.14.2085]

  7. Branham, K., Othman, M., Brumm, M., Karoukis, A. J., Atmaca-Sonmez, P., Yashar, B. M., Schwartz, S. B., Stover, N. B., Trzupek, K., Wheaton, D., Jennings, B., Ciccarelli, M. L., and 13 others. Mutations in RPGR and RP2 account for 15% of males with simplex retinal degenerative disease. Invest. Ophthal. Vis. Sci. 53: 8232-8237, 2012. [PubMed: 23150612] [Full Text: https://doi.org/10.1167/iovs.12-11025]

  8. Breuer, D. K., Yashar, B. M., Filippova, E., Hiriyanna, S., Lyons, R. H., Mears, A. J., Asaye, B., Acar, C., Vervoort, R., Wright, A. F., Musarella, M. A., Wheeler, P., and 9 others. A comprehensive mutation analysis of RP2 and RPG2 in a North American cohort of families with X-linked retinitis pigmentosa. Am. J. Hum. Genet. 70: 1545-1554, 2002. Note: Erratum: Am. J. Hum. Genet. 71: 1258 only, 2002. [PubMed: 11992260] [Full Text: https://doi.org/10.1086/340848]

  9. Buraczynska, M., Wu, W., Fujita, R., Buraczynska, K., Phelps, E., Andreasson, S., Bennett, J., Birch, D. G., Fishman, G. A., Hoffman, D. R., Inana, G., Jacobson, S. G., Musarella, M. A., Sieving, P. A., Swaroop, A. Spectrum of mutations in the RPGR gene that are identified in 20% of families with X-linked retinitis pigmentosa. Am. J. Hum. Genet. 61: 1287-1292, 1997. [PubMed: 9399904] [Full Text: https://doi.org/10.1086/301646]

  10. Comander, J., Weigel-DiFranco, C., Sandberg, M. A., Berson, E. L. Visual function in carriers of X-linked retinitis pigmentosa. Ophthalmology 122: 1899-1906, 2015. [PubMed: 26143542] [Full Text: https://doi.org/10.1016/j.ophtha.2015.05.039]

  11. Demirci, F. Y. K., Rigatti, B. W., Mah, T. S., Gorin, M. B. A novel RPGR exon ORF15 mutation in a family with X-linked retinitis pigmentosa and Coats'-like exudative vasculopathy. Am. J. Ophthal. 141: 208-210, 2006. [PubMed: 16387007] [Full Text: https://doi.org/10.1016/j.ajo.2005.07.077]

  12. Demirci, F. Y. K., Rigatti, B. W., Wen, G., Radak, A. L., Mah, T. S., Baic, C. L., Traboulsi, E. I., Alitalo, T., Ramser, J., Gorin, M. B. X-linked cone-rod dystrophy (locus COD1): identification of mutations in RPGR exon ORF15. Am. J. Hum. Genet. 70: 1049-1053, 2002. [PubMed: 11857109] [Full Text: https://doi.org/10.1086/339620]

  13. Dry, K. L., Manson, F. D. C., Lennon, A., Bergen, A. A. B., Van Dorp, D. B., Wright, A. F. Identification of a 5-prime splice site mutation in the RPGR gene in a family with X-linked retinitis pigmentosa (RP3). Hum. Mutat. 13: 141-145, 1999. [PubMed: 10094550] [Full Text: https://doi.org/10.1002/(SICI)1098-1004(1999)13:2<141::AID-HUMU6>3.0.CO;2-Q]

  14. Ebenezer, N. D., Michaelides, M., Jenkins, S. A., Audo, I., Webster, A. R., Cheetham, M. E., Stockman, A., Maher, E. R., Ainsworth, J. R., Yates, J. R., Bradshaw, K., Holder, G. E., Moore, A. T., Hardcastle, A. J. Identification of novel RPGR ORF15 mutations in X-linked progressive cone-rod dystrophy. Invest. Ophthal. Vis. Sci. 46: 1891-1898, 2005. [PubMed: 15914600] [Full Text: https://doi.org/10.1167/iovs.04-1482]

