Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Feb 22.
Published in final edited form as: J Med Genet. 2013 Jul 11;50(10):674–688. doi: 10.1136/jmedgenet-2013-101558

Comprehensive molecular diagnosis of 179 Leber congenital amaurosis and juvenile retinitis pigmentosa patients by targeted next generation sequencing

Xia Wang 1,2, Hui Wang 1,2, Vincent Sun 3, Han-Fang Tuan 1, Vafa Keser 3, Keqing Wang 2, Huanan Ren 3, Irma Lopez 3, Jacques E Zaneveld 1,2, Sorath Siddiqui 3, Stephanie Bowles 1, Ayesha Khan 3, Jason Salvo 1,4, Samuel G Jacobson 5, Alessandro Iannaccone 6, Feng Wang 1,2, David Birch 7, John R Heckenlively 8, Gerald A Fishman 9, Elias I Traboulsi 10, Yumei Li 1,2, Dianna Wheaton 7, Robert K Koenekoop 3, Rui Chen 1,2,4,11
PMCID: PMC3932025  NIHMSID: NIHMS543701  PMID: 23847139

Abstract

Background

Leber congenital amaurosis (LCA) and juvenile retinitis pigmentosa (RP) are inherited retinal diseases that cause early onset severe visual impairment. An accurate molecular diagnosis can refine the clinical diagnosis and allow gene specific treatments.

Methods

We developed a capture panel that enriches the exonic DNA of 163 known retinal disease genes. Using this panel, we performed targeted next generation sequencing (NGS) for a large cohort of 179 unrelated and prescreened patients with the clinical diagnosis of LCA or juvenile RP. Systematic NGS data analysis, Sanger sequencing validation, and segregation analysis were utilised to identify the pathogenic mutations. Patients were revisited to examine the potential phenotypic ambiguity at the time of initial diagnosis.

Results

Pathogenic mutations for 72 patients (40%) were identified, including 45 novel mutations. Of these 72 patients, 58 carried mutations in known LCA or juvenile RP genes and exhibited corresponding phenotypes, while 14 carried mutations in retinal disease genes that were not consistent with their initial clinical diagnosis. We revisited patients in the latter case and found that homozygous mutations in PRPH2 can cause LCA/juvenile RP. Guided by the molecular diagnosis, we reclassified the clinical diagnosis in two patients.

Conclusions

We have identified a novel gene and a large number of novel mutations that are associated with LCA/juvenile RP. Our results highlight the importance of molecular diagnosis as an integral part of clinical diagnosis.

INTRODUCTION

Leber congenital amaurosis (LCA) refers to a group of inherited retinal dystrophies that share the common feature of severe visual impairment within the first year of life. Clinically, LCA is defined by congenital blindness, congenital nystagmus, and lack of detectable signals on an electroretinogram (ERG).1,2 LCA affects 1 in every 50 000 individuals, but it accounts for 5% of all retinal dystrophies and 20% of blindness in school age children.3,4 To date, mutations in 19 genes are reported to cause LCA.512 Despite the breadth of current knowledge, genetic defects in about 30% of LCA cases remain unknown.11

The clinical phenotypes and genetic causes of LCA and juvenile retinitis pigmentosa (RP) largely overlap. Both diseases belong to a spectrum of retinal diseases termed early onset retinal dystrophies (EORD). In fact, LCA was initially considered to be a congenital form of RP.2 Compared with LCA, juvenile RP tends to have milder phenotypes and a later onset. Juvenile RP patients appear to have better visual function at birth than those with LCA, and later develop night blindness, narrowed visual fields, and eventually severe vision impairment. Mutations in several known LCA genes, such as CRB1 and RDH12, are reported to cause juvenile RP. 13 Interestingly, mutations in other retinal disease genes, such as IQCB1 and KCNJ13, are also known to be associated with LCA or ‘LCA-like’ phenotypes.10,11 These observations may be explained by a combination of allelic differences, genetic background, and environmental modifications. Also, it has been demonstrated that the clinical phenotypes of many retinal diseases overlap with that of LCA.11 It is likely that in some cases visual impairment is the most obvious phenotype in the initial evaluation, and that other syndromic features appear at a later time. Therefore, given the limited evaluation possible in infants and in early childhood, some patients initially diagnosed with LCA may actually have a different retinal disorder, such as Alström syndrome or Joubert syndrome.11 Despite these observations, systematic screening for mutations in all known retinal disease genes on a large LCA patient cohort has not yet been reported.

Because of the genetic heterogeneity of LCA and other retinal diseases, an accurate molecular diagnosis can improve the clinical diagnosis, facilitate a more accurate description of prognosis, and allow gene specific treatment. One of the most common methods for molecular diagnosis of LCA is the Arrayed Primer Extension (APEX) chip (Asper Ophthalmics). It is a microarray based genotyping method that tests a subset of known mutations in known LCA genes, leading to molecular diagnosis in approximately 17–32% of LCA patients.1416 With additional mutations added to the LCA APEX array, the estimated solving rate has been improved to about 50%.11 On the other hand, next generation sequencing (NGS) has been recently used for the molecular diagnosis of retinal diseases.17,18 Compared with the APEX chip, the NGS based approach is able to discover novel variants and genes. Recently, Coppieters and others described a workflow to screen the exons of known LCA genes, using amplicon PCR followed by NGS.19 However, this workflow was tested on a relatively small LCA patient cohort and did not cover other retinal disease genes.

The goal of this study was to develop a comprehensive molecular diagnostic method for LCA and potentially for other retinal diseases. For this purpose, we developed a targeted NGS method that allows us to systematically screen the exons of most known retinal disease genes at low cost (163 genes at the time of design, online supplementary files 1 and 2). We first evaluated this method on a standard control sample, and then applied it to the molecular diagnosis of a large cohort of unrelated and prescreened patients with the clinical diagnosis of either LCA or juvenile RP (n=179). Pathogenic mutations for 72 patients were identified by systematic NGS data analysis, Sanger sequencing validation, and segregation analysis. These 72 patients were classified into different confidence groups based on the clinical significance of their mutations. Among the 72 patients, 58 carried mutations in known LCA or juvenile RP genes and exhibited corresponding phenotypes, while 14 carried mutations in retinal disease genes that were not consistent with their initial clinical diagnosis. Guided by the molecular diagnosis, we revisited 12 out of the 14 patients. We found that homozygous mutations in PRPH2 can cause LCA/juvenile RP. We also reclassified or refined the initial clinical diagnosis for 10 patients.

METHODS

Study subjects

We initially collected a cohort of 389 patients from around the world and with a variety of backgrounds. Using a combination of LCA APEX array, Sanger sequencing, homozygosity mapping, and phenotype directed genotyping methods (eg, preserved para-arteriolar retinal pigment epithelium in an LCA patient is associated with mutations in CRB1), we had previously identified the genetic causes for 210 patients (most of whom are LCA patients).13,20 The remaining 179 patients were included in this study. The available prescreening information for the 179 patients is listed in online supplementary table S5.

The 179 patients were seen at McGill University (RKK), University of Pennsylvania (SGJ), The Lighthouse of Chicago (GAF), University of Tennessee Health Science Center (AI), and University of Michigan (JRH), by ophthalmologists with expertise in retinal dystrophies. Informed consents and research protocols were approved by the respective institutional review boards or research ethics board and adhered to the tenets of the Declaration of Helsinki. Complete histories, pedigree analysis, and ophthalmic examinations were performed. Eye exams consisted of cycloplegic refractions, fixation testing, Snellen visual acuities (when possible), pupillary responses, slit lamp exams, dilated fundus exam by indirect ophthalmoscopy, retinal photography, and Goldmann visual field testing (when possible). In most cases, ERGs were done according to ISCEV (International Society for Clinical Electrophysiology of Vision) standards. LCA is defined by the phenotypes mentioned in the introduction and the absence of overt systemic features. Juvenile RP represents a milder disease with later onset of signs and symptoms. In juvenile RP patients, vision can appear normal at birth, and the first symptom is progressive night blindness, with progressive visual loss at around age 2 years, with or without nystagmus.

DNA was extracted from whole blood using the FlexiGene kit or the QIAamp DNA blood kit according to the manufacturer’s protocol. The quantity and quality of DNA were verified by using NanoDrop.

Target DNA capture and NGS experiments

According to the manufacturer’s protocol, Illumina paired-end libraries were generated. Briefly, ∼1 µg of genomic DNA was sheared into fragments of approximately 300–500 bp. The DNA fragments were end-repaired and an extra ‘adenine’ base was added to the 3' end. Illumina Y-shape index adapters were ligated to the ends of the DNA fragments and eight cycles of PCR amplification were applied to each sample after ligation. The DNA libraries were quantified by the PicoGreen assay (Invitrogen). For each capture reaction, 24 to 48 libraries (3 µg of DNA in total) were pooled together. A design file (see online supplementary files 1 and 2) was submitted to Nimblegen for the design of the capture probe. NimbleGen SeqCap EZ Hybridisation and Wash Kits were used for the washing and recovery of captured DNA. Captured libraries were quantified and sequenced on the Illumina HiSeq 2000 as 100 bp paired-end reads, following the manufacturer’s protocols. Illumina sequencing was performed at the BCM-FGI core.

Evaluation of our method's sensitivity to detect SNPs on the Hapmap sample

Single nucleotide polymorphisms (SNPs) genotyping data of HapMap sample NA11831 were downloaded from 1000 Genome omni database (ftp://ftp-trace.ncbi.nih.gov/1000genomes/ftp/technical/working/20110527_bi_omni_1525_v2_genotypes/). This sample had been genotyped using the Illumina OMNI2.5 SNP genotyping array. A total of 1190 genotyped single nucleotide polymorphisms (SNPs) in this sample are within our design region, including 919 homozygous reference SNPs, 107 homozygous alternative SNPs, and 164 heterozygous alternative SNPs. A total of 1184 SNPs were detected by our targeted NGS method. Among the detected SNPs, 1183 out of 1184 had the same genotype between the SNP array and NGS. The single disconcordant SNP rs3763073 was heterozygous C/T on the SNP array but homozygous C/C in targeted NGS. To resolve the conflict, we performed direct Sanger sequencing and confirmed that rs3763073 was indeed homozygous for the reference C, indicating that NGS detected the SNP correctly (data not shown).

Data analysis

Sequencing reads were aligned to the human genome reference version hg19 using Burrows-Wheeler Aligner (BWA).21 Base quality recalibration and local realignment were performed using the Genome Analysis Toolkit (GATK).22 AtlasSNP was used for SNP calling and AtlasIndel2 was used for indel calling.23 The 1000 genome database, dbSNP, ESP5400, NIEHS95 exomes, and our internal database were used to filter out common SNPs and indels, with allele frequency cutoffs at 0.5% for recessive variants and at 0.1% for dominant variants (Exome Variant Server, NHLBI GO Exome Sequencing Project (ESP), Seattle, Washington (http://evs.gs.washington.edu/EVS/), NIEHS Environmental Genome Project, Seattle, Washington (http://evs.gs.washington.edu/niehsExome/).24,25 Variant annotation was performed using ANNOVAR.26 The Refseq gene sequences below were used for the mutation coordinates: AIPL1:NM_014336, ALMS1:NM_ 015120, BBS1:NM_024649, BBS7:NM_018190, CEP290:NM_ 025114, CERKL:NM_001160277, CLN3:NM_001042432, CRB1:NM_201253, GUCY2D:NM_000180, INPP5E:NM_ 019892, IQCB1:NM_001023570, LCA5:NM_181714, LRAT: NM_004744, NR2E3: NM_016346, OTX2:NM_172337, PDE6A:NM_000440, PRPF31:NM_015629, RDH12:NM_ 152443, RPE65:NM_000329, RPGR: NM_000328, RPGRIP1: NM_020366, SAG:NM_000541, SNRNP200:NM_014014, SPATA7:NM_018418, TULP1:NM_003322. The pathogenicity of novel missense mutations was predicted by dbNSFP, whose prediction score is derived from five algorithms (SIFT, Polyphen2, LRT, MutationTaster, and PhyloP).2732

PCR and direct Sanger sequencing

To validate the mutations detected by NGS, primers were designed (Primer3, http://biotools.umassmed.edu/bioapps/primer3_www.cgi) to PCR-amplify the 400–500 bp region flanking the mutation. To ensure the high quality of Sanger sequencing, the amplicon was designed to have a boundary at least 50 bp away from the mutation. The amplicon was then Sanger sequenced on Applied BioSystems (ABI) 3730×l capillary sequencer. The Sanger sequencing results were analysed with Sequencher software. The intronic mutation c.2991+1655A>G in CEP290 was not initially included in the original design of our exonic capture panel. Sanger sequencing of this mutation was performed and the results were combined with the NGS data.

