Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Human Molecular Genetics, 2016, Vol. 25, No.

8 1637–1647

doi: 10.1093/hmg/ddw041
Advance Access Publication Date: 11 February 2016
Original Article

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


ORIGINAL ARTICLE

Haploinsufficiency of RCBTB1 is associated with Coats


disease and familial exudative vitreoretinopathy
Jeng-Hung Wu1,†, Jorn-Hon Liu4,†, Yu-Chieh Ko3,5, Chi-Tang Wang1, Yu-Chien
Chung5, Kuo-Chang Chu7, Tze-Tze Liu2, Hsiao-Ming Chao4, Yun-Jin Jiang7, *,
Shih-Jen Chen5, * and Ming-Yi Chung1,6, *
1
Department of Life Sciences and Institute of Genome Sciences, 2Genome Research Center, 3Faculty of Medicine,
School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan, ROC, 4Department of Ophthalmology,
Cheng-Hsin General Hospital, Taipei, 11220, Taiwan, ROC, 5Department of Ophthalmology, 6Department of
Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan, ROC and 7Institute of Molecular and
Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
*To whom correspondence should be addressed. Email: mychung@ym.edu.tw (M.-Y.C.); sjchen@vghtpe.gov.tw (S.-J.C.); yjjiang@nhri.org.tw (Y.-J.J.)

Abstract
Familial exudative vitreoretinopathy (FEVR) belongs to a group of genetically and clinically heterogeneous disorders in retinal
vascular development. To date, in approximately 50% of patients with FEVR, pathogenic mutations have been detected in FZD4,
LRP5, TSPAN12, NDP and ZNF408. In this study, we identified two heterozygous frameshift mutations in RCBTB1 from three
Taiwanese cases through exome sequencing. In patient-derived lymphoblastoid cell lines (LCLs), the protein level of RCBTB1 is
approximately half that of unaffected control LCLs, which is indicative of a haploinsufficiency mechanism. By employing transient
transfection and reporter assays for the transcriptional activity of β-catenin, we demonstrated that RCBTB1 participates in the
Norrin/FZD4 signaling pathway and that knockdown of RCBTB1 by shRNA significantly reduced nuclear accumulation of β-catenin
under Norrin and Wnt3a treatments. Furthermore, transgenic fli1:EGFP zebrafish with rcbtb1 knockdown exhibited anomalies in
intersegmental and intraocular vessels. These results strongly support that reduced RCBTB1 expression may lead to defects in
angiogenesis through the Norrin-dependent Wnt pathway, and that RCBTB1 is a putative genetic cause of vitreoretinopathies.

Introduction sporadic retinal telangiectasis, is a rare disorder with an esti-


First described in 1969, familial exudative vitreoretinopathy mated incidence of 0.09 in 100 000 in UK (11). In addition to pre-
(FEVR; [OMIM 133780, 305390, 605750, 601813, 613310, and dominantly affecting males and typically affecting only one
616468]) is a rare developmental disorder of the retinal blood ves- eye, Coats disease is characterized by telangiectasia of the retinal
sels characterized by incomplete peripheral vascularization (1,2). vessels, subretinal lipid exudation, macular edema and RD re-
FEVR has two prominent clinical features: intrafamilial variabil- sulting from defects in retinal vascular development (12–14).
ity (3–8), in which individuals carrying the same mutation may Both FEVR and Coats disease, in addition to other congenital
present with a wide range of severities, from asymptomatic to vitreoretinopathies such as persistent hyperplastic primary
the most complicated retinal detachments (RDs); and disease vitreous (OMIM 611308) and Norrie disease (OMIM 310600),
asymmetry, with presentations of severity varying between the share similar fundus features, including avascularization of the
two eyes (9,10). Coats disease (OMIM 3 00 216), also known as peripheral retina and subretinal exudation. Therefore, they


These authors contributed equally to this work.
Received: December 15, 2015. Revised and Accepted: February 8, 2016
© The Author 2016. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com

1637
1638 | Human Molecular Genetics, 2016, Vol. 25, No. 8

have been proposed to be in a spectrum of vitreoretinopathies with informed consent (Fig. 1; Supplementary Material, Fig. S1).
(4,6,9,15–18). We performed direct sequencing of the exons and exon–intron
Various Mendelian inheritance patterns of FEVR have been boundaries of the five candidate genes, i.e. FDZ4, LRP5, NDP,
documented (15,19–21), mainly autosomal dominant with re- TSPAN12 and ZNF408, in the affected participants. We found
duced penetrance estimated to be less than 50% (22) and thus two previously identified mutations in FZD4 in two families,
may appear as sporadic cases. To date, mutations in five genes, specifically, c.313A>G in family E1 and c.1282_1285delGACA in
namely frizzled 4 (FZD4 [OMIM 6 04 579]), low-density lipoprotein family E10; and detected a novel single-nucleotide variant (NM_
receptor-related protein 5 (LRP5 [OMIM 6 03 506]), tetraspanin-12 000266.3:c.-77A>G) at the 5′UTR of NDP in family E9. This was not
(TSPAN12 [OMIM 6 13 138]), Norrin (NDP [OMIM 3 00 658]) and detected in 320 X chromosomes from ethnically matched popula-

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


zinc finger protein 408 (ZNF408 [OMIM 6 16 454]), have been iden- tion controls. To evaluate whether this 5′UTR mutation affects
tified to be highly associated with FEVR (6,15,17,23–26). All of the NDP expression level, we generated constructs with a wild-
these genes have been found to play a role in normal vascular de- type (WT) or mutant 5′UTR upstream of the firefly luciferase cod-
velopment in animal models, and the first four genes are crucial ing sequence. According to normalized luciferase activities, we
components of the Norrin-induced FZD4/β-catenin signaling estimated the expression level in the mutant 5′UTR to be half
pathway (23,27). However, mutations in these genes account for that of the WT 5′UTR (Supplementary Material, Fig. S2). To assess
only approximately 50% of cases of FEVR (6,17,28) and other the effect of this mutation on the Norrin-dependent Wnt signal-
vitreoretinopathies such as Coats disease and Norrie disease ing pathway, we cotransfected the NDP expression constructs
have also been demonstrated sharing the same genetic factors with a WT or mutant 5′UTR at two different doses with FZD4,
(NDP and/or FZD4) (29,30). Although Coats disease usually ap- LRP5 expression constructs and a reporter with TCF/LEF-binding
peared sporadic, there are genetic evidences that Coats disease sites into ARPE19 cells. The results revealed a dose-dependent re-
is linked to the NDP/wnt signaling pathway, one is a somatic mu- duction of Wnt signaling. A significant reduction of the reporter
tation in NDP identified in a boy (12) and the other is a germline was evident only in transfections with a higher dose of NDP con-
mutation also in NDP in an affected female who gave birth to a structs (150 ng), but not in the group with a lower construct dose
boy diagnosed with Norrie disease (31). These findings have sug- (75 ng) (Supplementary Material, Fig. S2). Because of limited
gested that vitreoretinopathies may result from mutations from information regarding the physiological concentration of Norrin
the same group of genes and that additional underlying genetic during retinal angiogenesis, we could not reach a definitive
factors for these disorders may also exist and function in early conclusion on whether this variant was the major genetic factor
retinal vascular development. and a sufficient cause of FEVR in family E9.