  15. Fishman, G. A., Grover, S., Buraczynska, M., Wu, W., Swaroop, A. A new 2-base pair deletion in the RPGR gene in a black family with X-linked retinitis pigmentosa. Arch. Ophthal. 116: 213-218, 1998. [PubMed: 9488274] [Full Text: https://doi.org/10.1001/archopht.116.2.213]

  16. Fishman, G. A., Grover, S., Jacobson, S. G., Alexander, K. R., Derlacki, D. J., Wu, W., Buraczynska, M., Swaroop, A. X-linked retinitis pigmentosa in two families with a missense mutation in the RPGR gene and putative change of glycine to valine at codon 60. Ophthalmology 105: 2286-2296, 1998. [PubMed: 9855162] [Full Text: https://doi.org/10.1016/S0161-6420(98)91231-3]

  17. Fujita, R., Buraczynska, M., Gieser, L., Wu, W., Forsythe, P., Abrahamson, M., Jacobson, S. G., Sieving, P. A., Andreasson, S., Swaroop, A. Analysis of the RPGR gene in 11 pedigrees with the retinitis pigmentosa type 3 genotype: paucity of mutations in the coding region but splice defects in two families. Am. J. Hum. Genet. 61: 571-580, 1997. [PubMed: 9326322] [Full Text: https://doi.org/10.1086/515523]

  18. Ghosh, A. K., Murga-Zamalloa, C. A., Chan, L., Hitchcock, P. F., Swaroop, A., Khanna, H. Human retinopathy-associated ciliary protein retinitis pigmentosa GTPase regulator mediates cilia-dependent vertebrate development. Hum. Molec. Genet. 19: 90-98, 2010. [PubMed: 19815619] [Full Text: https://doi.org/10.1093/hmg/ddp469]

  19. Hong, D.-H., Pawlyk, B. S., Adamian, M., Li, T. Dominant, gain-of-function mutant produced by truncation of RPGR. Invest. Ophthal. Vis. Sci. 45: 36-41, 2004. [PubMed: 14691151] [Full Text: https://doi.org/10.1167/iovs.03-0787]

  20. Hong, D.-H., Pawlyk, B. S., Adamian, M., Sandberg, M. A., Li, T. A single, abbreviated RPGR-ORF15 variant reconstitutes RPGR function in vivo. Invest. Ophthal. Vis. Sci. 46: 435-441, 2005. [PubMed: 15671266] [Full Text: https://doi.org/10.1167/iovs.04-1065]

  21. Hong, D.-H., Pawlyk, B. S., Shang, J., Sandberg, M. A., Berson, E. L., Li, T. A retinitis pigmentosa GTPase regulator (RPGR)-deficient mouse model for X-linked retinitis pigmentosa (RP3). Proc. Nat. Acad. Sci. 97: 3649-3654, 2000. [PubMed: 10725384] [Full Text: https://doi.org/10.1073/pnas.97.7.3649]

  22. Hong, D-H., Pawlyk, B., Sokolov, M., Strissel, K. J., Yang, Y., Tulloch, B., Wright, A. F., Arshavsky, V. Y., Li, T. RPGR isoforms in photoreceptor connecting cilia and the transitional zone of motile cilia. Invest. Ophthal. Vis. Sci. 44: 2413-2421, 2003. [PubMed: 12766038] [Full Text: https://doi.org/10.1167/iovs.02-1206]

  23. Iannaccone, A., Breuer, D. K., Wang, X. F., Kuo, S. F., Normando, E. M., Filippova, E., Baldi, A., Hiriyanna, S., MacDonald, C. B., Baldi, F., Cosgrove, D., Morton, C. C., Swaroop, A., Jablonski, M. M. Clinical and immunohistochemical evidence for an X linked retinitis pigmentosa syndrome with recurrent infections and hearing loss in association with an RPGR mutation. (Letter) J. Med. Genet. 40: e118, 2003. Note: Electronic Article. [PubMed: 14627685] [Full Text: https://doi.org/10.1136/jmg.40.11.e118]