RESULTS

A cohort of 179 patients clinically diagnosed with LCA or juvenile RP

After prescreening for known mutations in LCA and juvenile RP genes using a combination of conventional genotyping methods, the genetic defects in 173 LCA and six juvenile RP patients remained unexplained (see online supplementary table S1 and Methods). We hypothesised that a portion of these cases were caused by mutations in known LCA and juvenile RP genes that were not included in the conventional screening methods, or caused by mutations in other retinal disease genes that had not been previously associated with LCA or juvenile RP. To Sanger-sequence all known retinal disease genes for such a large sample set would be prohibitively expensive and time consuming. Therefore, we utilised a targeted NGS based method for the comprehensive molecular diagnosis of these patients.

Targeted NGS of a standard control sample from HapMap project

A capture panel was designed to enrich the target DNA, which consisted of 649 804 bp covering 2560 exons in 163 known retinal disease genes that had been reported and recorded in the RetNet at the time of design (see online supplementary files 1 and 2, https://sph.uth.tmc.edu/retnet/). The enriched DNA was then sent for NGS.

We first evaluated the coverage of our targeted NGS method on NA11831, a standard control sample from the original HapMap Centre d’Etude du Polymorphisme Humain (CEPH) cohort.33 DNA from NA11831 was captured and sequenced at high coverage. A total of 8 240 805 mappable reads were obtained, 39% of which mapped to the target region and resulted in a 234× mean per base coverage. As shown in figure 1A and B, the vast majority of the targeted regions were well covered. Indeed, 97% of the bases in target region had coverage >10× and 92% of the bases had coverage >50× (figure 1A). Also, 98% of the 2560 exons had mean coverage >5× (figure 1B). The low coverage exons were either within duplicate regions or those with a high GC content (see online supplementary tables S2 and S3).

Figure 1.

Figure 1

Systematic evaluation of targeted next generation sequencing method on a Hapmap sample NA11831. (A) The number of bases within different coverage groups. (B) The mean coverage of 2560 exons in the design region. (C) The sensitivity to detect single nucleotide polymorphisms versus the according mean coverage. (D) The high and uniform mean coverage for 12 multiplexed replicates. The blue dot line represents the average coverage for 12 replicates.

To systematically evaluate the accuracy of our method, we compared the genotyping data obtained from NGS to that from the SNP array. As part of the 1000 Genome project, sample NA11831 had been genotyped using the Illumina OMNI2.5 genotyping array. A total of 1190 genotyped SNPs in this sample were within our design region and were used as standards to test the accuracy of our method. As a result, 99.5% of SNPs (1184/ 1190) were detected by NGS (minimum coverage=3). The six undetected SNPs were within low coverage exons (data not shown). The genotypes of all 1184 NGS SNP calls were validated by either SNP array or Sanger sequencing (see Methods). Therefore, high quality SNP genotyping results were obtained by targeted NGS with a sensitivity of 99.5% (1184/1190) and a genotype concordance of 100% (1184/1184).

To further explore the effect of coverage on the sensitivity of SNP detection, sequencing reads generated from NA11831 were randomly sampled in silico to achieve different levels of coverage from 3× to 234×. As shown in figure 1C, sensitivity increased sharply from 38% to 96% as the coverage increased from 2× to 12×, then gradually reached a sensitivity of 99% at around 23×. Based on this result, we chose to sequence patient samples at around 50× coverage to achieve nearly saturated sensitivity with a relatively low cost (cost is linear to the depth of coverage). At 50× coverage, up to 100 samples could be sequenced in one lane of Illumina HiSeq 2000. To develop a more cost effective method, we tested the robustness of sample multiplexing. We molecularly barcoded 12 replicates of NA11831 DNA and performed targeted NGS for these 12 replicates in one capture reaction. As shown in figure 1D, uniform and high coverage of these replicates was achieved.

Targeted NGS of 179 patients

Using the capture panel described above, we applied targeted NGS to DNA obtained from a large cohort of 179 unrelated patients with the diagnosis of LCA or juvenile RP. The sequencing reads were processed by our bioinformatics pipeline that performed reads alignment, recalibration, realignment, variants calling, filtering, annotation, and quality control (see Methods). An average of 62× coverage was achieved for the 179 patient samples. Within the design region, 84% of bases had coverage >10× and 70% of bases had coverage >20×, indicating that sufficient coverage was achieved for high sensitivity of variants detection (table 1, figure 2A). For each individual, about 407 SNPs and small insertions/deletions (indels) were identified. Since LCA and juvenile RP are rare Mendelian diseases, common variants with a frequency >0.5% (for recessive variants) or >0.1% (for dominant variants) in any of the following databases were filtered out: the 1000 genome database, dbSNP135, the ESP5400 database, the NIEHS 95 exomes database, and our internal database (see Methods). As a result, an average of eight rare variants in retinal disease genes that lead to protein coding change were identified per sample (table 1). Furthermore, mutations known to cause retinal diseases in the Human Gene Mutation Database (HGMD) or the primary literature were identified.34 Finally, dbNSFP, a program that compiles prediction score from five well established prediction algorithms (PhyloP, SIFT, Polyphen2, LRT, and MutationTaster), was used to predict the pathogenicity of novel missense changes.2732 In this study, we only reported novel missense variants that are predicted to be pathogenic by at least three of the five algorithms (see online supplementary table S4). After all these stringent filtering steps, the remaining variants are likely to cause the disease in patients.

Table 1.

Target NGS statistics for 179 patients

Per base coverage 62
% Target bp covered >1× 97
% Target bp covered >10× 84
% Target bp covered >20× 70
Total number of variants (SNPs and indels) 407
Rare variants 24
Rare variants that lead to protein coding change: 8.23
  Missense change 3.81
  Nonsense change 0.28
  Splicing site change 0.15
  Non-frameshift indels 1.56
  Frameshift indels 2.46

All the values are the mean number derived from 179 patients.

NGS, next generation sequencing; SNPs, single nucleotide polymorphisms.

Figure 2.

Figure 2

The targeted next generation sequencing statistics for 179 patients. (A) The percentage of bases in design region in each coverage group for 179 patients. (B) The percentage of 179 patients in different categories. (C) The percentage of different types of pathogenic mutations identified in the 72 patients. LCA, Leber congenital amaurosis.

Identification of pathogenic mutations

To identify the potential pathogenic mutations among several rare variants in each patient, we looked for variants that matched the reported inheritance pattern of the respective genes:

  1. Homozygous or compound heterozygous variants in recessive retinal disease genes, or

  2. Reported heterozygous variants known to cause dominant retinal diseases, or

  3. Novel heterozygous loss-of-function (LOF) variants in dominant retinal disease genes, if heterozygous LOF mutations in those genes are previously known to cause dominant retinal diseases.

All potential pathogenic variants identified above were validated by Sanger sequencing. Segregation analysis was performed if DNA from family members was available. Through this procedure, we identified pathogenic mutations for 72 out of 179 patients (40%). Among the 72 patients, 58 patients carried mutations in known LCA or juvenile RP genes and exhibited corresponding phenotypes, while 14 harboured mutations in retinal disease genes that were not consistent with their initial clinical diagnosis (figure 2B). A total of 83 distinct pathogenic mutations were identified in the 72 patients, including a large number of novel mutations (n=45) (table 2). Most of these mutations were missense (39%) and nonsense (35%) mutations (figure 2C).

Table 2.

Number of pathogenic mutations identified in this study

Reported Novel Total
LCA and juvenile RP genes 30 38 68
Retinal disease genes inconsistent with initial
diagnosis
8 7 15
Total 38 45 83

LCA, Leber congenital amaurosis; RP, retinitis pigmentosa.

Molecular diagnosis of patients

Patients carrying mutations in known LCA or juvenile RP genes

In total, we identified 58 patients who carried mutations in known LCA or juvenile RP genes and exhibited corresponding phenotypes (tables 46). According to the American College of Medical Genetics standards to report sequence variants, mutations identified in our study can be classified into three categories with different clinical significance: (1) reported mutations that are known to cause retinal diseases; (2) novel LOF mutations that are expected to cause retinal diseases; (3) novel missense mutations that are predicted to be pathogenic by in silico prediction algorithms and may be causative of retinal diseases (see online supplementary table S4).35 To demonstrate the different confidence levels for different patients, we classified these patients into three groups based on the clinical significance of their mutations: patients in group 1 and 2 carried reported or novel LOF mutations with higher confidence, while patients in group 3 harboured one or more novel missense mutations with lower confidence (table 3).

Table 4.

Twenty-six patients carrying two reported mutations in LCA or juvenile RP genes

Patient ID Disease presentation Gene Type Mutations
1473 LCA AIPL1 Homozygous c.834G>A, p.W278X38
3745 LCA AIPL1 Compound Heterozygous c.834G>A, p.W278X38
c.547G>T, p.G183X4
3746 LCA AIPL1 Compound Heterozygous c.834G>A, p.W278X38
c.547G>T, p.G183X4
393 LCA AIPL1 Homozygous c.487C>T, p.Q163X39
3754 LCA AIPL1 Compound Heterozygous c.265T>C, p.C89R15
c.214T>C, p.W72R4
3638 LCA CEP290 Homozygous c.2991+1655A>G40
3656 LCA CEP290 Compound Heterozygous c.5668G>T, p.G1890X41
c.2991+1655A>G40
3661 LCA CEP290 Homozygous c.2991+1655A>G40
3793 LCA CEP290 Homozygous c.4723A>T, p.K1575X42
398 LCA CRB1 Homozygous c.610_616del, p.I205DfsX13343
3738 LCA CRB1 Compound Heterozygous c.1438T>C, p.C480R43
c.2945C>A, p.T982K4
1251 LCA CRB1 Homozygous c.3996C>A, p.C1332X43
3722 LCA GUCY2D Compound Heterozygous c.1343C>A, p.S448X44
c.2598G>C, p.K866N45
3778 LCA GUCY2D Homozygous c.1343C>A, p.S448X44
3750 LCA GUCY2D Compound Heterozygous c.2302C>T, p.R768W46
c.3271C>T, p.R1091X4
3577 LCA LCA5 Homozygous c.835C>T, p.Q279X47
54 LCA LRAT Homozygous c.217_218del, p.M73DfsX4848
622 LCA RDH12 Compound Heterozygous c.146C>T, p.T49M37
c.805_809del, p.A269GfsX237
1256 LCA RDH12 Compound Heterozygous c.146C>T, p.T49M37
c.805_809del, p.A269GfsX237
1278 Juvenile RP RDH12 Homozygous c.164C>T, p.T55M36
3916 LCA RDH12 Compound Heterozygous c.582C>G, p.Y194X36
c.805_809del, p.A269GfsX237
3784 LCA RPE65 Compound Heterozygous c.1205G>A, p.W402X4
c.1022T>C, p.L341S49
1259 LCA SPATA7 Homozygous c.322C>T, p.R108X9
1303 LCA TULP1 Homozygous c.1381C>G, p.L461V13
3670 LCA TULP1 Homozygous c.901C>T, p.Q301X50
3671 LCA TULP1 Homozygous c.901C>T, p.Q301X50

LCA, Leber congenital amaurosis; RP, retinitis pigmentosa.