Results
Exome sequencing and identification of potentially
Family recruitment and mutation screening for known pathogenic variants
FEVR-related genes
To detect disease-associated novel variants, we performed
Fifteen patients diagnosed with FEVR or Coats disease and their whole-exome sequencing in affected individuals from families
family members were recruited from nine Taiwanese families E5 (II-1), E6 (II-1) and E9 (III-2 and III-4). For each case, the average

Figure 1. Pedigrees of two families with heterozygous mutations in RCBTB1 and the clinical features of affected members. (A) Pedigree structure and partial
chromatograms of Sanger confirmation sequencing for families E5 and E9. Because of the reduced penetrance and phenotypic variability in FEVR, the mothers in
family E9 were assumed to be nonpenetrant carriers. (B) Fundi of three members in family E9. Both eyes were affected in individuals III-2 and III-4. In individual III-2,
disc-dragging with macular ectopia in the right eye and total RD in the left eye were evident. The fundi of individual III-4 exhibited a fibrovascular stalk (arrowhead)
emanating from the disc to the periphery in the right eye, and disc-dragging with macular ectopia in the left eye. The fundi of carrier II-4 appeared unremarkable. (C)
The fundus of individual II-1 of family E5 exhibited marked subretinal lipid exudates (arrowhead), with total exudative detachment in the right eye. The left eye was
unremarkable. The diagnosis was Coats disease on the right eye. The retina was reattached after cryopexy, pars plana vitrectomy and removal of the subretinal
fibrous cord and encircling buckle. However, a massive lipid exudate remained at the posterior pole. OD, right eye; OS, left eye. *Reference sequences for RCBTB1:
NM_018191.3, NC_000013.10.
Human Molecular Genetics, 2016, Vol. 25, No. 8 | 1639

Table 1. Clinical characterization of subjects

Family ID E9 E5
Individual ID III-2 III-4 II-2 II-4 I-1 I-2 II-2

Sex M M F F M M F
Mutation NDP: c.-77A>G RCBTB1: c.707delA –
RCBTB1: c.1172+1G>A ( p.Glu349Glyfs*17) ( p.Asn236Thrfs*11)
Onset age Congenital Congenital – – 3 y/o – –

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


Clinical features
Anterior segment
OD Unremarkable Status post intraocular lens implantation Unremarkable Unremarkable Aphakia ND ND
OS Cornea opacity Unremarkable Unremarkable
Fundoscopic exam
OD Macular ectopia Fibrovascular stalk Unremarkable Unremarkable Exudative RD ND ND
OS Traction RD and Macular ectopia Unremarkable
retinal break

Reference sequences for NDP and RTBCB1 are NM_000266.3 and NM_018191.3, respectively.
+, Present; −, absent; F, female; ID, intellectual disability; M, male; ND, not documented; OD, right eye; OS, left eye.

coverage was at least 90-fold, with more than 90% of the targeted and a frameshift with a truncated protein was predicted (NP_
exons covered at least 20 times. Because of the availability of a 060661.3:p.Glu349Glyfs*17) (Fig. 2A and B). Immunoblot analysis
pair of third-degree relatives (i.e. III-2 and III-4 in E9), we first ana- of RCBTB1 using total proteins from LCLs from carriers of family
lyzed their exome data. After conducting variant filtering to fulfill E9 revealed an expression level that was approximately half that
an autosomal dominant or X-linked inheritance and to exclude of control LCLs without predicted truncated proteins (Fig. 2C),
known variants in dbSNP 138, we identified 130 candidate var- suggesting the haploinsufficiency of this variant on RCBTB1.
iants in both III-2 and III-4 in family E9 (Supplementary Material,
Table S1), 27 of which passed confirmation based on information
Functional characterization of rcbtb1 in zebrafish
provided by the Ensembl Genome Browser. To further narrow
down the candidate gene list, we included the exome data of II- To examine the role of RCBTB1 in angiogenesis in vivo, we con-
1 in E5 and II-1 in E6, and applied the modified overlap strategy ducted morpholino (MO)-mediated knockdown of rcbtb1 in the
(32) by focusing on novel variants in the same gene in at least Tg(fli1:EGFP) zebrafish line (34) to investigate its effects on embry-
two of the three families. We identified two novel variants with onic blood vessel development. Compared with those injected
predicted damaging effects in RCBTB1 in families E5 and E9. with control MOs containing five mismatched nucleotides, mor-
In family E5, we detected a single-nucleotide deletion (NM_ phants with rcbtb1 translation-blocking MOs revealed compar-
018191.3:c.707delA) in RCBTB1 for individual II-1, who had been able gross development except some anomalies in the patterns
diagnosed with Coats disease, and in his unaffected father (indi- of the vasculatures in the intersegmental vessels (ISVs) and the
vidual I-1) (Fig. 1A). In family E9, a canonical splice-site variant intraocular vessels (IOVs). Some ISVs exhibited a smaller average
(NM_018191.3:c.1172+1G>A, NC_000013.10:g.50118872C>T) was area and even appeared as a streak of cells without lumen. These
found in two cousins diagnosed with FEVR and their asymptom- abnormalities resembled truncated ISVs at 3 days post fertiliza-
atic mothers (Fig. 1A). Neither of these two variants was detected tion (dpf ) and could be partially rescued through coinjection
in 186 unrelated, ethnic-matched population controls and was with human RCBTB1 mRNA (Fig. 3A and B). In a similar manner,
not present as rare variants in the Phase 3 1000 Genomes variants injections with rcbtb1 splicing MOs resulted in dose-dependent
(33). The clinical characteristics of these two families are aberrant splicing of rcbtb1 (Supplementary Material, Fig. S4) as
summarized in Table 1 and Figure 1. We also performed direct se- well as thinner ISVs, suggesting that rcbtb1 plays a role in early
quencing of all the exons and exon–intron boundaries of RCBTB1 embryonic angiogenesis in zebrafish. To accurately model clinic-
in the affected participants of the other recruited families. The al phenotypes of FEVR, we further investigated the effect of rcbtb1
results revealed only common single-nucleotide polymorphisms knockdown on IOVs in zebrafish using the ndp translation-block-
(SNPs) rather than the novel damaging SNVs (Supplementary ing MOs as a positive control. As expected, injections with ndp
Material, Table S2). MOs resulted in narrow IOVs and an increased area of avascular-
ization at 4 dpf, without inducing obvious effects on the ISVs
(Fig. 3C–E). We categorized the IOV phenotypes of the ndp mor-
Effect of novel variants on gene products
phants into three classes according to the proportion of the avas-
The RCBTB1 gene contains 13 exons encoding a 531-amino-acid cular area: class I was the baseline; class II indicated thinner IOVs
protein with two predicted domains, namely RCC1 at the N-ter- and an avascular area range between 25 and 50% and class III
minus and the BTB domain at the C-terminus (Fig. 2A), and it is represented thinner IOVs and an avascular area larger than
ubiquitously expressed in various tissues (Supplementary Mater- 50% (Fig. 3C). Compared with the IOV phenotypes in the ndp mor-
ial, Fig. S3). The single-nucleotide deletion (c.707delA) in family phants, a milder effect was found in the rcbtb1 morphants, specif-
E5 is located in exon 7, whereas the novel variant (c.1172+1G>A) ically 79 and 87% in classes II to III for rcbtb1 and ndp morphant
in family E9 was located at the splicing donor site of intron 10. To groups, respectively. To determine the genetic interactions be-
verify the effect of the splice-site variant on the RCBTB1 tran- tween Norrin signaling and rcbtb1 during IOV development in
script, we performed RT-PCR followed by amplicon sequencing, vivo, we investigated the effect of combined MOs targeting ndp
where we used the leukocyte RNA from the participants of family and rcbtb1. Coinjections of half-dose ndp MOs (3 ng) with rcbtb1
E9. An aberrant transcript with a skipped exon 10 was found, mismatch-control MOs (3 ng) resulted in mild IOV anomalies
1640 | Human Molecular Genetics, 2016, Vol. 25, No. 8