  24. Khanna, H., Hurd, T. W., Lillo, C., Shu, X., Parapuram, S. K., He, S., Akimoto, M., Wright, A. F., Margolis, B., Williams, D. S., Swaroop, A. RPGR-ORF15, which is mutated in retinitis pigmentosa, associates with SMC1, SMC3, and microtubule transport proteins. J. Biol. Chem. 280: 33580-33587, 2005. [PubMed: 16043481] [Full Text: https://doi.org/10.1074/jbc.M505827200]

  25. Kirschner, R., Erturk, D., Zeitz, C., Sahin, S., Ramser, J., Cremers, F. P. M., Ropers, H.-H., Berger, W. DNA sequence comparison of human and mouse retinitis pigmentosa GTPase regulator (RPGR) identifies tissue-specific exons and putative regulatory elements. Hum. Genet. 109: 271-278, 2001. [PubMed: 11702207] [Full Text: https://doi.org/10.1007/s004390100572]

  26. Kirschner, R., Rosenberg, T., Schultz-Heienbrok, R., Lenzner, S., Feil, S., Roepman, R., Cremers, F. P. M., Ropers, H.-H., 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. Molec. Genet. 8: 1571-1578, 1999. [PubMed: 10401007] [Full Text: https://doi.org/10.1093/hmg/8.8.1571]

  27. Mavlyutov, T. A., Zhao, H., Ferreira, P. A. Species-specific subcellular localization of RPGR and RPGRIP isoforms: implications for the phenotypic variability of congenital retinopathies among species. Hum. Molec. Genet. 11: 1899-1907, 2002. [PubMed: 12140192] [Full Text: https://doi.org/10.1093/hmg/11.16.1899]

  28. McGuire, R. E., Sullivan, L. S., Blanton, S. H., Church, M. W., Heckenlively, J. R., Daiger, S. P. X-linked dominant cone-rod degeneration: linkage mapping of a new locus for retinitis pigmentosa (RP15) to Xp22.13-p22.11. Am. J. Hum. Genet. 57: 87-94, 1995. [PubMed: 7611300]

  29. Mears, A. J., Hiriyanna, S., Vervoort, R., Yashar, B., Gieser, L., Fahrner, S., Daiger, S. P., Heckenlively, J. R., Sieving, P. A., Wright, A. F., Swaroop, A. Remapping of the RP15 locus for X-linked cone-rod degeneration to Xp11.4-p21.1, and identification of a de novo insertion in the RPGR exon ORF15. Am. J. Hum. Genet. 67: 1000-1003, 2000. [PubMed: 10970770] [Full Text: https://doi.org/10.1086/303091]

  30. Meindl, A., Dry, K., Herrmann, K., Manson, F., Ciccodicola, A., Edgar, A., Carvalho, M. R. S., 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). Nature Genet. 13: 35-42, 1996. [PubMed: 8673101] [Full Text: https://doi.org/10.1038/ng0596-35]

  31. Miano, M. G., Testa, F., Strazzullo, M., Trujillo, M., De Bernardo, C., Grammatico, B., Simonelli, F., Mangino, M., Torrente, I., Ruberto, G., Beneyto, M., Antinolo, G., Rinaldi, E., Danesino, C., Ventruto, V., D'Urso, M., Ayuso, C., Baiget, M., Ciccodicola, A. Mutation analysis of the RPGR gene reveals novel mutations in south European patients with X-linked retinitis pigmentosa. Europ. J. Hum. Genet. 7: 687-694, 1999. [PubMed: 10482958] [Full Text: https://doi.org/10.1038/sj.ejhg.5200352]

  32. Moore, A., Escudier, E., Roger, G., Tamalet, A., Pelosse, B., Marlin, S., Clement, A., Geremek, M., Delaisi, B., Bridoux, A.-M., 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. 43: 326-333, 2006. [PubMed: 16055928] [Full Text: https://doi.org/10.1136/jmg.2005.034868]