Table 6.

Ten patients carrying one or more novel missense mutations in LCA or juvenile RP genes

Patient ID Disease presentation Gene Type Mutations
3319 LCA CRB1 Homozygous c.1439G>C, p.C480S
3611 LCA GUCY2D Homozygous c.2132C>T, p.P711L
3799 LCA GUCY2D Compound Heterozygous c.743C>G, p.S248W
c.3224+1G>C
3725 LCA GUCY2D Compound Heterozygous c.1343C>A, p.S448X44
c.2678C>T, p.S893F
1272 LCA GUCY2D Compound Heterozygous c.1933T>C, p.S645P
c.2207T>G, p.M736R
1313 Juvenile RP PDE6A Compound Heterozygous c.2333A>T, p.D778V
c.1363A>T, p.K455×
3740 LCA RDH12 Compound Heterozygous c.692G>A, p.G231D
c.823G>T, p.E275×
1268 LCA TULP1 Compound Heterozygous c.1518C>A, p.F506L
c.1277C>T, p.P426L
3771 LCA TULP1 Compound Heterozygous c.1199G>A, p.R400Q58
c.961T>G, p.Y321D
3681 LCA TULP1 Compound Heterozygous c.1102G>T, p.G368W59
c.1064A>T, p.D355V

LCA, Leber congenital amaurosis; RP, retinitis pigmentosa.

Table 3.

Classification of patients according to clinical significance of mutations

Mutated genes Group Allele 1 Allele 2 Homo Compound hetero Hetero Total
LCA/juvenile RP genes 1 Reported Reported 15 11 26
2 Novel LOF Novel LOF 8 5 13
Novel LOF Reported 8 8
Novel LOF 1 1
3 Novel missense Novel missense 2 2 4
Novel missense Reported 3 3
Novel missense Novel LOF 3 3
Retinal disease genes inconsistent 1 Reported Reported 7 7
with initial diagnosis Reported 1 1
2 Novel LOF Novel LOF 1* 1 2
3 Novel missense Novel missense 4 4
Total 37 33 2 72

The numbers in the four columns on the right represent the number of patients in each category.

*

This patient carried a hemizygous mutation.

Compound hetero, compound heterozygous; Hetero, Heterozygous; Homo, homozygous; LCA, Leber congenital amaurosis; RP, retinitis pigmentosa.

We identified 26 patients in group 1 who carried homozygous or compound heterozygous mutations that were known to cause recessive LCA or juvenile RP (tables 3 and 4, online supplementary table S1). For example, patient 3916 carried compound heterozygous reported nonsense mutations c.582C>G (p.Y194X) and frameshift deletion c.805_809del (p. A269GfsX2) in RDH12 (table 4). The patient exhibited LCA phenotypes and the two mutations were previously known to cause LCA (see online supplementary figure S1, table S1).36,37 In this group of patients, AIPL1 was the most frequently mutated gene, which appeared in five patients. The nonsense mutation c.834G>A (p.W278X) in AIPL1, the intronic mutation c.2991+1655A>G in CEP290, and the frameshift insertion c.805_809del (p.A269GfsX2) in RDH12 were the most frequent mutations, all carried by three patients.

We identified 22 patients in group 2 who carried novel LOF mutations in known LCA or juvenile RP genes (tables 3 and 5). First, 13 patients carried homozygous or compound heterozygous novel LOF mutations. For example, a novel homozygous frameshift deletion c.613_614del (p.S205YfsX27) was identified in exon3 of LRAT in patient 4019. To our knowledge, this is the first reported disease allele outside LRAT exon2.48,5153 The c.613_614del is predicted to change the 205–230 amino acids in the C terminus of LRAT protein, which is thought to be important for the LRAT protein enzymatic activity and its localisation to the endoplasmic reticulum membrane.54,55 Second, eight patients carried one reported mutation plus one novel LOF mutation. Third, patient 3561 carried a novel heterozygous frameshift insertion in OTX2. This insertion is likely to be pathogenic because a heterozygous protein truncating mutation in OTX2 was previously reported to cause ocular malformation and LCA.56 In this group of patients, CEP290 was the most frequently mutated gene, which appeared in seven patients.

Table 5.

Twenty-two patients carrying novel LOF mutations in LCA or juvenile RP genes

Patient ID Disease presentation Gene Type Mutations
3739 LCA CEP290 Compound Heterozygous c.5344C>T, p.R1782×
c.1667_1668insA, p.I556Nfs×20
3640 LCA CEP290 Compound Heterozygous c.1260_1264del, p.K421Gfs×2
c.2991+1655A>G
3645 LCA CEP290 Compound Heterozygous c.3811C>T, p.R1271×
c.2991+1655A>G40
3650 LCA CEP290 Compound Heterozygous c.547_550del, p.Y183Rfs×4
c.2991+1655A>G40
3653 LCA CEP290 Compound Heterozygous c.4882C>T, p.Q1628×
c.2991+1655A>G40
3666 LCA CEP290 Compound Heterozygous c.1219_1220del, p.M407Efs×14
c.2991+1655A>G40
3741 LCA CEP290 Compound Heterozygous c.4723A>T, p.K1575×42
c.2052+1_2052+2del
418 LCA CRB1 Homozygous c.984G>A, p.W328×
3557 LCA CRB1 Homozygous c.3687C>A, p.C1229×
1413 LCA GUCY2D Homozygous c.1116G>A, p.W372×
3796 LCA IQCB1 Compound Heterozygous c.1518_1519del, p.H506QfsX1357
c.1381C>T, p.R461×
3752 LCA IQCB1 Compound Heterozygous c.1518_1519del, p.H506Qfs×1357
c.1465C>T, p.R489×
3737 LCA IQCB1 Compound Heterozygous c.1465C>T, p.R489×
c.1381C>T, p.R461×
4019 LCA LRAT Homozygous c.613_614del, p.S205Yfs×27
3561 LCA OTX2 Heterozygous c.543_544insCTCA, p.Q181Hfs×7
1842 Juvenile RP PDE6A Homozygous c.205C>T, p.Q69×
3676 LCA RPGRIP1 Compound Heterozygous c.1083_1084insGA, p.V364Efs×12
c.3749-1G>T
3677 LCA RPGRIP1 Compound Heterozygous c.1083_1084insGA, p.V364Efs×12
c.3749-1G>T
1315 LCA SPATA7 Homozygous c.1216—1G>A
3679 LCA SPATA7 Homozygous c.1373del, p.V458Efs×48
3757 LCA TULP1 Compound Heterozygous c.1376_1377del, p.I459Rfs×12
1271 LCA TULP1 Homozygous c.725_728del, p.P242Qfs×16
c.1113—2A>C

LCA, Leber congenital amaurosis; LOF, loss-of-function; RP, retinitis pigmentosa.

We identified 10 patients in group 3 who carried one or more novel missense mutations in known LCA or juvenile RP genes (table 6). Specifically, four patients carried homozygous or compound heterozygous novel missense mutations, three patients had a novel missense mutation plus a reported mutation, and three patients had a novel missense plus a novel LOF mutation (table 3). For example, patient 3319 carried a homozygous novel missense mutation (c.1439G>C, p.C480S) in CRB1 that changes a cysteine to a serine. The cysteine is conserved across mammals and this mutation is predicted to be damaging to protein function/structure by in silico prediction (see online supplementary table S4). Interestingly, similar missense mutations p.C480R and p.C480G at this residue were reported to cause LCA, further supporting the pathogenicity of p.C480S.43 In this group of patients, GUCY2D was the most frequently mutated gene, which appeared in four patients.

Patients carrying mutations in other retinal disease genes

We also identified 14 patients who carried mutations in retinal disease genes that were not consistent with their initial clinical diagnosis, representing 19% of the 72 diagnosed patients. Using the criteria mentioned above, we classified these 14 patients into three groups based on the clinical significance of their mutations.

We identified eight patients in group 1 who carried reported mutations known to cause retinal disease genes that were not consistent with their initial clinical diagnosis (tables 3 and 7). Within this group, seven patients carried homozygous mutations. In addition, juvenile RP patient 3311 carried a heterozygous reported mutation known to cause autosomal dominant RP (adRP).

Table 7.

Eight patients carrying mutations known to cause other retinal diseases

Patient ID Disease presentation Gene Previously reported disease Type Mutations
704 Juvenile RP BBS1 BBS Homozygous c.1169T>G, p.M390R60
647 LCA CERKL Cone–rod dystrophy Homozygous c.375C>G, p.C125W61
3748 LCA CLN3 Batten disease Homozygous c.597C>A, p.Y199X62
617 LCA NR2E3 Enhanced S-cone syndrome Homozygous c.119—2A>C63
3311 Juvenile RP PRPF31 adRP Heterozygous c.220C>T, p.Q74X64
1318 LCA PRPH2 adRP Homozygous c.637T>C, p.C213R65
3256 LCA PRPH2 adRP Homozygous c.554T>C, p.L185P66
3425 LCA SAG Oguchi disease Homozygous c.874C>T, p.R292X67

BBS, Bardet–Biedl syndrome; LCA, Leber congenital amaurosis; RP, retinitis pigmentosa.

We identified two LCA patients in group 2 who carried homozygous or compound heterozygous novel LOF mutations in other retinal disease genes (tables 3 and 8). For example, patient 3688 carried a hemizygous novel splice site mutation c.248–1G>T in RPGR. Previously reported splice site mutations in RPGR were known to cause X-linked RP, supporting that this mutation may cause the retinal defects in patient 3688.

Table 8.

Two patients carrying novel LOF mutations in other retinal disease genes

Patient ID Disease presentation Gene Previously reported disease Type Mutations
3494 LCA ALMS1 Alström syndrome Compound Heterozygous c.2996C>G, p.S999X
c.11410C>T, p.R3804×
3688 LCA RPGR X-linked RP Hemizygous c.248–1G>T

LCA, Leber congenital amaurosis; LOF, loss-of-function; RP, retinitis pigmentosa.

We identified four patients in group 3 who carried homozy-gous novel missense mutations in retinal diseases genes that were not consistent with their initial clinical diagnosis (tables 3 and 9). For example, patient 1327 carried a homozygous novel missense mutation c.728G>A (p.C243Y) in Bardet–Biedl syndrome (BBS) gene BBS7. This mutation changes a cysteine residue that is conserved across vertebrates. It was predicted to be damaging by all of the five in silico prediction algorithms, supporting that this mutation is likely to be pathogenic (see online supplementary table S4).

Table 9.

four patients carrying homozygous novel missense mutations in other retinal disease genes

Patient ID Disease presentation Gene Previously reported disease Type Mutations
3779 LCA ALMS1 Alström syndrome Homozygous c.9764C>G, p.S3255C
1327 Juvenile RP BBS7 BBS Homozygous c.728G>A, p.C243Y
3773 LCA INPP5E Joubert syndrome Homozygous c.1861C>T, p.R621W
3795 LCA SNRNP200 adRP Homozygous c.3133C>A, p.P1045T

BBS, Bardet–Biedl syndrome; LCA, Leber congenital amaurosis; adRP, autosomal dominant retinitis pigmentosa.