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


Figure 2. Effect of splice-site mutation on mRNA and protein expression. (A) RCBTB1 gene structure and predicted domains. The UTRs and coding sequences are indicated
in the clear and solid boxes, respectively, and are labeled with exon numbers. Domains in RCBTB1 (UniProt Q8NDN9) include RCC1 (blue) and BTB (green). The nucleotide
positions and predicted changes in amino acids of the two heterozygous mutations identified in families E5 and E9 are indicated below the gene structure. The predicted
additional amino acid residues located downstream of the frameshift mutations are in red. RCC1, the regulator of chromosome condensation 1; BTB, BR-C, ttk and bab. (B)
Aberrant RTBCB1 transcripts with skipped exon 10 were identified through RT-PCR by using RNA extracted from peripheral blood leukocytes from the members of family
E9. Direct sequencing of the RT-PCR products revealed the direct joining of exons 9 and 11 of RCBTB1, and a frameshift mutation with premature termination ( p.
Glu349Glyfs*17) was predicted. Messenger RNA without reverse transcription was used as the template for the negative control, RT(–). (C) Immunoblot analysis of
RCBTB1 (apparent mobility at ∼55 kDa) in LCLs derived from the members of family E9 or baseline controls. The predicted truncated form (∼38 kDa) was not detected,
and approximately half the expression level of WT RCBTB1 was noted in all heterozygotes from family E9.

(25% of class II, but 0% of class III), whereas coinjections of half- fractionation and immunoblot analyses. The quality of the frac-
dose ndp MOs (3 ng) with rcbtb1 MOs (3 ng) severely impaired the tionation was assessed using Lamin B1 and GAPDH as markers
IOV structure, and resembled those observed in the working-dose for nuclear and cytosolic fractions, respectively (Fig. 4B). In
ndp morphants (Fig. 3C and D, respectively). These findings APRE19 cells transfected with FZD4/LRP5, the nonphosphory-
suggest that rcbtb1 is involved in the Norrin-dependent IOV lated β-catenin was upregulated significantly in the nuclear frac-
development in zebrafish. tion under Norrin/Wnt3a activation for 4 h. However, the
increase of nuclear β-catenin under Norrin/Wnt3a activation
was abolished in RCBTB1 knockdown groups without affecting
RCBTB1 participates in the Norrin/β-catenin signaling
the level of β-catenin in the cytosolic fractions (Fig. 4B and C).
pathway by regulating the nuclear accumulation
This effect was similarly revealed at the single-cell level in im-
of β-catenin
munofluorescence analysis using an anti-β-catenin antibody in
We determined the role of RCBTB1 in the Norrin/β-catenin signal- ARPE19 cells transfected with FZD4, LRP5 and either RCBTB1-tar-
ing by performing TCF/LEF reporter assays in ARPE19 cells. Cells geting shRNA or the pLKO.1 vector under serum-free media with
were cotransfected with plasmids expressing FZD4 and LRP5, re- 250 ng/mL rhNorrin or Wnt3a for 4 h. The cells in RCBTB1 knock-
porter constructs and one of the two RCBTB1-targeting shRNA down groups revealed significantly reduced nuclear accumula-
plasmids (shRCBTB1-1 or shRCBTB1-2) with different knockdown tion of β-catenin under treatments with either Norrin or Wnt3a
efficiency, or the pLKO.1 vector as a sham control. At 24 h after (Fig. 4D), suggesting that RCBTB1 is located upstream of β-catenin
transfection, the cells were cultured in the presence of 0, 62.5 or and regulates its nuclear accumulation in the FZD/β-catenin sig-
125 ng/mL recombinant human Norrin (rhNorrin) for another naling pathway.
24 h. Compared with no-ligand and mutant reporter controls
(M51), both concentrations of rhNorrin can effectively activate
TCF/LEF-driven expression of luciferase in a dose-dependent
Discussion
manner (Fig. 4A). In the RCBTB1 knockdown groups, we estimated By employing exome sequencing, bioinformatic analysis and the
the Norrin-induced activation to be approximately 50 and 33% of modified overlap strategy, we identified two frameshift mutations
that of the vector control under high and low doses of Norrin, re- in RCBTB1 (c.707delA and c.1172+1G>A) in two unrelated Taiwan-
spectively, and the reduction of Norrin-induced signaling was ese families affected by Coats disease and FEVR. To date, in add-
proportional to the knockdown efficiency of the shRNA plasmids ition to the five FEVR-associated genes, RCBTB1 was identified as
(Fig. 4A; Supplementary Material, Fig. S5). A more pronounced re- the sixth putative causative gene in two out of nine families,
duction of Norrin-induced signaling upon RCBTB1 knockdown at reflecting the genetic heterogeneity of FEVR and suggesting that
a lower dose of Norrin may reflect a synergistic effect between other genetic factors may also contribute to the development of
NDP and RCBTB1, indicating that RCBTB1 may be involved in the vitreoretinopathies.
Norrin/β-catenin signaling pathway. To further investigate the RCBTB1, also known as CLLD7, is located at chromosome
ligand-specific response of RCBTB1 in Norrin-dependent signal- 13q14.3, which is frequently deleted in cases of B-cell chronic
ing, cells cotransfected with FZD4, LRP5, reporter constructs and lymphocytic leukemia and other neoplasms (35). Although
RCBTB1-targeting shRNA plasmids or the pLKO.1 vector were in- RCBTB1 was thought to be a tumor suppressor gene (35), its physio-
cubated in a medium with 100 ng/mL Wnt3a, and we also found a logic and pathologic roles remain unclear because of limited ex-
reduction in reporter activity for the RCBTB1 knockdown group perimental data (36). RCBTB1 is composed of two domains: the
(Fig. 4A). These results suggest that RCBTB1 may be a common N-terminal RCC1 domain may act as a guanine exchange factor
downstream component of the FZD4/LRP5 signaling pathway. for Ran (37), a critical protein for nuclear transport and the BTB do-
Because β-catenin is tightly regulated in both cytoplasm and nu- main is implicated in protein–protein interactions (38). Moreover,
cleus, the effect of RCBTB1 on β-catenin in different cellular com- a previous study found that the BTB domain of RCBTB1 can act as a
partments in ARPE19 cells with or without Norrin/Wnt3a substrate adaptor for the CUL3-based E3 ligase and can interact
activation was analyzed and quantified by subcellular with UbcM2, a highly conserved E2 ubiquitin-conjugating enzyme
Human Molecular Genetics, 2016, Vol. 25, No. 8 | 1641

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019

Figure 3. Functional characterization of RCBTB1 through morpholino (MO)-mediated gene knockdown in zebrafish embryos. (A) Morphant rcbtb1 zebrafish showed thinner
and irregular ISVs. Lateral views of trunk vasculatures (dorsal side facing up) in fli1:EGFP transgenic larvae at 3 dpf injected with the control MO, rcbtb1-ATG MO, or
combinations of rcbtb1-ATG MO and mRNA encoding human WT RCBTB1. ISVs project from the dorsal aorta toward the dorsal longitudinal vessel at the top. (B)
Quantitative analysis of the average areas of eight ISVs counted posteriorly from the urogenital pole in different morphant groups normalized to the average of no-
injection controls. The values are represented as the mean ± standard error of the mean (SEM). Asterisks indicate a significant difference, with a P of <0.05. (C)
1642 | Human Molecular Genetics, 2016, Vol. 25, No. 8