  33. Murga-Zamalloa, C. A., Atkins, S. J., Peranen, J., Swaroop, A., Khanna, H. Interaction of retinitis pigmentosa GTPase regulator (RPGR) with RAB8A GTPase: implications for cilia dysfunction and photoreceptor degeneration. Hum. Molec. Genet. 19: 3591-3598, 2010. [PubMed: 20631154] [Full Text: https://doi.org/10.1093/hmg/ddq275]

  34. Neidhardt, J., Glaus, E., Barthelmes, D., Zeitz, C., Fleischhauer, J., Berger, W. Identification and characterization of a novel RPGR isoform in human retina. Hum. Mutat. 28: 797-807, 2007. [PubMed: 17405150] [Full Text: https://doi.org/10.1002/humu.20521]

  35. Nishiguchi, K. M., Tearle, R. G., Liu, Y. P., Oh, E. C., Miyake, N., Benaglio, P., Harper, S., Koskiniemi-Kuendig, H., Venturini, G., Sharon, D., Koenekoop, R. K., Nakamura, M., and 10 others. Whole genome sequencing in patients with retinitis pigmentosa reveals pathogenic DNA structural changes and NEK2 as a new disease gene. Proc. Nat. Acad. Sci. 110: 16139-16144, 2013. [PubMed: 24043777] [Full Text: https://doi.org/10.1073/pnas.1308243110]

  36. Pelletier, V., Jambou, M., Delphin, N., Zinovieva, E., Stum, M., Gigarel, N., Dollfus, H., Hamel, C., Toutain, A., Dufier, J.-L., Roche, O., Munnich, A., Bonnefont, J.-P., Kaplan J., Rozet, J.-M. Comprehensive study of mutations in RP2 and RPGR in patients affected with distinct retinal dystrophies: genotype-phenotype correlations and impact on genetic counseling. Hum. Mutat. 28: 81-91, 2007. [PubMed: 16969763] [Full Text: https://doi.org/10.1002/humu.20417]

  37. Roepman, R., Bernoud-Hubac, N., Schick, D. E., Maugeri, A., Berger, W., Ropers, H.-H., Cremers, F. P. M., Ferreira, P. A. The retinitis pigmentosa GTPase regulator (RPGR) interacts with novel transport-like proteins in the outer segments of rod photoreceptors. Hum. Molec. Genet. 9: 2095-2105, 2000. [PubMed: 10958648] [Full Text: https://doi.org/10.1093/hmg/9.14.2095]

  38. Roepman, R., van Duijnhoven, G., Rosenberg, T., Pinckers, A. J. L. G., Bleeker-Wagemakers, L. M., Bergen, A. A. B., Post, J., Beck, A., Reinhardt, R., Ropers, H.-H., Cremers, F. P. M., Berger, W. Positional cloning of the gene for X-linked retinitis pigmentosa 3: homology with the guanine-nucleotide-exchange factor RCC1. Hum. Molec. Genet. 5: 1035-1041, 1996. [PubMed: 8817343] [Full Text: https://doi.org/10.1093/hmg/5.7.1035]

  39. Rozet, J.-M., Perrault, I., Gigarel, N., Souied, E., Ghazi, I., Gerber, S., Dufier, J.-L., Munnich, A., Kaplan, J. Dominant X linked retinitis pigmentosa is frequently accounted for by truncating mutations in exon ORF15 of the RPGR gene. J. Med. Genet. 39: 284-285, 2002. [PubMed: 11950860] [Full Text: https://doi.org/10.1136/jmg.39.4.284]

  40. Sandberg, M. A., Rosner, B., Weigel-DiFranco, C., Dryja, T. P., Berson, E. L. Disease course of patients with X-linked retinitis pigmentosa due to RPGR gene mutations. Invest. Ophthal. Vis. Sci. 48: 1298-1304, 2007. [PubMed: 17325176] [Full Text: https://doi.org/10.1167/iovs.06-0971]

  41. Sharon, D., Sandberg, M. A., Rabe, V. W., Stillberger, M., Dryja, T. P., Berson, E. L. RP2 and RPGR mutations and clinical correlations in patients with X-linked retinitis pigmentosa. Am. J. Hum. Genet. 73: 1131-1146, 2003. [PubMed: 14564670] [Full Text: https://doi.org/10.1086/379379]