Revisiting patients carrying mutations in other retinal disease genes

In our study we observed that 14 patients carried mutations in genes that were not consistent with their initial clinical diagnosis. This observation may be explained by novel genotype– phenotype correlations, or by the difficulty assigning clinical diagnosis at the time of initial visit. In most cases, the first visit of a blind or low vision infant occurs shortly after birth. The initial clinical diagnosis may be difficult and influenced by the most obvious ophthalmic and visual findings at that time. To test these two possibilities, we managed to revisit 12 of these 14 patients.

Homozygous mutations in PRPH2 cause EORD with LCA/juvenile RP phenotypes

After revisiting, we confirmed the clinical diagnosis of LCA in patients 1318 and 3256 (figure 3, online supplementary table S1). Each patient carried a reported homozygous missense mutation in gene PRPH2: c.637T>C (p.C213R) and c.554T>C (p.L185P), respectively (table 7). PRPH2 encodes peripherin, a membrane glycoprotein that is important for the stabilisation and compaction of photoreceptor outer segment discs.68 The p.C213R mutation is associated with autosomal dominant pattern dystrophy, and the p.L185P mutation, together with a null mutation in ROM1, has been reported to cause digenic RP.65,66 However, it has not been reported that homozygous mutations in PRPH2 cause severe EORD. To further validate this finding, we sequenced PRPH2 in another 135 unsolved LCA or juvenile RP patients and found the same homozygous missense mutation p.L185P in PRPH2 in a third juvenile RP patient, 741. These mutations were confirmed by Sanger sequencing and their segregations with the disease in the families were examined (figure 4). All the patients with homozygous mutations in PRPH2 exhibited LCA or juvenile RP phenotypes, including visual impairment within the first year of life, nystagmus in the two LCA patients (1318 and 3256), non-detectable or reduced ERGs, and a very similar form of maculopathies in the fundus (figure 3, online supplementary table S1). By contrast, family members who carried heterozygous mutations in PRPH2 were asymptomatic but showed detectable maculopathy phenotypes. For example, the 56-year-old father of patient 741 had macular pattern dystrophy and clear-cut foveal changes, but his visual acuity was essentially normal in both eyes (see online supplementary figure S2A–C, table S1). Similarly, the mother and the son of patient 1318 were both carriers of the mutation c.637T>C (p.C213R) (figure 4). At 57 years of age, the mother was asymptomatic with 20/20 visual acuity but had a florid butterfly-shaped macular pattern dystrophy and a number of other retinal flecks upon examination (see online supplementary figure S2D). The 7-year-old son had a significant refractive defect whereby visual acuity was reduced due to partial amblyopia. His fundus showed a miniature form of foveal butterfly-shaped macular pattern dystrophy that was consistent with an early stage PRPH2 related phenotype (data not shown). The brother of patient 1318, who was homozygous wild-type for the mutation, had normal visual acuity (20/20) and no maculo-pathy (data not shown). To our knowledge, our study reported for the first time that homozygous mutations in PRPH2 cause EORD with LCA/juvenile RP phenotypes.

Figure 3.

Figure 3

The phenotypes of patients 1318, 3256, and 741 who carry homozygous mutations in PRPH2. (A) Fundus photograph of patient 1318 shows a prominent multilobulated central atrophic maculopathy surrounded by concentric rings of yellow deposits, with vessel narrowing and fine diffuse peripheral retinal changes. (B–D) Fundus photographs of patient 3256 show pigment deposits both peripherally and in the macular region, extensive disease with choroidal sclerosis, vessel narrowing, and optic disc pallor; a central extensive atrophic maculopathy is seen in C and D. (E) Optical coherence tomography images of patient 3256 confirm the extensive maculopathy and unusual globular lesions in the foveal region. (F) Fundus photograph of patient 741 shows the obvious diffuse retinal dystrophy with retinal vessel narrowing, retinal pigment epithelium mottling and loss, and a multilobulated maculopathy. (G) Fundus autofluorescence of patient 741 shows diffuse retinal abnormalities, perifoveal hyper-autofluorescence and a multilobulated foveal abnormality. (H) Optical coherence tomography images of patient 741 shows inner segment/ outer segment junction confined to the central macula, which explains well the fairly good visual acuity (20/40), and an unusual appearing deposit in the foveal regions. Note the similarity with the maculopathy shown in A, D, F, and G. The photographs of patients 1318, 3256, and 741 were taken at the age of 29, 66, and 30 years, respectively.

Figure 4.

Figure 4

Pathogenic mutations of PRPH2 identified in three patients. (A–C) Pedigree information of three patients and Sanger sequencing results for the two mutations in patients and controls. (A) The c.637T>C mutation is homozygous in patient 1318, heterozygous in both parents and the son, and homozygous wild-type in the brother. (B) The c.554T>C mutation is homozygous in patient 3256. (C) The c.554T>C mutation is homozygous in patient 741 and heterozygous in both parents. (D) Amino acid residues affected by the two missense mutations are conserved across different species. Solid symbols: affected; open symbols: unaffected; squares: male; circles: female; arrow: proband; asterisk: the DNA was not available for both parents of 3256; M1 and M2 represent the two mutations, respectively.

Revision of the initial clinical diagnosis in two patients

After revisiting, two patients were reclassified to retinal diseases that were consistent with their molecular diagnosis (tables 7 and 8, online supplementary table S1). The clinical diagnosis of the first patient 3425 who carried a reported homozygous nonsense mutation in the known Oguchi disease gene SAG was revised to Oguchi disease, which presents as congenital stationary night blindness, fundus discolouration, and slowed dark adaptation.67 The second patient 3494 carried novel compound heterozygous nonsense mutations in the Alström syndrome gene ALMS1 (Otable 8). Both mutations segregated with the disease in the family (see online supplementary table S1). Patient 3494 was initially diagnosed with LCA at the age of 8; however, revisiting this patient at the age of 11 revealed other syndromic features including obesity, diabetes mellitus, and learning difficulties (see online supplementary table S1). Furthermore, the fundus examination showed an atrophic bull’s eye-like maculopathy, which was often seen in Alström syndrome patients (see online supplementary figure S3). These results indicate that molecular diagnosis can be a useful tool to revise or correct the initial clinical diagnosis.

The LCA-like or juvenile RP-like presentations in eight patients

Guided by the molecular diagnosis, revisiting the phenotypes of an additional eight patients revealed their ‘LCA-like’ or ‘juvenile RP-like’ phenotypes that may represent spectrums of corresponding retinal diseases (see online supplementary table S1). For example, patient 3688 carried a novel hemizygous splicing site mutation in X-linked RP gene RPGR (table 8). This patient exhibited ‘LCA-like’ phenotypes including nystagmus at birth, which is typically absent in X-linked RP (see online supplementary table S1). However, it is already known that X-linked RP patients may lose central and peripheral vision more rapidly than average RP patients.69 Similarly, two patients (647, 617) carried reported mutations in cone-rod dystrophy gene CERKL and enhanced S-cone syndrome gene NR2E3, respectively (table 7). They exhibited ‘LCA-like’ phenotypes, including congenital visual impairment and nystagmus at birth (see online supplementary table S1). However, based on the available clinical information, the phenotypes of the two patients may also represent severe spectrums of cone-rod dystrophy and S-cone syndrome, respectively. Patient 3311 carried a heterozygous mutation in PRPF31 that is known to cause RP with late onset and mild phenotypes.64 This patient exhibited early onset ‘juvenile RP-like’, possibly due to modifier effect from another gene (see online supplementary table S1, figure S4).

In addition, four patients (704, 1327, 3748, and 3773) carried mutations in BBS1, BBS7, CLN3, and INPP5E, respectively (tables 7 and 9). Mutations in these genes were known to cause syndromes that are characterised by visual impairment and other systemic features.7072 It was also reported in some cases that these genes were associated with ‘LCA-like’ or ‘RP-like’ phenotypes without defects in other organs. 60,62,73 In our study, revisiting these patients confirmed their severe retinal degenerations without other syndromic features (see online supplementary table S1). For example, patient 704 carried a reported homozygous missense mutation in BBS gene BBS1 (table 7). This mutation segregated with disease within the family (see online supplementary table S1). Revisiting this patient at the age of 53 confirmed the ‘juvenile RP-like’ phenotypes without other syndromic features (see online supplementary table S1). However, the retinal features of this patient were consistent with those observed in other BBS patients with BBS1 mutations (see online supplementary figure S5).72 Due to these molecular findings and retinal features, we should still follow up the potential development of syndromic phenotypes in these patients.

Collectively, these results suggest that the clinical manifestations of LCA/juvenile RP and related retinal diseases are overlapped, and that patients with ‘LCA-like’ or ‘juvenile RP-like’ phenotypes may actually carry mutations in non-canonical LCA/ juvenile RP genes.74 Therefore, molecular diagnosis should be used to refine the clinical diagnosis and get a better understanding of the disease. To achieve a more accurate diagnosis for these patients, it is essential to screen for mutations in a larger set of retinal disease genes.

DISCUSSION

In this study, we developed a targeted NGS based method for the molecular diagnosis of LCA and most other retinal diseases. We systematically evaluated this method on a HapMap sample and then applied it to 179 unrelated and prescreened LCA or juvenile RP patients. To our knowledge, our sample set represents the largest cohort of unrelated patients diagnosed with LCA or juvenile RP that is systematically screened for all known LCA genes and most other known retinal disease genes. In-depth analysis of this dataset led to several important findings.

A large number of novel mutations have been identified in our study, representing 54% (45/83) of the identified mutations in this patient cohort (table 2). Our observations are consistent with the 1000 genome project’s finding that every individual’s genome contains a large number of rare variants.25 Compared with common variants that arose earlier during the evolution, these recent rare variants may have greater impact on disease pathogenesis.75 Therefore, we expect that a significant number of novel mutations will continue to be discovered every time a new patient is sequenced. Since NGS based molecular diagnosis can capture novel mutations, it is likely to achieve a high diagnosis rate. Among the 45 novel mutations that we identified, 29 were LOF mutations and 16 were missense mutations. All these novel mutations are likely to be pathogenic. First, these mutations are rare in large control databases. Collectively the databases used in our study contain more than 7400 control individuals. Second, all of these mutations match the reported inheritance pattern of the respective genes. In particular, the pathogenicity of all novel missense mutations reported in our study was supported by five well-established algorithms (see online supplementary table S4). Our study adds a significant number of novel pathogenic mutations to our current knowledge of disease causing mutations. These mutations can serve as references and directly benefit the future molecular diagnosis of patients clinically diagnosed with LCA or juvenile RP.