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


Figure 4. Knockdown of RCBTB1 significantly reduced the activation of the Norrin- or Wnt3a-mediated Wnt/β-catenin pathway by reducing the nuclear accumulation of β-
catenin. (A) Luciferase assays were conducted using ARPE19 cells. The cells were cotransfected with shRNA targeting RCBTB1 (shRCBTB1-1 and shRCBTB1-2) or the vector
control ( pLKO.1), with expression vectors encoded for human FZD4, LRP5 and pmirGlo-derived dual luciferase constructs with seven human TCF/LEF-binding sequences
(M50) or mutant TCF/LEF-binding sequences (M51) at the firefly luciferase promoter. The transfected cells were treated with 0, 62.5, 125 ng/mL rhNorrin or 100 ng/mL
Wnt3a ligands. Compared with no ligand control and mutant reporter controls (M51), both rhNorrin and Wnt3a can activate TCF/LEF-driven expression of luciferase
and the activation was significantly reduced by the expression of RCBTB1-targeting shRNA. The RLA is the ratio of Firefly and Renilla luciferase activities and is
presented as the mean ± SEM (*P < 0.05, ns, not significant). (B) Subcellular fractionation and immunoblot analysis of ARPE19 cells cotransfected with expression
vectors encoding human FZD4 and LRP5 with or without RCBTB1-targeting shRNA (shRCBTB1-1), in the presence of 250 ng/mL rhNorrin or Wnt3a for 4 h. The
nonphosphorylated β-catenin was upregulated significantly in the nuclear fraction in control groups, which was abolished in RCBTB1 knockdown groups without
affecting the level of β-catenin in the cytosolic fractions. (C) Quantification of (B) in at least triplicate. The non-P-β-catenin levels in each groups were normalized to
control groups without Norrin or Wnt3a treatment and presented as the mean ± SEM (*P < 0.05, ns, not significant). (D) Immunofluorescence with anti-β-catenin
(green) in ARPE19 cells cotransfected with expression vectors encoding human FZD4 and LRP5, shRNA targeting RCBTB1 (shRCBTB1-1) or the vector-only control
( pLKO.1) in the presence of 250 ng/mL rhNorrin or Wnt3a for 4 h. Significant nuclear accumulation of β-catenin was noted in the vector control group rather than in
the RCBTB1 knockdown group. The arrowheads indicate the locations of nuclei in cells.

(39). The abundant expression of UbcM2 in the retina (40) and an- morpholino-mediated gene knockdown were reported (42), injec-
other E3 ubiquitin ligase, Fbxw7, has been demonstrated to act as a tions with two different rcbtb1 MOs, a mismatch control, the res-
potent positive regulator in angiogenesis by inhibiting Notch sig- cue experiment using human WT and mutant RCBTB1 mRNA as
naling in zebrafish (41). These findings raise the possibility that well as CRISPR-mediated transcriptional knockdown were
RCBTB1-mediated ubiquitination may be involved in the regula- carried out to minimize the possibilities for false-positive pheno-
tion of angiogenic pathways such as Norrin-induced β-catenin sig- types and off-target effects of MOs in this study (Fig. 3; Supple-
naling. How these multiple roles and interactions of RCBTB1 are mentary Material, Figs S6 and S7). In this way, delayed sprouting
conducted at the cellular level has yet to be investigated. of trunk vessels and defects in ISVs and IOVs were revealed in
To link RCBTB1 to vascular development in vivo, knockdown of rcbtb1 morphants, which mimic the avascularization in FEVR
rcbtb1 by morpholinos in zebrafish was performed to investigate and Coats disease under haploinsufficient RCBTB1 genotypes
the role of rcbtb1 in angiogenesis. Although some poor correlation and thus support the correlation between the genotypes and phe-
between phenotypes in germline mutants and those in notypes. Moreover, compared with the phenotypes of rcbtb1

Morphant rcbtb1 zebrafish exhibited moderate defects in IOV development. The images depict the IOVs of fli1:EGFP transgenic zebrafish larvae at 4 dpf. Phenotypes of the
IOVs were categorized into three classes: baseline (class I), moderately affected (class II) and severely affected (class III). The IOVs in class I had a normal width and a
regular radial configuration, with an avascular area occupied <25% of the total space. However, narrow and less regular radial vasculatures with an avascular area
ranging between 25 and 50%, or >50%, were classified into classes II and III, respectively. (D) Semiquantitative analyses of IOV phenotypes in morphants injected with
the control MO (6 ng), rcbtb1-ATG MO (6 ng), ndp ATG MO (6 ng), or combinations of 3 ng of the rcbtb1 control or ATG MO with 3 ng of ndp ATG MO. At least 40 larvae per
condition from three independent experiments were evaluated. (E) Comparison of IOV and ISV phenotypes in fli1:EGFP transgenic larvae at 4 dpf, with the injection of 6 ng
ndp ATG MO or rcbtb1-ATG MO. Both morphant groups had class II IOV phenotypes, but the thinner and irregular ISV phenotypes (white arrowhead) were noted only in
rcbtb1-ATG morphants.
Human Molecular Genetics, 2016, Vol. 25, No. 8 | 1643

knockdown, the effect of reduced ndp is restricted to the IOVs but factors associated with FEVR. The frequency of gene mutations
not observed in the ISVs. Further, we showed genetic interaction may reflect their roles in angiogenesis, i.e. mutations in crucial
between NDP and RCBTB1 in vitro and in vivo. These findings sug- rate-limiting factors such as FZD4 and LRP5 tend to be more fre-
gest that ubiquitously expressed RCBTB1 may act as a common quently identified, whereas fewer mutations occur in the regula-
downstream component of angiogenesis in general, while the tis- tory components that modulate the major signaling pathways. A
sue- or organ-specific angiogenesis is guided by other specific fac- recent study involving a cohort of 92 FEVR families identified mu-
tors, e.g. Norrin and TSPAN12. tations in LRP5 and FZD4 in 19% and 15% of cases, respectively. Of
In the patient cohort, we identified two heterozygous frame- the families recruited, 48.5% had a confirmed molecular diagno-
shift mutations in RCBTB1, one of which is a splice-site variant sis (28), indicating that the Norrin-induced β-catenin signaling