  42. Shu, X., Black, G. C., Rice, J. M., Hart-Holden, N., Jones, A., O'Grady, A., Ramsden, S., Wright, A. F. RPGR mutation analysis and disease: an update. Hum. Mutat. 28: 322-328, 2007. [PubMed: 17195164] [Full Text: https://doi.org/10.1002/humu.20461]

  43. Shu, X., Fry, A. M., Tulloch, B., Manson, F. D. C., Crabb, J. W., Khanna, H., Faragher, A. J., Lennon, A., He, S., Trojan, P., Giessl, A., Wolfrum, U., Vervoort, R., Swaroop, A., Wright, A. F. RPGR ORF15 isoform co-localizes with RPGRIP1 at centrioles and basal bodies and interacts with nucleophosmin. Hum. Molec. Genet. 14: 1183-1197, 2005. [PubMed: 15772089] [Full Text: https://doi.org/10.1093/hmg/ddi129]

  44. Shu, X., Zeng, Z., Gautier, P., Lennon, A., Gakovic, M., Patton, E. E., Wright, A. F. Zebrafish Rpgr is required for normal retinal development and plays a role in dynein-based retrograde transport processes. Hum. Molec. Genet. 19: 657-670, 2010. [PubMed: 19955120] [Full Text: https://doi.org/10.1093/hmg/ddp533]

  45. Souied, E., Segues, B., Ghazi, I., Rozet, J.-M., Chatelin, S., Gerber, S., Perrault, I., Michel-Awad, A., Briard, M.-L., Plessis, G., Dufier, J.-L., Munnich, A., Kaplan, J. Severe manifestations in carrier females in X linked retinitis pigmentosa. J. Med. Genet. 34: 793-797, 1997. [PubMed: 9350809] [Full Text: https://doi.org/10.1136/jmg.34.10.793]

  46. van Dorp, D. B., Wright, A. F., Carothers, A. D., Bleeker-Wagemakers, E. M. A family with RP3 type of X-linked retinitis pigmentosa: an association with ciliary abnormalities. Hum. Genet. 88: 331-334, 1992. [PubMed: 1733835] [Full Text: https://doi.org/10.1007/BF00197269]

  47. Vervoort, R., Lennon, A., Bird, A. C., Tulloch, B., Axton, R., Miano, M. G., Meindl, A., Meitinger, T., Ciccodicola, A., Wright, A. F. Mutational hot spot within a new RPGR exon in X-linked retinitis pigmentosa. Nature Genet. 25: 462-466, 2000. [PubMed: 10932196] [Full Text: https://doi.org/10.1038/78182]

  48. Vervoort, R., Wright, A. F. Mutations of RPGR in X-linked retinitis pigmentosa (RP3). Hum. Mutat. 19: 486-500, 2002. [PubMed: 11968081] [Full Text: https://doi.org/10.1002/humu.10057]

  49. Yang, Z., Peachey, N. S., Moshfeghi, D. M., Thirumalaichary, S., Chorich, L., Shugart, Y. Y., Fan, K., Zhang, K. Mutations in the RPGR gene cause X-linked cone dystrophy. Hum. Molec. Genet. 11: 605-611, 2002. [PubMed: 11875055] [Full Text: https://doi.org/10.1093/hmg/11.5.605]

  50. Yokoyama, A., Maruiwa, F., Hayakawa, M., Kanai, A., Vervoort, R., Wright, A. F., Yamada, K., Niikawa, N., Naoi, N. Three novel mutations of the RPGR gene exon ORF15 in three Japanese families with X-linked retinitis pigmentosa. Am. J. Med. Genet. 104: 232-238, 2001. [PubMed: 11754050]

  51. Zeiss, C. J., Ray, K., Acland, G. M., Aguirre, G. D. Mapping of X-linked progressive retinal atrophy (XLPRA), the canine homolog of retinitis pigmentosa 3 (RP3). Hum. Molec. Genet. 9: 531-537, 2000. [PubMed: 10699176] [Full Text: https://doi.org/10.1093/hmg/9.4.531]