We identified the genetic defects in 40% of our patient cohort. This lower ratio is primarily due to the fact that our patient cohort had been prescreened. Among the initial cohort of 389 patients, we had previously identified mutations in known LCA genes for 210 patients (see Methods). Among the remaining 179 patients included in this study, mutations in known LCA genes were identified in 56 patients. Therefore, about 68% ((210+56)/389) of our initial cohort can be explained by mutations in known LCA genes. This is concordant with the estimation that mutations in current known LCA genes explain about 70% of LCA cases.11 Among the 56 patients who carry mutations in known LCA genes, 24 patients have prescreening information available (see online supplementary table S5). We found that 16 patients had neither been screened by LCA APEX array nor been Sanger sequenced for the corresponding genes identified in this study. The remaining eight patients had been screened by LCA APEX array and/or Sanger sequencing for the corresponding genes identified in this study. Their mutations had not been identified in the prescreening because the mutations had not been covered by LCA APEX array and/or because Sanger sequencing only covered the frequently mutated exons of related genes.15

To our knowledge, our results demonstrate for the first time that homozygous mutations in PRPH2 cause EORD. The phenotypes of the three patients with homozygous mutations in PRPH2 were severe and quite consistent, especially with regard to the maculopathy phenotypes. By contrast, their family members who carried heterozygous mutations in PRPH2 had milder phenotypes. These results are consistent with the previous observations in PRPH2 mouse models. The rds/rds mouse that carried a homozygous null mutation in PRPH2 failed to develop photoreceptor outer segments and showed early onset and severe retinal degeneration, whereas the heterozygous rds/ +mouse displayed milder retinal degeneration and visual loss, suggesting that dose dependent phenotypic expression is an essential feature in the working of the PRPH2 gene.76,77 Until the discovery of these three patients homozygous for PRPH2 mutations, the full severity of the retinal degeneration seen in the rds/rds mouse had not yet been observed in humans. In our study, individuals with heterozygous mutations in PRPH2 were asymptomatic but had detectable macular flecks upon subsequent examination, exhibiting the clinical presentation of a macular pattern dystrophy, which is fully consistent with previously reported PRPH2 mediated phenotypes.65 By contrast, the severe early onset retinal defects in the three patients with homozygous mutations in PRPH2 are novel and likely due to dose dependent effect.

It may be argued that the rds/rds mouse and our patients harboured different mutations and that individuals with the heterozygous p.L185P mutation in previously reported digenic RP families were originally reported as asymptomatic.78 However, the p.L185P mutation is now known to exert a measurable partial LOF effect. Work from Molday and co-workers established that this peripherin mutant is conditionally defective with respect to subunit assembly, and is capable of forming peripherin dimers but not tetramers.79,80 Furthermore, Kedzierski et al have shown that rds/+mice overexpressing L185P peripherin mutant indeed exhibited a mild phenotype. These mice had outer nuclear layer loss, partially disorganised outer segments, and reduced ERG responses. As observed in our patients homozygous for the p.L185P mutation, rds/rds mice overexpressing L185P peripherin mutant exhibited dramatically reduced levels of peripherin expression in their retinas, and a much more severe histological and electroretinographic retinal phenotype.81 Taken together, these findings suggest that, although asymptomatic, individuals harbouring the heterozygous p.L185P mutation should be expected to exhibit a subclinical phenotype and it is possible that, as in our cases, later in life they may all consistently develop asymptomatic macular flecks or other minor yet measurable phenotypic manifestations.

Interestingly, similar examples have been reported for many other genes.82,83 PITX3 is a gene that is usually mutated in dominant congenital cataracts and anterior segment dysgenesis. However, patients with two mutations in this gene exhibited microphthalmia and central nervous system (CNS) abnormalities.84 For another example, homozygous mutations in the low density lipoprotein (LDL) receptor gene were known to cause much more severe phenotypes of hypercholesterolaemia than heterozygous mutations.85

From a therapeutic standpoint, the implication for patients with retinal degenerations caused by homozygous mutations in PRPH2 is that their diseases can be modelled by the rds/rds mouse, which has been treated by gene augmentation therapy in proof-of-concept research.86,87 There is also a long history of investigation of the severe phenotype in this model, features of which can now be studied in the patients to determine how representative the model is in relation to the newly identified human condition.

There are two main explanations for the 60% of our patients for whom we were unable to find pathogenic mutations in this study. First, mutations that were not covered by our method, including intronic mutations, synonymous mutations, large structural variations, and copy number variations, may account for diseases in these patients. Second, these unsolved cases may due to novel disease-causing genes. Indeed, whole exome sequencing (WES) of some of these unsolved cases has led to the identification of a novel LCA gene NMNAT16. Therefore, we expect that additional novel disease-causing genes will be identified by performing WES on these unsolved cases.

Our results highlight the utility of molecular information in diagnosing clinically heterogeneous diseases. Assigning clinical diagnosis at the time of initial visit is difficult in some cases, and molecular diagnosis can guide the health care provider to reassess the phenotypes of their patients and achieve a more accurate diagnosis. Indeed, guided by their molecular diagnosis, two patients in our study were reclassified with other retinal diseases. Additionally, the clinical manifestations of different retinal diseases are sometimes overlapped, and molecular diagnosis can help us to better define the disease. In our report, eight patients exhibited ‘LCA-like’ or ‘juvenile RP-like’ presentations. Based on the available clinical information, the diagnosis of these patients may be either LCA/juvenile RP, or extreme spectrums of other related retinal diseases, due to the allelic differences or genetic background. Despite the phenotypic similarity between different clinical diagnoses, diseases can be well defined by the molecular diagnosis. Therefore, with the rapid drop of sequencing costs, comprehensive mutation screening that covers all known retinal disease genes should become an integral part of diagnosis in the near future.

In addition to aiding the diagnosis, molecular information can directly contribute to better patient management. Recently, studies on gene therapy for LCA have made significant progress.8891 An accurate molecular diagnosis is the first step toward realising the promise of gene therapy. Additionally, it can clarify the prognosis and change the focus of the clinical follow-up. Patients with different molecularly defined diseases may receive a different prognosis and clinical interventions. For example, patients who exhibit LCA phenotypes but carry mutations in syndromic retinal disease genes should be followed for the development of syndromic features and be given corresponding clinical management. Finally, it can facilitate the genetic counselling and decision-making. Carrier tests or predictive tests for retinal diseases can inform prospective parents of their reproductive risk and possibly influence their decisions.

The low coverage regions in our design either had a higher GC content or were within duplicate regions (see online supplementary tables S2 and S3). Indeed, the GC content bias of coverage in Illumina sequencing data has been previously reported and the bias could be potentially introduced in many steps during sequencing.9294 It was recently recognised that PCR amplification before sequencing may be the major source of GC content bias; protocols to minimise such bias were proposed accordingly.95,96 In addition, low coverage in duplicated regions is likely due to the inability to map reads to a single unique position. The relatively shorts reads (90∼300 bp) generated by most currently available NGS platforms lack enough sequence specificity to be mapped to a single location among multiple duplicated regions. To uncover the genomic information of duplicated regions, long range PCR or NGS sequencer producing longer reads may be utilised.

In summary, we were able to identify pathogenic mutations for 40% of this prescreened patient cohort. A total of 45 novel pathogenic mutations were found. Interestingly, we found that homozygous mutations in PRPH2 can cause LCA and juvenile RP. Our study highlighted the utility of comprehensive molecular information as an integral part of the diagnosis process to achieve more accurate diagnosis and potentially better disease treatment and management.

Supplementary Material

supplementFigure
supplementlist1
supplementlist2
supplementtable

Acknowledgements

We sincerely thank all the patients and their families for their participation. We thank Ms Shirley Briand, Ms Alcira Vieiri and Ms Renee Pigeon for coordinating the blindness clinics at the McGill Ocular Genetics Laboratory. XW is supported by predoctoral fellowship: The Burroughs Wellcome Fund, The Houston Laboratory and Population Sciences Training Program in Gene Environment Interaction. HW is supported by NIH postdoctoral fellowship 5F32EY19430. JEZ is supported by NIH training grant T32 EY007102. RKK is supported by the Foundation Fighting Blindness Canada, the Canadian Institutes for Health Research, FRSQ, the Foundation for Retinal Research, and Reseau Vision. AI is supported by grant from Research to Prevent Blindness, Inc, New York, NY (unrestricted grant to UTHSC Department of Ophthalmology and a Physician Scientist Award to AI). This work is supported by grants from the Retinal Research Foundation and the National Eye Institute (R01EY018571 and R01EY020540) to RC.

Footnotes

Contributors RC, XW and RKK designed the study. SGJ, AI, DB, JRH, GAF, EIT and DW performed the clinical study. XW, HW, VS, HFT, VK, KW, HR, IL, JEZ, SS, SB, AK, JS, FW and YL performed the molecular study. XW integrated the data and performed the analysis. XW, HFT, RC, RKK, JEZ, SGJ, AI wrote the manuscript.

Competing interests None.

Provenance and peer review Not commissioned; externally peer reviewed.