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


(c.1172+1G>A) resulting in the skip of exon 10 in RCBTB1 tran- pathway plays an essential role in retinal angiogenesis. To date,
scripts, thereby leading to half the amount of normal RCBTB1 in mutations have been identified in only approximately 50% of
LCLs. Furthermore, partially reducing RCBTB1 by using shRNA in cases, providing an opportunity to investigate (retinal) angiogen-
the transiently transfected cell model and MOs in zebrafish signifi- esis by identifying additional genetic factors associated with
cantly lowered Norrin-induced β-catenin signaling by reduced FEVR and related vitreoretinopathy.
nuclear accumulation of β-catenin and led to poorly developed In conclusion, we identified RCBTB1 as a gene associated with
vasculatures, respectively. These results suggest that RCBTB1 hap- vitreoretinopathy and found that it plays a role in retinal angiogen-
loinsufficiency may lead to the observed disease phenotypes. In esis through Norrin-induced β-catenin signaling. Further investiga-
addition, according to the reporter assay, the novel variant at the tion of its physiological role may elucidate its detailed functions in
5′UTR of NDP (c.-77A>G) may have resulted in a halved Norrin level retinal angiogenesis.
in family E9, in which both children affected were hemizygous for
the mutant allele, whereas their unaffected mothers were hetero-
Subjects, Materials and Methods
zygous. On the basis of the Norrin and RCBTB1 levels estimated ac-
cording to the genotypes of the members of family E9 (Figs 1B and Ethics statement for human subject research
2C) and additional evidence obtained from cell transfection ex-
This study was approved by the Institutional Review Boards of
periments (Fig. 4A; Supplementary Material, Fig. S2B) and zebra-
Taipei Veterans General Hospital (for FEVR) and Cheng-Hsin Gen-
fish morphant analyses (Fig. 3D), we propose a possible disease
eral Hospital (for Coats disease). All research studies involving
mechanism, in which normal angiogenesis can be achieved as
humans were conducted according to the principles expressed
long as the Norrin-induced signals exceed a threshold. Thus, com-
in the Declaration of Helsinki. Written informed consent was
bined genetic factors, such as mutations in RCBTB1 and NDP, both
obtained from all participants.
influencing Norrin-induced signaling, enhance disease suscepti-
bility in a dynamic microenvironment, leading to defects charac-
terized by a wide spectrum of severity in retinal angiogenesis. Patient recruitment
Reduced penetrance and asymmetric retinal phenotypes have Fifteen patients diagnosed with FEVR or Coats disease and their
long been noted in vitreoretinopathies (22). In all affected cases family members were recruited from nine Taiwanese families
with RCBTB1 mutations, asymmetry in retinal manifestation was and provided informed consent (Fig. 1; Supplementary Material,
obvious as documented in Table 1. In addition, there are heterozy- Fig. S1). The patients provided a detailed history by answering
gous nonpenetrating parents with RCBTB1 mutations in both fam- questions in order to rule out retinopathy of prematurity, and oph-
ilies E5 and E9. These characteristics were also found in family E10 thalmic examinations included assessments of corrected visual
with a reported FZD4 mutation (Supplementary material, Fig. S1A; acuity, slit-lamp examinations, intraocular pressure application
clinical feature not shown). These phenomena could also result and dilated fundus examination with photographs with or without
from the effect of genetic background in heterozygous individuals fluorescein angiography. The fundus features were grouped into
and microenvironmental factors in the eyes. Recently, a rare the following categories for diagnosis: avascular zone only; extra-
variant in RCBTB1 was detected in a consanguineous family with retinal neovascularization; exudative RD; tractional RD; rhegmato-
retinal ciliopathy (43), a disease of photoreceptors. A follow-up genous RD; retinal breaks without RD; retinoschisis and macular
electroretinogram (ERG) was performed in the proband, individual ectopia. Approximately 20 ml of peripheral venous blood was
III-2, of family E9 (Supplementary Material, Fig. S1B). It was drawn, with 10 ml being used for genomic DNA and total RNA iso-
revealed that the rod and cone responses were preserved in his lation conducted according to our established protocol and the
relatively normal eye with only mild disc dragging, while the other 10 ml being used for the establishment of LCLs by the Biore-
ERG was flat in the other eye with severe RD. Based on his fol- source Collection and Research Center (BCRC), Hsinchu, Taiwan,
low-up histories during past 20 years, including the preserved for additional RNA and protein analyses.
ERG, lack of night blindness and stable clinical features of FEVR
without bony spicules or sheathing vessels, all suggest that he is
Direct sequencing of candidate genes and mutation
a patient with FEVR without clinical overlap of retinitis pigment-
analysis
osa (RP). Besides, the pathophysiologies of vitreoretinopathies
and RP are quite different, where angiogenesis is the primary Genomic DNA was extracted from the peripheral blood leuko-
defect in vitreoretinopathies and usually asymmetric while RP cytes of all participants, according to standard lab protocols. All
affects the photoreceptors and almost always in both eyes. Thus, exons and exon–intron boundaries of the candidate genes (NDP,
homozygous RCBTB1 variants in retinal ciliopathy could be an FZD4, LRP5, TSPAN12 and ZNF408) were amplified under opti-
example of clinical heterogeneity. mized PCR conditions. Subsequently, direct sequence analysis
In addition to known FEVR-associated genes, many other was conducted using the ABI PRISM Big Dye Terminator Cycle Se-
angiogenesis-related genes have been identified in zebrafish quencing V2.0 Ready Reaction Kit and ABI PRISM 3730 DNA ana-
(44), indicating the presence of numerous complex signaling lyzer (Applied Biosystems). The allele frequencies of all three
pathways involved in angiogenesis. On the basis of the findings, novel variants (c.-77A>G of NDP; c.707delA and c.1172+1G>A of
we can speculate the existence of additional unidentified genetic RCBTB1) in at least 150 Taiwanese controls were assessed by
1644 | Human Molecular Genetics, 2016, Vol. 25, No. 8

conducting direct Sanger sequencing. Reference sequences for Transient transfections of ARPE19 with expression vectors or
NDP and RTBCB1 are NM_000266.3 and NM_018191.3, respectively. shRNA were performed using Lipofectamine® 3000, according
These variants have been submitted to the ClinVar database to manufacturer instructions.
(http://www.ncbi.nlm.nih.gov/clinvar/).
RNA extraction and RT-PCR
Exome sequencing analysis and variant filtering Total RNA was extracted from peripheral blood leukocytes or LCLs
Exome sequencing was performed by the Genome Research Center by using the TRIzol® Reagent (Ambion) according to the manufac-
of National Yang-Ming University for four affected individuals (i.e. II- turer protocol. Reverse transcriptase reactions were performed on