  52. Zhang, Q., Acland, G. M., Wu, W. X., Johnson, J. L., Pearce-Kelling, S., Tulloch, B., Vervoort, R., Wright, A. F., Aguirre, G. D. Different RPGR exon ORF15 mutations in Canids provide insights into photoreceptor cell degeneration. Hum. Molec. Genet. 11: 993-1003, 2002. [PubMed: 11978759] [Full Text: https://doi.org/10.1093/hmg/11.9.993]

  53. Zito, I., Downes, S. M., Patel, R. J., Cheetham, M. E., Ebenezer, N. D., Jenkins, S. A., Bhattacharya, S. S., Webster, A. R., Holder, G. E., Bird, A. C., Bamiou, D. E., Hardcastle, A. J. RPGR mutation associated with retinitis pigmentosa, impaired hearing, and sinorespiratory infections. J. Med. Genet. 40: 609-615, 2003. [PubMed: 12920075] [Full Text: https://doi.org/10.1136/jmg.40.8.609]


Contributors:
Jane Kelly - updated : 4/7/2016
Marla J. F. O'Neill - updated : 12/13/2013
Jane Kelly - updated : 5/22/2013
Patricia A. Hartz - updated : 4/26/2012
Patricia A. Hartz - updated : 8/31/2011
George E. Tiller - updated : 2/8/2011
George E. Tiller - updated : 11/12/2010
George E. Tiller - updated : 5/19/2008
Jane Kelly - updated : 4/16/2008
Marla J. F. O'Neill - updated : 2/1/2008
Jane Kelly - updated : 12/5/2007
Jane Kelly - updated : 11/20/2007
Cassandra L. Kniffin - updated : 10/9/2007
Cassandra L. Kniffin - updated : 5/29/2007
Victor A. McKusick - updated : 3/28/2007
Jane Kelly - updated : 9/13/2006
Marla J. F. O'Neill - updated : 7/6/2006
Jane Kelly - updated : 11/18/2005
Jane Kelly - updated : 6/2/2005
Cassandra L. Kniffin - updated : 5/13/2005
Jane Kelly - updated : 6/14/2004
Cassandra L. Kniffin - updated : 1/16/2004
Victor A. McKusick - updated : 12/12/2003
Jane Kelly - updated : 10/27/2003
Victor A. McKusick - updated : 10/1/2003
Jane Kelly - updated : 8/22/2003
George E. Tiller - updated : 7/9/2003
George E. Tiller - updated : 12/12/2002
George E. Tiller - updated : 10/9/2002
Victor A. McKusick - updated : 10/1/2002
Victor A. McKusick - updated : 6/11/2002
Victor A. McKusick - updated : 5/14/2002
Victor A. McKusick - updated : 4/12/2002
Victor A. McKusick - updated : 12/4/2001
Victor A. McKusick - updated : 10/12/2001
George E. Tiller - updated : 12/4/2000
Victor A. McKusick - updated : 10/19/2000
Victor A. McKusick - updated : 10/19/2000
Ada Hamosh - updated : 8/1/2000
Jane Kelly - updated : 6/28/2000
Victor A. McKusick - updated : 4/20/2000
George E. Tiller - updated : 4/14/2000
Victor A. McKusick - updated : 11/8/1999
Ada Hamosh - updated : 8/18/1999
Victor A. McKusick - updated : 2/25/1999
Michael J. Wright - updated : 6/5/1998
Victor A. McKusick - updated : 5/12/1998
Victor A. McKusick - updated : 2/25/1998
Victor A. McKusick - updated : 10/7/1997
Moyra Smith - updated : 8/21/1996
Iosif W. Lurie - updated : 7/4/1996
Moyra Smith - updated : 4/26/1996