REFERENCES

  • 1.Franceschetti A, Dieterle P. [Diagnostic and prognostic importance of the electroretinogram in tapetoretinal degeneration with reduction of the visual field and hemeralopia] Confin Neurol. 1954;14:184–186. [PubMed] [Google Scholar]
  • 2.Leber T. Ueber Retinitis pigmentosa und angeborene Amaurose. Graefes Arch Clin Exp Ophthalmol. 1869;15:1–25. [Google Scholar]
  • 3.Koenekoop RK. An overview of Leber congenital amaurosis: a model to understand human retinal development. Surv Ophthalmol. 2004;49:379–398. doi: 10.1016/j.survophthal.2004.04.003. [DOI] [PubMed] [Google Scholar]
  • 4.Stone EM. Leber congenital amaurosis—a model for efficient genetic testing of heterogeneous disorders: LXIV Edward Jackson Memorial Lecture. Am J Ophthalmol. 2007;144:791–811. doi: 10.1016/j.ajo.2007.08.022. [DOI] [PubMed] [Google Scholar]
  • 5.Estrada-Cuzcano A, Koenekoop RK, Coppieters F, Kohl S, Lopez I, Collin RW, De Baere EB, Roeleveld D, Marek J, Bernd A, Rohrschneider K, van den Born LI, Meire F, Maumenee IH, Jacobson SG, Hoyng CB, Zrenner E, Cremers FP, den Hollander AI. IQCB1 mutations in patients with Leber congenital amaurosis. Invest Ophthalmol Vis Sci. 2011;52:834–839. doi: 10.1167/iovs.10-5221. [DOI] [PubMed] [Google Scholar]
  • 6.Koenekoop RK, Wang H, Majewski J, Wang X, Lopez I, Ren H, Chen Y, Li Y, Fishman GA, Genead M, Schwartzentruber J, Solanki N, Traboulsi EI, Cheng J, Logan CV, McKibbin M, Hayward BE, Parry DA, Johnson CA, Nageeb M, Poulter JA, Mohamed MD, Jafri H, Rashid Y, Taylor GR, Keser V, Mardon G, Xu H, Inglehearn CF, Fu Q, Toomes C, Chen R. Mutations in NMNAT1 cause Leber congenital amaurosis and identify a new disease pathway for retinal degeneration. Nat Genet. 2012;44:1035–1039. doi: 10.1038/ng.2356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Falk MJ, Zhang Q, Nakamaru-Ogiso E, Kannabiran C, Fonseca-Kelly Z, Chakarova C, Audo I, Mackay DS, Zeitz C, Borman AD, Staniszewska M, Shukla R, Palavalli L, Mohand-Said S, Waseem NH, Jalali S, Perin JC, Place E, Ostrovsky J, Xiao R, Bhattacharya SS, Consugar M, Webster AR, Sahel JA, Moore AT, Berson EL, Liu Q, Gai X, Pierce EA. NMNAT1 mutations cause Leber congenital amaurosis. Nat Genet. 2012;44:1040–1045. doi: 10.1038/ng.2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Perrault I, Hanein S, Zanlonghi X, Serre V, Nicouleau M, Defoort-Delhemmes S, Delphin N, Fares-Taie L, Gerber S, Xerri O, Edelson C, Goldenberg A, Duncombe A, Le Meur G, Hamel C, Silva E, Nitschke P, Calvas P, Munnich A, Roche O, Dollfus H, Kaplan J, Rozet JM. Mutations in NMNAT1 cause Leber congenital amaurosis with early-onset severe macular and optic atrophy. Nat Genet. 2012;44:975–977. doi: 10.1038/ng.2357. [DOI] [PubMed] [Google Scholar]
  • 9.Wang H, den Hollander AI, Moayedi Y, Abulimiti A, Li Y, Collin RW, Hoyng CB, Lopez I, Abboud EB, Al-Rajhi AA, Bray M, Lewis RA, Lupski JR, Mardon G, Koenekoop RK, Chen R. Mutations in SPATA7 cause Leber congenital amaurosis and juvenile retinitis pigmentosa. Am J Hum Genet. 2009;84:380–387. doi: 10.1016/j.ajhg.2009.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sergouniotis PI, Davidson AE, Mackay DS, Li Z, Yang X, Plagnol V, Moore AT, Webster AR. Recessive mutations in KCNJ13, encoding an inwardly rectifying potassium channel subunit, cause Leber congenital amaurosis. Am J Hum Genet. 2011;89:183–190. doi: 10.1016/j.ajhg.2011.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.den Hollander AI, Roepman R, Koenekoop RK, Cremers FP. Leber congenital amaurosis: genes, proteins and disease mechanisms. Prog Retin Eye Res. 2008;27:391–419. doi: 10.1016/j.preteyeres.2008.05.003. [DOI] [PubMed] [Google Scholar]
  • 12.Chiang PW, Wang J, Chen Y, Fu Q, Zhong J, Yi X, Wu R, Gan H, Shi Y, Barnett C, Wheaton D, Day M, Sutherland J, Heon E, Weleber RG, Gabriel LA, Cong P, Chuang K, Ye S, Sallum JM, Qi M. Exome sequencing identifies NMNAT1 mutations as a cause of Leber congenital amaurosis. Nat Genet. 2012;44:972–974. doi: 10.1038/ng.2370. [DOI] [PubMed] [Google Scholar]
  • 13.den Hollander AI, Lopez I, Yzer S, Zonneveld MN, Janssen IM, Strom TM, Hehir-Kwa JY, Veltman JA, Arends ML, Meitinger T, Musarella MA, van den Born LI, Fishman GA, Maumenee IH, Rohrschneider K, Cremers FP, Koenekoop RK. Identification of novel mutations in patients with Leber congenital amaurosis and juvenile RP by genome-wide homozygosity mapping with SNP microarrays. Invest Ophthalmol Vis Sci. 2007;48:5690–5698. doi: 10.1167/iovs.07-0610. [DOI] [PubMed] [Google Scholar]
  • 14.Yzer S, Leroy BP, De Baere E, de Ravel TJ, Zonneveld MN, Voesenek K, Kellner U, Ciriano JP, de Faber JT, Rohrschneider K, Roepman R, den Hollander AI, Cruysberg JR, Meire F, Casteels I, van Moll-Ramirez NG, Allikmets R, van den Born LI, Cremers FP. Microarray-based mutation detection and phenotypic characterization of patients with Leber congenital amaurosis. Invest Ophthalmol Vis Sci. 2006;47:1167–1176. doi: 10.1167/iovs.05-0848. [DOI] [PubMed] [Google Scholar]
  • 15.Zernant J, Kulm M, Dharmaraj S, den Hollander AI, Perrault I, Preising MN, Lorenz B, Kaplan J, Cremers FP, Maumenee I, Koenekoop RK, Allikmets R. Genotyping microarray (disease chip) for Leber congenital amaurosis: detection of modifier alleles. Invest Ophthalmol Vis Sci. 2005;46:3052–3059. doi: 10.1167/iovs.05-0111. [DOI] [PubMed] [Google Scholar]
  • 16.Henderson RH, Waseem N, Searle R, van der Spuy J, Russell-Eggitt I, Bhattacharya SS, Thompson DA, Holder GE, Cheetham ME, Webster AR, Moore AT. An assessment of the apex microarray technology in genotyping patients with Leber congenital amaurosis and early-onset severe retinal dystrophy. Invest Ophthalmol Vis Sci. 2007;48:5684–5689. doi: 10.1167/iovs.07-0207. [DOI] [PubMed] [Google Scholar]
  • 17.Shanks ME, Downes SM, Copley RR, Lise S, Broxholme J, Hudspith KA, Kwasniewska A, Davies WI, Hankins MW, Packham ER, Clouston P, Seller A, Wilkie AO, Taylor JC, Ragoussis J, Nemeth AH. Next-generation sequencing (NGS) as a diagnostic tool for retinal degeneration reveals a much higher detection rate in early-onset disease. Eur J Hum Genet. 2012 doi: 10.1038/ejhg.2012.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Neveling K, Collin RW, Gilissen C, van Huet RA, Visser L, Kwint MP, Gijsen SJ, Zonneveld MN, Wieskamp N, de Ligt J, Siemiatkowska AM, Hoefsloot LH, Buckley MF, Kellner U, Branham KE, den Hollander AI, Hoischen A, Hoyng C, Klevering BJ, van den Born LI, Veltman JA, Cremers FP, Scheffer H. Next-generation genetic testing for retinitis pigmentosa. Hum Mutat. 2012;33:963–972. doi: 10.1002/humu.22045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Coppieters F, De Wilde B, Lefever S, De Meester E, De Rocker N, Van Cauwenbergh C, Pattyn F, Meire F, Leroy BP, Hellemans J, Vandesompele J, De Baere E. Massively parallel sequencing for early molecular diagnosis in Leber congenital amaurosis. Genet Med. 2012;14:576–585. doi: 10.1038/gim.2011.51. [DOI] [PubMed] [Google Scholar]
  • 20.Galvin JA, Fishman GA, Stone EM, Koenekoop RK. Evaluation of genotype-phenotype associations in leber congenital amaurosis. Retina. 2005;25:919–929. doi: 10.1097/00006982-200510000-00016. [DOI] [PubMed] [Google Scholar]
  • 21.Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M, DePristo MA. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303. doi: 10.1101/gr.107524.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Challis D, Yu J, Evani US, Jackson AR, Paithankar S, Coarfa C, Milosavljevic A, Gibbs RA, Yu F. An integrative variant analysis suite for whole exome next-generation sequencing data. BMC Bioinformatics. 2012;13:8. doi: 10.1186/1471-2105-13-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sherry ST, Ward MH, Kholodov M, Baker J, Phan L, Smigielski EM, Sirotkin K. dbSNP: the NCBI database of genetic variation. Nucleic Acids Res. 2001;29:308–311. doi: 10.1093/nar/29.1.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.1000 Genomes Project Consortium. A map of human genome variation from population-scale sequencing. Nature. 2010;467:1061–1073. doi: 10.1038/nature09534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164. doi: 10.1093/nar/gkq603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Liu X, Jian X, Boerwinkle E. dbNSFP: a lightweight database of human nonsynonymous SNPs and their functional predictions. Hum Mutat. 2011;32:894–899. doi: 10.1002/humu.21517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kumar P, Henikoff S, Ng PC. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4:1073–1081. doi: 10.1038/nprot.2009.86. [DOI] [PubMed] [Google Scholar]
  • 29.Pollard KS, Hubisz MJ, Rosenbloom KR, Siepel A. Detection of nonneutral substitution rates on mammalian phylogenies. Genome Res. 2010;20:110–121. doi: 10.1101/gr.097857.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chun S, Fay JC. Identification of deleterious mutations within three human genomes. Genome Res. 2009;19:1553–1561. doi: 10.1101/gr.092619.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS, Sunyaev SR. A method and server for predicting damaging missense mutations. Nat Methods. 2010;7:248–249. doi: 10.1038/nmeth0410-248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Schwarz JM, Rodelsperger C, Schuelke M, Seelow D. MutationTaster evaluates disease-causing potential of sequence alterations. Nat Methods. 2010;7:575–576. doi: 10.1038/nmeth0810-575. [DOI] [PubMed] [Google Scholar]
  • 33.The International HapMap Consortium. The International HapMap Project. Nature. 2003;426:789–796. doi: 10.1038/nature02168. [DOI] [PubMed] [Google Scholar]
  • 34.Stenson PD, Mort M, Ball EV, Howells K, Phillips AD, Thomas NS, Cooper DN. The Human Gene Mutation Database: 2008 update. Genome Med. 2009;1:13. doi: 10.1186/gm13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Richards CS, Bale S, Bellissimo DB, Das S, Grody WW, Hegde MR, Lyon E, Ward BE. ACMG recommendations for standards for interpretation and reporting of sequence variations: revisions 2007. Genet Med. 2008;10:294–300. doi: 10.1097/GIM.0b013e31816b5cae. [DOI] [PubMed] [Google Scholar]