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


1 of family E5, II-1 of family E6, III-2 and III-4 of family E9) after ex- 500 ng of total RNA by using M-MLV Reverse Transcriptase (Life
cluding possible pathogenic mutations in the five candidate genes. Technologies), and 1 μl of cDNA was used to conduct PCR. The
Exon sequences were enriched from genomic DNA by using the Agi- PCR products were electrophoresed in a 2% TBE gel, and the pro-
lent SureSelect All Exon 50 Mb or ROCHE NimbleGen SeqCap EZ ducts of the aberrant transcripts were excised from the gel and
Exome V3+UTR (96 Mb) kits and were paired-end sequenced on an extracted using the Zymoclean™ Gel DNA Recovery Kit (Zymo Re-
Illumina HiSeq 2000 or 2500 platform. The CLC Genomics Work- search) prior to sequencing. Details of the primer sequences are
bench v7.0.4 was used for basic quality assessment and variant call- shown in Supplementary Material, Table S3.
ing. For each patient sample, at least 90% of the targeted region was
covered by at least 20 folds, as displayed in Supplementary Material, Whole cell lysate preparation
Table S1. Variants that were not in the dbSNP138 common-variant
The cultured cells were lysed with RIPA buffer (50 m Tris–HCl, pH
list were filtered for being heterozygous and were present in both
8.0, 150 m NaCl, 0.1% SDS, 1% NP-40 and 0.5% sodium deoxycho-
III-2 and III-4 of family E9. Filtering was followed by removing var-
late) supplemented with 1× protease inhibitor cocktail (cOmplete
iants listed in the in-house exome database for unrelated pheno-
Protease Inhibitor Cocktail Tablets, Roche). The cell lysates were
types, and the remaining variants were analyzed using the
cleared through centrifugation at 14 000 × g for 20 min, and the
Variant Effect Predictor of the Ensembl Genome Browser. The 130
protein concentration was quantified using the Bradford protein
variants with predicted consequences of in-frame deletion, mis-
assay (BioRad).
sense, stop-gained or splice-site variants were individually verified
using Ensembl (release 75, Feb. 2014). The remaining 27 variants
were then prioritized through segregation according to the results Nuclear and cytosolic fractionation
obtained from Sanger sequencing, pathway analyses with known
The method for subcellular fractionation was based on Dr David
candidate genes and the presence of other variants in the same
Ron’s Lab’s protocol (http://ron.cimr.cam.ac.uk/protocols.html).
gene in individuals II-1 of family E5 and/or II-1 of family E6.
Briefly, cells were washed twice with ice-cold calcium-tris-buffered
saline (CTMS) (10 m Tris–HCl, pH 7.4, 140 m NaCl, 2 m CaCl2).
Plasmids and constructs For one 10-cm dish, cells were scraped from the dish in 1 ml of
CTBS buffer supplemented with 2 m DTT, 5 m EDTA and 1× pro-
The WT and mutant TCF/LEF-binding sequences were amplified
tease inhibitor cocktail (Roche). The harvested cells were pelleted,
using M50 Super 8× TOPFlash and M51 Super 8× FOPFlash plas-
resuspended in 0.4 ml serum-free media containing with 7.5 m
mids (Addgene) as the template, respectively, and were cloned up-
N-ethylmaleimide (Sigma-Aldrich) (45) and incubated at room
stream of the firefly luciferase gene in pmirGlo vectors (Promega).
temperature for 10 min. After washed in ice-cold CTBS, cells were
The expression constructs for human NDP with the 5′UTR region,
lysed in Harvest buffer (10 m HEPES, pH 7.9, 50 m NaCl, 0.5  su-
FZD4, and LRP5 were generated using pcDNA3.1myc-His vectors
crose, 0.1 m EDTA, 0.5% Triton × 100) containing 1× protease in-
(Invitrogen). Site-directed mutagenesis was performed through
hibitor cocktail (ROCHE) and 1× phosphatase inhibitor cocktail
PCR by using Phusion DNA polymerase (New England Biolabs) as
(Sigma). The cleared supernatant is the cytoplasmic/membrane
well as the corresponding primers shown in Supplementary Ma-
fraction. The washed nuclear pellets were lysed in buffer 2 × C
terial, Table S3, to generate mutant constructs. All DNA constructs
(20 m HEPES, pH 7.9, 1  NaCl, 0.2 m EDTA, 0.2 m EGTA, 0.2%
after site-directed mutagenesis were validated through Sanger se-
NP40). The cleared supernatant contains the nuclear fraction.
quencing. Two RCBTB1-targeting shRNA plasmids and the pLKO.1
vector were obtained from the National RNAi Core Facility at the
Institute of Molecular Biology/Genomic Research Center, Aca- Immunoblot analysis
demia Sinica (Taipei, Taiwan), with the following oligo sequences: For immunoblot analysis, protein lysates were electrophoresed on
shRCBTB1-1 (TRCN0000160987): CCGGGCCAAATTACAAGTGGGT 10% SDS-PAGE gels and transferred to PVDF membranes (Milli-
GAACTCGAGTTCACCCACTTGTAATTTGGCTTTTTTG, shRCBTB1-2 pore). Antibodies to RCBTB1 (ab154649, Abcam®), active (nonpho-
(TRCN0000273669): CCGGCCTTACTGTGAAGGAATAATTCTCGAG spho) β-catenin (#8814, Cell Signaling Technology®), Lamin B1
AATTATTCCTTCACAGTAAGGTTTTTG, pLKO.1 empty vector: (ab133741, Abcam®), GAPDH (NB300-322, Novus Biologicals) and
CCGGACACTCGAGCACTTTTTG. β-actin (8H10D10, Cell Signaling Technology®) were used to detect
and quantify the proteins. Chemiluminescent images were cap-
tured using the BioSpectrum Imaging System (UVP), and signal in-
Cell culture and transient transfection
tensities were analyzed using ImageJ (Image Processing and
ARPE19 cells (ATCC number: CRL-2302) were cultured in DMEM/ Analysis in Java, http://imagej.nih.gov/ij/).
F12 (GIBCO®) supplemented with 10% fetal bovine serum (FBS)
in a 5% CO2 incubator. LCLs derived from Epstein-Barr-virus-
Dual luciferase assay
transformed lymphocytes (BCRC, Hsinchu, Taiwan) from the
members of family E9 were cultured in RPMI 1640 supplemented In 24-well plates, 105 ARPE19 cells/well were transfected with
with 20% FBS, -glutamine and penicillin–streptomycin solution 550 ng of DNA by using 1 μl of the Lipofectamine® 3000 Reagent
(GIBCO®) in a 5% CO2 incubator standing in the upright position. (Invitrogen). The DNA mixture contained 150 ng each of the
Human Molecular Genetics, 2016, Vol. 25, No. 8 | 1645

expression constructs for FZD and LRP5, 150 ng of the RCBTB1- those of the control group. For characterizing the intraocular
targeting shRNA plasmid or vector control and 100 ng of reporter phenotype at 4 dpf, injected embryos were classified into three