Creation Date:
Victor A. McKusick : 10/12/1987

Edit History:
carol : 01/13/2023
alopez : 03/15/2022
carol : 01/08/2019
carol : 04/07/2016
carol : 11/19/2014
joanna : 1/22/2014
carol : 12/17/2013
mcolton : 12/13/2013
tpirozzi : 7/3/2013
carol : 5/22/2013
mgross : 5/2/2012
mgross : 5/2/2012
terry : 4/26/2012
mgross : 9/9/2011
terry : 8/31/2011
wwang : 3/14/2011
carol : 2/15/2011
terry : 2/8/2011
wwang : 11/19/2010
terry : 11/12/2010
alopez : 10/14/2010
joanna : 7/27/2010
alopez : 7/17/2009
alopez : 7/15/2009
alopez : 7/15/2009
alopez : 7/14/2009
carol : 3/9/2009
alopez : 2/16/2009
wwang : 10/30/2008
wwang : 5/22/2008
terry : 5/19/2008
carol : 4/16/2008
wwang : 2/5/2008
terry : 2/1/2008
carol : 12/5/2007
carol : 11/20/2007
wwang : 10/17/2007
ckniffin : 10/9/2007
carol : 7/18/2007
wwang : 6/11/2007
ckniffin : 5/29/2007
alopez : 4/3/2007
terry : 3/28/2007
carol : 2/28/2007
carol : 9/13/2006
wwang : 7/6/2006
wwang : 2/24/2006
alopez : 11/18/2005
tkritzer : 6/2/2005
tkritzer : 5/31/2005
ckniffin : 5/13/2005
terry : 7/19/2004
alopez : 6/14/2004
tkritzer : 2/18/2004
ckniffin : 1/16/2004
cwells : 12/19/2003
terry : 12/12/2003
ckniffin : 12/4/2003
tkritzer : 11/6/2003
tkritzer : 10/28/2003
tkritzer : 10/27/2003
tkritzer : 10/23/2003
tkritzer : 10/23/2003
tkritzer : 10/10/2003
tkritzer : 10/1/2003
carol : 8/22/2003
cwells : 7/9/2003
terry : 5/15/2003
tkritzer : 1/3/2003
tkritzer : 1/2/2003
cwells : 12/12/2002
cwells : 10/9/2002
carol : 10/2/2002
tkritzer : 10/1/2002
tkritzer : 10/1/2002
alopez : 6/18/2002
terry : 6/11/2002
alopez : 5/17/2002
terry : 5/14/2002
alopez : 4/19/2002
alopez : 4/17/2002
terry : 4/12/2002
carol : 1/2/2002
mcapotos : 12/10/2001
terry : 12/4/2001
mcapotos : 10/30/2001
mcapotos : 10/12/2001
carol : 12/4/2000
mcapotos : 11/28/2000
terry : 11/17/2000
carol : 11/6/2000
carol : 10/19/2000
terry : 10/19/2000
alopez : 8/2/2000
terry : 8/1/2000
alopez : 6/28/2000
mcapotos : 5/11/2000
mcapotos : 5/9/2000
terry : 4/20/2000
terry : 4/14/2000
carol : 3/13/2000
alopez : 11/12/1999
terry : 11/8/1999
alopez : 8/20/1999
terry : 8/18/1999
carol : 5/5/1999
carol : 3/9/1999
terry : 2/25/1999
dkim : 9/10/1998
alopez : 6/17/1998
terry : 6/5/1998
carol : 5/26/1998
alopez : 5/21/1998
terry : 5/12/1998
joanna : 5/7/1998
mark : 2/25/1998
mark : 2/25/1998
mark : 2/23/1998
terry : 12/11/1997
mark : 10/8/1997
terry : 10/7/1997
mark : 11/4/1996
jamie : 10/23/1996
jamie : 10/16/1996
mark : 8/21/1996
terry : 8/21/1996
mark : 8/20/1996
carol : 7/4/1996
carol : 6/27/1996
carol : 4/26/1996
mark : 1/10/1996
terry : 1/5/1996
carol : 11/18/1994
davew : 7/5/1994
mimadm : 2/28/1994
carol : 6/7/1993
carol : 1/27/1993
carol : 1/19/1993