  • 36.Thompson DA, Janecke AR, Lange J, Feathers KL, Hubner CA, McHenry CL, Stockton DW, Rammesmayer G, Lupski JR, Antinolo G, Ayuso C, Baiget M, Gouras P, Heckenlively JR, den Hollander A, Jacobson SG, Lewis RA, Sieving PA, Wissinger B, Yzer S, Zrenner E, Utermann G, Gal A. Retinal degeneration associated with RDH12 mutations results from decreased 11-cis retinal synthesis due to disruption of the visual cycle. Hum Mol Genet. 2005;14:3865–3875. doi: 10.1093/hmg/ddi411. [DOI] [PubMed] [Google Scholar]
  • 37.Janecke AR, Thompson DA, Utermann G, Becker C, Hubner CA, Schmid E, McHenry CL, Nair AR, Ruschendorf F, Heckenlively J, Wissinger B, Nurnberg P, Gal A. Mutations in RDH12 encoding a photoreceptor cell retinol dehydrogenase cause childhood-onset severe retinal dystrophy. Nat Genet. 2004;36:850–854. doi: 10.1038/ng1394. [DOI] [PubMed] [Google Scholar]
  • 38.Sohocki MM, Bowne SJ, Sullivan LS, Blackshaw S, Cepko CL, Payne AM, Bhattacharya SS, Khaliq S, Qasim Mehdi S, Birch DG, Harrison WR, Elder FF, Heckenlively JR, Daiger SP. Mutations in a new photoreceptor-pineal gene on 17p cause Leber congenital amaurosis. Nat Genet. 2000;24:79–83. doi: 10.1038/71732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sohocki MM, Perrault I, Leroy BP, Payne AM, Dharmaraj S, Bhattacharya SS, Kaplan J, Maumenee IH, Koenekoop R, Meire FM, Birch DG, Heckenlively JR, Daiger SP. Prevalence of AIPL1 mutations in inherited retinal degenerative disease. Mol Genet Metab. 2000;70:142–150. doi: 10.1006/mgme.2000.3001. [DOI] [PubMed] [Google Scholar]
  • 40.den Hollander AI, Koenekoop RK, Yzer S, Lopez I, Arends ML, Voesenek KE, Zonneveld MN, Strom TM, Meitinger T, Brunner HG, Hoyng CB, van den Born LI, Rohrschneider K, Cremers FP. Mutations in the CEP290 (NPHP6) gene are a frequent cause of Leber congenital amaurosis. Am J Hum Genet. 2006;79:556–561. doi: 10.1086/507318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Sayer JA, Otto EA, O’Toole JF, Nurnberg G, Kennedy MA, Becker C, Hennies HC, Helou J, Attanasio M, Fausett BV, Utsch B, Khanna H, Liu Y, Drummond I, Kawakami I, Kusakabe T, Tsuda M, Ma L, Lee H, Larson RG, Allen SJ, Wilkinson CJ, Nigg EA, Shou C, Lillo C, Williams DS, Hoppe B, Kemper MJ, Neuhaus T, Parisi MA, Glass IA, Petry M, Kispert A, Gloy J, Ganner A, Walz G, Zhu X, Goldman D, Nurnberg P, Swaroop A, Leroux MR, Hildebrandt F. The centrosomal protein nephrocystin-6 is mutated in Joubert syndrome and activates transcription factor ATF4. Nat Genet. 2006;38:674–681. doi: 10.1038/ng1786. [DOI] [PubMed] [Google Scholar]
  • 42.Perrault I, Delphin N, Hanein S, Gerber S, Dufier JL, Roche O, Defoort-Dhellemmes S, Dollfus H, Fazzi E, Munnich A, Kaplan J, Rozet JM. Spectrum of NPHP6/CEP290 mutations in Leber congenital amaurosis and delineation of the associated phenotype. Hum Mutat. 2007;28:416. doi: 10.1002/humu.9485. [DOI] [PubMed] [Google Scholar]
  • 43.Lotery AJ, Jacobson SG, Fishman GA, Weleber RG, Fulton AB, Namperumalsamy P, Heon E, Levin AV, Grover S, Rosenow JR, Kopp KK, Sheffield VC, Stone EM. Mutations in the CRB1 gene cause Leber congenital amaurosis. Arch Ophthalmol. 2001;119:415–420. doi: 10.1001/archopht.119.3.415. [DOI] [PubMed] [Google Scholar]
  • 44.Perrault I, Rozet JM, Gerber S, Ghazi I, Ducroq D, Souied E, Leowski C, Bonnemaison M, Dufier JL, Munnich A, Kaplan J. Spectrum of retGC1 mutations in Leber’s congenital amaurosis. Eur J Hum Genet. 2000;8:578–582. doi: 10.1038/sj.ejhg.5200503. [DOI] [PubMed] [Google Scholar]
  • 45.Coppieters F, Casteels I, Meire F, De Jaegere S, Hooghe S, van Regemorter N, Van Esch H, Matuleviciene A, Nunes L, Meersschaut V, Walraedt S, Standaert L, Coucke P, Hoeben H, Kroes HY, Vande Walle J, de Ravel T, Leroy BP, De Baere E. Genetic screening of LCA in Belgium: predominance of CEP290 and identification of potential modifier alleles in AHI1 of CEP290-related phenotypes. Hum Mutat. 2010;31:E1709–E66. doi: 10.1002/humu.21336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lotery AJ, Namperumalsamy P, Jacobson SG, Weleber RG, Fishman GA, Musarella MA, Hoyt CS, Heon E, Levin A, Jan J, Lam B, Carr RE, Franklin A, Radha S, Andorf JL, Sheffield VC, Stone EM. Mutation analysis of 3 genes in patients with Leber congenital amaurosis. Arch Ophthalmol. 2000;118:538–543. doi: 10.1001/archopht.118.4.538. [DOI] [PubMed] [Google Scholar]
  • 47.den Hollander AI, Koenekoop RK, Mohamed MD, Arts HH, Boldt K, Towns KV, Sedmak T, Beer M, Nagel-Wolfrum K, McKibbin M, Dharmaraj S, Lopez I, Ivings L, Williams GA, Springell K, Woods CG, Jafri H, Rashid Y, Strom TM, van der Zwaag B, Gosens I, Kersten FF, van Wijk E, Veltman JA, Zonneveld MN, van Beersum SE, Maumenee IH, Wolfrum U, Cheetham ME, Ueffing M, Cremers FP, Inglehearn CF, Roepman R. Mutations in LCA5, encoding the ciliary protein lebercilin, cause Leber congenital amaurosis. Nat Genet. 2007;39:889–895. doi: 10.1038/ng2066. [DOI] [PubMed] [Google Scholar]
  • 48.Senechal A, Humbert G, Surget MO, Bazalgette C, Arnaud B, Arndt C, Laurent E, Brabet P, Hamel CP. Screening genes of the retinoid metabolism: novel LRAT mutation in leber congenital amaurosis. Am J Ophthalmol. 2006;142:702–704. doi: 10.1016/j.ajo.2006.04.057. [DOI] [PubMed] [Google Scholar]
  • 49.Morimura H, Fishman GA, Grover SA, Fulton AB, Berson EL, Dryja TP. Mutations in the RPE65 gene in patients with autosomal recessive retinitis pigmentosa or leber congenital amaurosis. Proc Natl Acad Sci U S A. 1998;95:3088–3093. doi: 10.1073/pnas.95.6.3088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li Y, Wang H, Peng J, Gibbs RA, Lewis RA, Lupski JR, Mardon G, Chen R. Mutation survey of known LCA genes and loci in the Saudi Arabian population. Invest Ophthalmol Vis Sci. 2009;50:1336–1343. doi: 10.1167/iovs.08-2589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Thompson DA, Li Y, McHenry CL, Carlson TJ, Ding X, Sieving PA, Apfelstedt-Sylla E, Gal A. Mutations in the gene encoding lecithin retinol acyltransferase are associated with early-onset severe retinal dystrophy. Nat Genet. 2001;28:123–124. doi: 10.1038/88828. [DOI] [PubMed] [Google Scholar]
  • 52.Clark GR, Crowe P, Muszynska D, O’Prey D, O’Neill J, Alexander S, Willoughby CE, McKay GJ, Silvestri G, Simpson DA. Development of a diagnostic genetic test for simplex and autosomal recessive retinitis pigmentosa. Ophthalmology. 2010;117:2169–2177. doi: 10.1016/j.ophtha.2010.02.029. e3. [DOI] [PubMed] [Google Scholar]
  • 53.Collin RW, van den Born LI, Klevering BJ, de Castro-Miro M, Littink KW, Arimadyo K, Azam M, Yazar V, Zonneveld MN, Paun CC, Siemiatkowska AM, Strom TM, Hehir-Kwa JY, Kroes HY, de Faber JT, van Schooneveld MJ, Heckenlively JR, Hoyng CB, den Hollander AI, Cremers FP. High-resolution homozygosity mapping is a powerful tool to detect novel mutations causative of autosomal recessive RP in the Dutch population. Invest Ophthalmol Vis Sci. 2011;52:2227–2239. doi: 10.1167/iovs.10-6185. [DOI] [PubMed] [Google Scholar]
  • 54.Moise AR, Golczak M, Imanishi Y, Palczewski K. Topology and membrane association of lecithin: retinol acyltransferase. J Biol Chem. 2007;282:2081–2090. doi: 10.1074/jbc.M608315200. [DOI] [PubMed] [Google Scholar]
  • 55.Hu J, Ng PC. Predicting the effects of frameshifting indels. Genome Biol. 2012;13:R9. doi: 10.1186/gb-2012-13-2-r9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Henderson RH, Williamson KA, Kennedy JS, Webster AR, Holder GE, Robson AG, FitzPatrick DR, van Heyningen V, Moore AT. A rare de novo nonsense mutation in OTX2 causes early onset retinal dystrophy and pituitary dysfunction. Mol Vis. 2009;15:2442–2447. [PMC free article] [PubMed] [Google Scholar]
  • 57.Otto EA, Loeys B, Khanna H, Hellemans J, Sudbrak R, Fan S, Muerb U, O’Toole JF, Helou J, Attanasio M, Utsch B, Sayer JA, Lillo C, Jimeno D, Coucke P, De Paepe A, Reinhardt R, Klages S, Tsuda M, Kawakami I, Kusakabe T, Omran H, Imm A, Tippens M, Raymond PA, Hill J, Beales P, He S, Kispert A, Margolis B, Williams DS, Swaroop A, Hildebrandt F. Nephrocystin-5, a ciliary IQ domain protein, is mutated in Senior-Loken syndrome and interacts with RPGR and calmodulin. Nat Genet. 2005;37:282–288. doi: 10.1038/ng1520. [DOI] [PubMed] [Google Scholar]
  • 58.Singh HP, Jalali S, Narayanan R, Kannabiran C. Genetic analysis of Indian families with autosomal recessive retinitis pigmentosa by homozygosity screening. Invest Ophthalmol Vis Sci. 2009;50:4065–4071. doi: 10.1167/iovs.09-3479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hanein S, Perrault I, Gerber S, Tanguy G, Barbet F, Ducroq D, Calvas P, Dollfus H, Hamel C, Lopponen T, Munier F, Santos L, Shalev S, Zafeiriou D, Dufier JL, Munnich A, Rozet JM, Kaplan J. Leber congenital amaurosis: comprehensive survey of the genetic heterogeneity, refinement of the clinical definition, and genotype-phenotype correlations as a strategy for molecular diagnosis. Hum Mutat. 2004;23:306–317. doi: 10.1002/humu.20010. [DOI] [PubMed] [Google Scholar]
  • 60.Estrada-Cuzcano A, Koenekoop RK, Senechal A, De Baere EB, de Ravel T, Banfi S, Kohl S, Ayuso C, Sharon D, Hoyng CB, Hamel CP, Leroy BP, Ziviello C, Lopez I, Bazinet A, Wissinger B, Sliesoraityte I, Avila-Fernandez A, Littink KW, Vingolo EM, Signorini S, Banin E, Mizrahi-Meissonnier L, Zrenner E, Kellner U, Collin RW, den Hollander AI, Cremers FP, Klevering BJ. BBS1 mutations in a wide spectrum of phenotypes ranging from nonsyndromic retinitis pigmentosa to Bardet-Biedl syndrome. Arch Ophthalmol. 2012;130:1425–1432. doi: 10.1001/archophthalmol.2012.2434. [DOI] [PubMed] [Google Scholar]
  • 61.Littink KW, Koenekoop RK, van den Born LI, Collin RW, Moruz L, Veltman JA, Roosing S, Zonneveld MN, Omar A, Darvish M, Lopez I, Kroes HY, van Genderen MM, Hoyng CB, Rohrschneider K, van Schooneveld MJ, Cremers FP, den Hollander AI. Homozygosity mapping in patients with cone-rod dystrophy: novel mutations and clinical characterizations. Invest Ophthalmol Vis Sci. 2010;51:5943–5951. doi: 10.1167/iovs.10-5797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sarpong A, Schottmann G, Ruther K, Stoltenburg G, Kohlschutter A, Hubner C, Schuelke M. Protracted course of juvenile ceroid lipofuscinosis associated with a novel CLN3 mutation (p.Y199X) Clin Genet. 2009;76:38–45. doi: 10.1111/j.1399-0004.2009.01179.x. [DOI] [PubMed] [Google Scholar]
  • 63.Haider NB, Jacobson SG, Cideciyan AV, Swiderski R, Streb LM, Searby C, Beck G, Hockey R, Hanna DB, Gorman S, Duhl D, Carmi R, Bennett J, Weleber RG, Fishman GA, Wright AF, Stone EM, Sheffield VC. Mutation of a nuclear receptor gene, NR2E3, causes enhanced S cone syndrome, a disorder of retinal cell fate. Nat Genet. 2000;24:127–131. doi: 10.1038/72777. [DOI] [PubMed] [Google Scholar]