constructs (M50) or mutant reporter constructs (M51). The cells classes of phenotypes according to the relative severity and the
were stimulated 24 h post-transfection with 0, 62.5 or 125 ng/ ratio of the avascularization area compared with the MO-injected
mL human recombinant Norrin (rhNorrin) or 100 ng/mL Wnt3a (6 ng) control embryos. To determine the efficiency of splice
(R&D Systems), or by cotransfecting the indicated amount blocking, the RNA was isolated from 60 control MOs and different
of NDP-expressing constructs with WT or mutant alleles for doses of rcbtb1 splice MO-injected embryos at 1 dpf or 2 dpf by
16–18 h. Firefly and Renilla luciferase activity was measured using RNAzol®RT (Molecular Research Center) according to the
using the Dual-Luciferase® Reporter Assay System (Promega) manufacturer protocol. Reverse-transcriptase reaction was per-

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


according to the manufacturer instructions. Relative luciferase formed using 1 μg of RNA and Maxima Reverse Transcriptase
activity (RLA) is the ratio between the Firefly and Renilla lucifer- (Thermo Scientific) according to the manufacturer protocol. PCR
ase activities and is used to normalize transfection efficiency and was then conducted, as follows: (forward: CGGACCTCATGTT
changes in cell survival and growth. TTGCTGG and reverse: AACCCCAGCTGGCCATTAC). Fragments
were resolved through electrophoresis, extracted from the gel by
using the Zymoclean™ Gel DNA Recovery Kit (Zymo Research),
Immunofluorescence
and Sanger-sequenced as described.
ARPE19 cells transfected with DNA mixture contained 150 ng each
of the expression constructs for FZD and LRP5, 200 ng of the
Statistical analysis
RCBTB1-targeting shRNA plasmids or vector controls were cul-
tured in four-well chamber slides (Nunc® Lab-Tek®). Twenty- The means and standard error of the mean for all experimental
four hours after transfection, culture media were replaced with data were calculated. Comparisons between the experimental
serum-free only media or media containing 250 ng/ml rhNorrin conditions were conducted using a two-tailed Student t-test
or rhWnt3a for another 4 h and then washed with PBS for three for unpaired samples, with α < 0.05 considered to indicate
times. The cells were fixed using 4% paraformaldehyde at 37°C significance.
for 15 min and followed by three PBS washes. Then 0.25% Triton
X-100 in PBS was added to permeabilize cells at room temperature
Authors’ contributions
for 5 min and followed by PBST wash for three times. The cells
were incubated in 1% BSA in PBST for 1 h and then incubated in Project design: M.-Y.C., J.-H.W., J.-H.L., S.-J.C., Y.-J.J.; manuscript
1:100 diluted anti-β-catenin antibody (NBP1-54467SS, Novus) at writing: J.-H.W., M.-Y.C., S.-J.C., Y.-J.J.; project coordination and
4°C overnight. After washed for three times with PBST, the slides senior leaders of the groups: M.-Y.C., S.-J.C., Y.-J.J.; patient recruit-
were incubated in 1:200 diluted AlexaFluor488-conjugated Donkey ment and phenotyping: J.-H.L., H.-M.C., Y.-C.K., Y.-C.C., S.-J.C.;
anti-Mouse IgG H&L (ab150105, Abcam) for 1 h and afterwards genetic sequencing and interpretation: J.-H.W., M.-Y.C., C.-T.W.,
washed with PBST for three times. After mounted with SlowFade T.-T.L.; cell biology experiments: J.-H.W., M.-Y.C., C.T.-W.; and
Gold Antifade Mountant with DAPI (Life Technologies, S36942), the zebrafish experiments: J.-H.W., K.-C.C., Y.-J.J.
slides were examined by confocal microscope FV10i and analyzed
by the FV10-ASW 4.0 Viewer (Olympus).
Supplementary Material
Supplementary Material is available at HMG online.
Morpholinos and zebrafish embryo manipulations
The Tg(fli1:EGFP) zebrafish were maintained and mated as de-
scribed (34), and all experiments were conducted with the ap- Acknowledgements
proval of the Institutional Animal Care and Use Committee, We thank all the patients and their family members for participa-
National Health Research Institutes (NHRI-IACUC-103003). tion; Professors Chen-Kung Chou, Ming-Yuan Cheng and Yann-
Translation-blocking (GAGGCCACTTACTCACATCCACCAT), mis- Jang Chen for their valuable discussions and comments. We
matched-control (GAcGCCAaTTACTaACATaCACaAT) and splice also thank Dr Didier Y. R. Stainier for dCas9 and gRNA plasmids
blocking (GTTAAAAAGCATTCCCTCACCTCAC) MOs for the zebra- and the staff in the Zebrafish Facility of NHRI for their efforts in
fish rcbtb1 ortholog and translation-blocking (GAGCGACCGA maintaining fish stocks. Also, we are grateful to Chien-Ming
GTTCCTCATAGTGTC) MOs for ndp were designed and synthe- Wang and Chih-Hao Tang for their technical help.
sized using LLC (Gene Tools) and diluted in nuclease-free, deio-
nized, sterile water supplemented with 0.8% phenol red. To Conflict of Interest statement. None declared.
determine the most effective dose of the MOs, 2.3 nL of diluted
MO (containing 2, 4 and 6 ng) was injected into cells at one- or
two-cell-stage embryos by using an oil-PicoPump pv280 (World
Funding
Precision Instruments). For in vivo rescue experiments, human This study was supported by grants from Ministry of Science
WT RCBTB1 mRNAs were prepared using the mMESSAGE mMA- and Technology (MOST) (104-2314-B-010-049) and Cheng-Yang
CHINE Kit (Ambion) by following the manufacturer instructions. Collaborative Research Foundation (98F117CY02, 99F167CY05)
The mRNAs (200 pg) were coinjected with MOs as described. After to M.-Y.C.; the High-throughput Genome Analysis and RNAi
injection, the embryos were cultured at 28.5°C in the E3 embryo Core Facilities supported by the National Core Facility Program
medium and were subsequently phenotyped at 3 dpf or 4 dpf for Biotechnology (NSC-103-2319-B-010-001 and NSC100-
through confocal microscopy by using the Leica AF6000 LX. The 2319-B-001-002) for Illumina sequencing and RNAi reagents;
average areas of eight ISVs counted posteriorly from the urogeni- EBV-transformation service of the Resource Center supported
tal pole were analyzed using ImageJ, and thresholds were opti- by the National Research Program for Biopharmaceuticals
mized to enable the imaging signals to cover the ISV area. The (SB3, NSC100-2325-B-080-001). The work was also supported
pixels in the covered area were then counted and normalized to by grants from the National Health Research Institutes, Taiwan
1646 | Human Molecular Genetics, 2016, Vol. 25, No. 8