  • 64.Sullivan LS, Bowne SJ, Birch DG, Hughbanks-Wheaton D, Heckenlively JR, Lewis RA, Garcia CA, Ruiz RS, Blanton SH, Northrup H, Gire AI, Seaman R, Duzkale H, Spellicy CJ, Zhu J, Shankar SP, Daiger SP. Prevalence of disease-causing mutations in families with autosomal dominant retinitis pigmentosa: a screen of known genes in 200 families. Invest Ophthalmol Vis Sci. 2006;47:3052–3064. doi: 10.1167/iovs.05-1443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Payne AM, Downes SM, Bessant DA, Bird AC, Bhattacharya SS. Founder effect, seen in the British population, of the 172 peripherin/RDS mutation-and further refinement of genetic positioning of the peripherin/RDS gene. Am J Hum Genet. 1998;62:192–195. doi: 10.1086/301679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kajiwara K, Hahn LB, Mukai S, Travis GH, Berson EL, Dryja TP. Mutations in the human retinal degeneration slow gene in autosomal dominant retinitis pigmentosa. Nature. 1991;354:480–483. doi: 10.1038/354480a0. [DOI] [PubMed] [Google Scholar]
  • 67.Nakamura M, Yamamoto S, Okada M, Ito S, Tano Y, Miyake Y. Novel mutations in the arrestin gene and associated clinical features in Japanese patients with Oguchi’s disease. Ophthalmology. 2004;111:1410–1414. doi: 10.1016/j.ophtha.2003.11.006. [DOI] [PubMed] [Google Scholar]
  • 68.Travis GH, Sutcliffe JG, Bok D. The retinal degeneration slow (rds) gene product is a photoreceptor disc membrane-associated glycoprotein. Neuron. 1991;6:61–70. doi: 10.1016/0896-6273(91)90122-g. [DOI] [PubMed] [Google Scholar]
  • 69.Iannaccone A. The genetics of hereditary retinopathies and optic neuropathies. Compr Ophthalmol Update. 2005;6:39–62. [Google Scholar]
  • 70.Badano JL, Ansley SJ, Leitch CC, Lewis RA, Lupski JR, Katsanis N. Identification of a novel Bardet-Biedl syndrome protein, BBS7, that shares structural features with BBS1 and BBS2. Am J Hum Genet. 2003;72:650–658. doi: 10.1086/368204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.The International Batten Disease Consortium. Isolation of a novel gene underlying Batten disease, CLN3. Cell. 1995;82:949–957. doi: 10.1016/0092-8674(95)90274-0. [DOI] [PubMed] [Google Scholar]
  • 72.Bielas SL, Silhavy JL, Brancati F, Kisseleva MV, Al-Gazali L, Sztriha L, Bayoumi RA, Zaki MS, Abdel-Aleem A, Rosti RO, Kayserili H, Swistun D, Scott LC, Bertini E, Boltshauser E, Fazzi E, Travaglini L, Field SJ, Gayral S, Jacoby M, Schurmans S, Dallapiccola B, Majerus PW, Valente EM, Gleeson JG. Mutations in INPP5E, encoding inositol polyphosphate-5-phosphatase E, link phosphatidyl inositol signaling to the ciliopathies. Nat Genet. 2009;41:1032–1036. doi: 10.1038/ng.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Cox KF, Kerr NC, Kedrov M, Nishimura D, Jennings BJ, Stone EM, Sheffield VC, Iannaccone A. Phenotypic expression of Bardet-Biedl syndrome in patients homozygous for the common M390R mutation in the BBS1 gene. Vision Res. 2012;75:77–87. doi: 10.1016/j.visres.2012.08.005. [DOI] [PubMed] [Google Scholar]
  • 74.Wang X, Wang H, Cao M, Li Z, Chen X, Patenia C, Gore A, Abboud EB, Al-Rajhi AA, Lewis RA, Lupski JR, Mardon G, Zhang K, Muzny D, Gibbs RA, Chen R. Whole-exome sequencing identifies ALMS1, IQCB1, CNGA3, and MYO7A mutations in patients with Leber congenital amaurosis. Hum Mutat. 2011;32:1450–1459. doi: 10.1002/humu.21587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lupski JR, Belmont JW, Boerwinkle E, Gibbs RA. Clan genomics and the complex architecture of human disease. Cell. 2011;147:32–43. doi: 10.1016/j.cell.2011.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.van Nie R, Ivanyi D, Demant P. A new H-2-linked mutation, rds, causing retinal degeneration in the mouse. Tissue Antigens. 1978;12:106–108. doi: 10.1111/j.1399-0039.1978.tb01305.x. [DOI] [PubMed] [Google Scholar]
  • 77.Hawkins RK, Jansen HG, Sanyal S. Development and degeneration of retina in rds mutant mice: photoreceptor abnormalities in the heterozygotes. Exp Eye Res. 1985;41:701–720. doi: 10.1016/0014-4835(85)90179-4. [DOI] [PubMed] [Google Scholar]
  • 78.Kajiwara K, Berson EL, Dryja TP. Digenic retinitis pigmentosa due to mutations at the unlinked peripherin/RDS and ROM1 loci. Science. 1994;264:1604–1608. doi: 10.1126/science.8202715. [DOI] [PubMed] [Google Scholar]
  • 79.Goldberg AF, Molday RS. Defective subunit assembly underlies a digenic form of retinitis pigmentosa linked to mutations in peripherin/rds and rom-1. Proc Natl Acad Sci U S A. 1996;93:13726–13730. doi: 10.1073/pnas.93.24.13726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Loewen CJ, Moritz OL, Molday RS. Molecular characterization of peripherin-2 and rom-1 mutants responsible for digenic retinitis pigmentosa. J Biol Chem. 2001;276:22388–22396. doi: 10.1074/jbc.M011710200. [DOI] [PubMed] [Google Scholar]
  • 81.Kedzierski W, Nusinowitz S, Birch D, Clarke G, McInnes RR, Bok D, Travis GH. Deficiency of rds/peripherin causes photoreceptor death in mouse models of digenic and dominant retinitis pigmentosa. Proc Natl Acad Sci USA. 2001;98:7718–7723. doi: 10.1073/pnas.141124198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Wittstrom E, Ekvall S, Schatz P, Bondeson ML, Ponjavic V, Andreasson S. Morphological and functional changes in multifocal vitelliform retinopathy and biallelic mutations in BEST1. Ophthalmic Genet. 2011;32:83–96. doi: 10.3109/13816810.2010.535890. [DOI] [PubMed] [Google Scholar]
  • 83.Burgess R, Millar ID, Leroy BP, Urquhart JE, Fearon IM, De Baere E, Brown PD, Robson AG, Wright GA, Kestelyn P, Holder GE, Webster AR, Manson FD, Black GC. Biallelic mutation of BEST1 causes a distinct retinopathy in humans. Am J Hum Genet. 2008;82:19–31. doi: 10.1016/j.ajhg.2007.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Bidinost C, Matsumoto M, Chung D, Salem N, Zhang K, Stockton DW, Khoury A, Megarbane A, Bejjani BA, Traboulsi EI. Heterozygous and homozygous mutations in PITX3 in a large Lebanese family with posterior polar cataracts and neurodevelopmental abnormalities. Invest Ophthalmol Vis Sci. 2006;47:1274–1280. doi: 10.1167/iovs.05-1095. [DOI] [PubMed] [Google Scholar]
  • 85.Naoumova RP, Thompson GR, Soutar AK. Current management of severe homozygous hypercholesterolaemias. Curr Opin Lipidol. 2004;15:413–422. doi: 10.1097/01.mol.0000137222.23784.2a. [DOI] [PubMed] [Google Scholar]
  • 86.Ali RR, Sarra GM, Stephens C, Alwis MD, Bainbridge JW, Munro PM, Fauser S, Reichel MB, Kinnon C, Hunt DM, Bhattacharya SS, Thrasher AJ. Restoration of photoreceptor ultrastructure and function in retinal degeneration slow mice by gene therapy. Nat Genet. 2000;25:306–310. doi: 10.1038/77068. [DOI] [PubMed] [Google Scholar]
  • 87.Schlichtenbrede FC, da Cruz L, Stephens C, Smith AJ, Georgiadis A, Thrasher AJ, Bainbridge JW, Seeliger MW, Ali RR. Long-term evaluation of retinal function in Prph2Rd2/Rd2 mice following AAV-mediated gene replacement therapy. J Gene Med. 2003;5:757–764. doi: 10.1002/jgm.401. [DOI] [PubMed] [Google Scholar]
  • 88.Bennett J, Ashtari M, Wellman J, Marshall KA, Cyckowski LL, Chung DC, McCague S, Pierce EA, Chen Y, Bennicelli JL, Zhu X, Ying GS, Sun J, Wright JF, Auricchio A, Simonelli F, Shindler KS, Mingozzi F, High KA, Maguire AM. AAV2 gene therapy readministration in three adults with congenital blindness. Sci Transl Med. 2012;4 doi: 10.1126/scitranslmed.3002865. 120ra15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Maguire AM, Simonelli F, Pierce EA, Pugh EN, Jr., Mingozzi F, Bennicelli J, Banfi S, Marshall KA, Testa F, Surace EM, Rossi S, Lyubarsky A, Arruda VR, Konkle B, Stone E, Sun J, Jacobs J, Dell’Osso L, Hertle R, Ma JX, Redmond TM, Zhu X, Hauck B, Zelenaia O, Shindler KS, Maguire MG, Wright JF, Volpe NJ, McDonnell JW, Auricchio A, High KA, Bennett J. Safety and efficacy of gene transfer for Leber’s congenital amaurosis. N Engl J Med. 2008;358:2240–2248. doi: 10.1056/NEJMoa0802315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K, Viswanathan A, Holder GE, Stockman A, Tyler N, Petersen-Jones S, Bhattacharya SS, Thrasher AJ, Fitzke FW, Carter BJ, Rubin GS, Moore AT, Ali RR. Effect of gene therapy on visual function in Leber’s congenital amaurosis. N Engl J Med. 2008;358:2231–2239. doi: 10.1056/NEJMoa0802268. [DOI] [PubMed] [Google Scholar]
  • 91.Jacobson SG, Cideciyan AV, Ratnakaram R, Heon E, Schwartz SB, Roman AJ, Peden MC, Aleman TS, Boye SL, Sumaroka A, Conlon TJ, Calcedo R, Pang JJ, Erger KE, Olivares MB, Mullins CL, Swider M, Kaushal S, Feuer WJ, Iannaccone A, Fishman GA, Stone EM, Byrne BJ, Hauswirth WW. Gene therapy for Leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years. Arch Ophthalmol. 2011;130:9–24. doi: 10.1001/archophthalmol.2011.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hansen KD, Brenner SE, Dudoit S. Biases in Illumina transcriptome sequencing caused by random hexamer priming. Nucleic Acids Res. 2010;38:e131. doi: 10.1093/nar/gkq224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Quail MA, Kozarewa I, Smith F, Scally A, Stephens PJ, Durbin R, Swerdlow H, Turner DJ. A large genome center’s improvements to the Illumina sequencing system. Nat Methods. 2008;5:1005–1010. doi: 10.1038/nmeth.1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Dohm JC, Lottaz C, Borodina T, Himmelbauer H. Substantial biases in ultra-short read data sets from high-throughput DNA sequencing. Nucleic Acids Res. 2008;36:e105. doi: 10.1093/nar/gkn425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kozarewa I, Ning Z, Quail MA, Sanders MJ, Berriman M, Turner DJ. Amplification-free Illumina sequencing-library preparation facilitates improved mapping and assembly of (G+C)-biased genomes. Nat Methods. 2009;6:291–295. doi: 10.1038/nmeth.1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Aird D, Ross MG, Chen WS, Danielsson M, Fennell T, Russ C, Jaffe DB, Nusbaum C, Gnirke A. Analyzing and minimizing PCR amplification bias in Illumina sequencing libraries. Genome Biol. 2011;12:R18. doi: 10.1186/gb-2011-12-2-r18. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

supplementFigure
supplementlist1
supplementlist2
supplementtable

RESOURCES