(MG-104-PP-12 and MG-104-PP-13) and the Ministry of Science 16. Kashani, A.H., Brown, K.T., Chang, E., Drenser, K.A., Capone,
and Technology, Taiwan (MOST103-2311-B-400-001 and A. and Trese, M.T. (2014) Diversity of retinal vascular anomal-
MOST104-2319-B-400-001) to Y.-J.J. ies in patients with familial exudative vitreoretinopathy. Oph-
thalmology, 121, 2220–2227.
17. Poulter, J.A., Ali, M., Gilmour, D.F., Rice, A., Kondo, H., Hayashi,
K., Mackey, D.A., Kearns, L.S., Ruddle, J.B., Craig, J.E. et al. (2010)
References Mutations in TSPAN12 cause autosomal-dominant familial
1. Criswick, V.G. and Schepens, C.L. (1969) Familial exudative exudative vitreoretinopathy. Am. J. Hum. Genet., 86, 248–253.
vitreoretinopathy. Am. J. Ophthalmol., 68, 578–594. 18. Simunovic, M.P. and Maberley, D.A. (2014) Familial exudative

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


2. Canny, C.L. and Oliver, G.L. (1976) Fluorescein angiographic vitreoretinopathy mimicking macular telangiectasia type 1.
findings in familial exudative vitreoretinopathy. Arch. Can. J. Ophthalmol., 49, e28–e30.
Ophthalmol., 94, 1114–1120. 19. Gow, J. and Oliver, G.L. (1971) Familial exudative vitreoretino-
3. Zaremba, J., Feil, S., Juszko, J., Myga, W., van Duijnhoven, G. pathy. An expanded view. Arch. Ophthalmol., 86, 150–155.
and Berger, W. (1998) Intrafamilial variability of the ocular 20. Jiao, X., Ventruto, V., Trese, M.T., Shastry, B.S. and Hejtman-
phenotype in a Polish family with a missense mutation cik, J.F. (2004) Autosomal recessive familial exudative vitreor-
(A63D) in the Norrie disease gene. Ophthalmic Genet., 19, etinopathy is associated with mutations in LRP5. Am. J. Hum.
157–164. Genet., 75, 878–884.
4. Robitaille, J.M., Wallace, K., Zheng, B., Beis, M.J., Samuels, M., 21. van Nouhuys, C.E. (1982) Dominant exudative vitreoretinopa-
Hoskin-Mott, A. and Guernsey, D.L. (2009) Phenotypic overlap thy and other vascular developmental disorders of the per-
of familial exudative vitreoretinopathy (FEVR) with persistent ipheral retina. Doc. Ophthalmol., 54, 1–414.
fetal vasculature (PFV) caused by FZD4 mutations in two dis- 22. Kashani, A.H., Learned, D., Nudleman, E., Drenser, K.A., Ca-
tinct pedigrees. Ophthalmic Genet., 30, 23–30. pone, A. and Trese, M.T. (2014) High prevalence of peripheral
5. Berger, W., Kloeckener-Gruissem, B. and Neidhardt, J. (2010) retinal vascular anomalies in family members of patients
The molecular basis of human retinal and vitreoretinal dis- with familial exudative vitreoretinopathy. Ophthalmology,
eases. Prog. Retin. Eye Res., 29, 335–375. 121, 262–268.
6. Nikopoulos, K., Venselaar, H., Collin, R.W., Riveiro-Alvarez, R., 23. Collin, R.W., Nikopoulos, K., Dona, M., Gilissen, C., Hoischen,
Boonstra, F.N., Hooymans, J.M., Mukhopadhyay, A., Shears, A., Boonstra, F.N., Poulter, J.A., Kondo, H., Berger, W., Toomes,
D., van Bers, M., de Wijs, I.J. et al. (2010) Overview of the mu- C. et al. (2013) ZNF408 is mutated in familial exudative vitreor-
tation spectrum in familial exudative vitreoretinopathy and etinopathy and is crucial for the development of zebrafish
Norrie disease with identification of 21 novel variants in retinal vasculature. Proc. Natl. Acad. Sci. USA, 110, 9856–9861.
FZD4, LRP5, and NDP. Hum. Mutat., 31, 656–666. 24. Nikopoulos, K., Gilissen, C., Hoischen, A., van Nouhuys, C.E.,
7. Robitaille, J.M., Zheng, B., Wallace, K., Beis, M.J., Tatlidil, C., Boonstra, F.N., Blokland, E.A., Arts, P., Wieskamp, N., Strom,
Yang, J., Sheidow, T.G., Siebert, L., Levin, A.V., Lam, W.C. T.M., Ayuso, C. et al. (2010) Next-generation sequencing of a
et al. (2011) The role of Frizzled-4 mutations in familial exuda- 40 Mb linkage interval reveals TSPAN12 mutations in patients
tive vitreoretinopathy and Coats disease. Br. J. Ophthalmol., 95, with familial exudative vitreoretinopathy. Am. J. Hum. Genet.,
574–579. 86, 240–247.
8. Schafer, N.F., Luhmann, U.F., Feil, S. and Berger, W. (2009) Dif- 25. Robitaille, J., MacDonald, M.L., Kaykas, A., Sheldahl, L.C., Zeis-
ferential gene expression in Ndph-knockout mice in retinal ler, J., Dube, M.P., Zhang, L.H., Singaraja, R.R., Guernsey, D.L.,
development. Invest. Ophthalmol. Vis. Sci., 50, 906–916. Zheng, B. et al. (2002) Mutant frizzled-4 disrupts retinal angio-
9. Gilmour, D.F. (2015) Familial exudative vitreoretinopathy and genesis in familial exudative vitreoretinopathy. Nat. Genet.,
related retinopathies. Eye (Lond.), 29, 1–14. 32, 326–330.
10. Gal, M., Levanon, E.Y., Hujeirat, Y., Khayat, M., Pe’er, J. and 26. Toomes, C., Bottomley, H.M., Jackson, R.M., Towns, K.V.,
Shalev, S. (2014) Novel mutation in TSPAN12 leads to auto- Scott, S., Mackey, D.A., Craig, J.E., Jiang, L., Yang, Z., Trembath,
somal recessive inheritance of congenital vitreoretinal dis- R. et al. (2004) Mutations in LRP5 or FZD4 underlie the com-
ease with intra-familial phenotypic variability. Am. J. Med. mon familial exudative vitreoretinopathy locus on chromo-
Genet. A., 164A, 2996–3002. doi: 2910.1002/ajmg.a.36739. some 11q. Am. J. Hum. Genet., 74, 721–730.
Epub 32014 Sep 36722. 27. Junge, H.J., Yang, S., Burton, J.B., Paes, K., Shu, X., French, D.M.,
11. Morris, B., Foot, B. and Mulvihill, A. (2010) A population-based Costa, M., Rice, D.S. and Ye, W. (2009) TSPAN12 regulates ret-
study of Coats disease in the United Kingdom I: epidemiology inal vascular development by promoting Norrin- but not
and clinical features at diagnosis. Eye (Lond), 24, 1797–1801. Wnt-induced FZD4/beta-catenin signaling. Cell, 139, 299–311.
12. Black, G.C., Perveen, R., Bonshek, R., Cahill, M., Clayton- 28. Salvo, J., Lyubasyuk, V., Xu, M., Wang, H., Wang, F., Nguyen,
Smith, J., Lloyd, I.C. and McLeod, D. (1999) Coats’ disease of D., Wang, K., Luo, H., Wen, C., Shi, C. et al. (2015) Next-gener-
the retina (unilateral retinal telangiectasis) caused by somat- ation sequencing and novel variant determination in a cohort
ic mutation in the NDP gene: a role for norrin in retinal angio- of 92 familial exudative vitreoretinopathy patients. Invest.
genesis. Hum. Mol. Genet., 8, 2031–2035. Ophthalmol. Vis. Sci., 56, 1937–1946.
13. Shields, J.A. and Shields, C.L. (2002) Review: Coats disease: the 29. Aponte, E.P., Pulido, J.S., Ellison, J.W., Quiram, P.A. and Moh-
2001 LuEsther T. Mertz lecture. Retina, 22, 80–91. ney, B.G. (2009) A novel NDP mutation in an infant with uni-
14. Smithen, L.M., Brown, G.C., Brucker, A.J., Yannuzzi, L.A., Klais, lateral persistent fetal vasculature and retinal vasculopathy.
C.M. and Spaide, R.F. (2005) Coats’ disease diagnosed in adult- Ophthalmic Genet., 30, 99–102.
hood. Ophthalmology, 112, 1072–1078. 30. Meindl, A., Berger, W., Meitinger, T., van de Pol, D., Achatz, H.,
15. Chen, Z.Y., Battinelli, E.M., Fielder, A., Bundey, S., Sims, K., Dorner, C., Haasemann, M., Hellebrand, H., Gal, A., Cremers,
Breakefield, X.O. and Craig, I.W. (1993) A mutation in the F. et al. (1992) Norrie disease is caused by mutations in an
Norrie disease gene (NDP) associated with X-linked familial extracellular protein resembling C-terminal globular domain
exudative vitreoretinopathy. Nat. Genet., 5, 180–183. of mucins. Nat. Genet., 2, 139–143.
Human Molecular Genetics, 2016, Vol. 25, No. 8 | 1647

31. Lin, P., Shankar, S.P., Duncan, J., Slavotinek, A., Stone, E.M. and 39. Plafker, K.S., Singer, J.D. and Plafker, S.M. (2009) The ubiquitin
Rutar, T. (2010) Retinal vascular abnormalities and dragged conjugating enzyme, UbcM2, engages in novel interactions
maculae in a carrier with a new NDP mutation (c.268delC) that with components of cullin-3 based E3 ligases. Biochemistry,
caused severe Norrie disease in the proband. J. AAPOS, 14, 93–96. 48, 3527–3537.
32. Ng, S.B., Bigham, A.W., Buckingham, K.J., Hannibal, M.C., 40. Mirza, S., Plafker, K.S., Aston, C. and Plafker, S.M. (2010)
McMillin, M.J., Gildersleeve, H.I., Beck, A.E., Tabor, H.K., Cooper, Expression and distribution of the class III ubiquitin-
G.M., Mefford, H.C. et al. (2010) Exome sequencing identifies conjugating enzymes in the retina. Mol. Vis., 16, 2425–2437.
MLL2 mutations as a cause of Kabuki syndrome. Nat. Genet., 41. Izumi, N., Helker, C., Ehling, M., Behrens, A., Herzog, W. and
42, 790–793. Adams, R.H. (2012) Fbxw7 controls angiogenesis by regulating

Downloaded from https://academic.oup.com/hmg/article-abstract/25/8/1637/2384939 by University of Leeds - Library user on 29 July 2019


33. Auton, A., Brooks, L.D., Durbin, R.M., Garrison, E.P., Kang, H.M., endothelial Notch activity. PLoS One, 7, e41116.
Korbel, J.O., Marchini, J.L., McCarthy, S., McVean, G.A. and 42. Kok, F.O., Shin, M., Ni, C.W., Gupta, A., Grosse, A.S., van Impel,
Abecasis, G.R. (2015) A global reference for human genetic A., Kirchmaier, B.C., Peterson-Maduro, J., Kourkoulis, G.,
variation. Nature, 526, 68–74. doi: 10.1038/nature15393. Male, I. et al. (2015) Reverse genetic screening reveals poor
34. Ellertsdottir, E., Lenard, A., Blum, Y., Krudewig, A., Herwig, L., correlation between morpholino-induced and mutant phe-
Affolter, M. and Belting, H.G. (2010) Vascular morphogenesis notypes in zebrafish. Dev. Cell, 32, 97–108.
in the zebrafish embryo. Dev. Biol., 341, 56–65. 43. Coppieters, F., Ascari, G., Karlstetter, M., Bauwens, M.,
35. Mabuchi, H., Fujii, H., Calin, G., Alder, H., Negrini, M., Rassen- De Rocker, N., Boel, A., Vleminckx, K., Van der Eecken, M.,
ti, L., Kipps, T.J., Bullrich, F. and Croce, C.M. (2001) Cloning and Leroy, B.P., Meire, F. et al. (2015) Identification of RCBTB1 as
characterization of CLLD6, CLLD7, and CLLD8, novel candi- novel disease gene for retinal ciliopathy (Program number
date genes for leukemogenesis at chromosome 13q14, a re- 283). Presented at the 65th Annual Meeting of the American
gion commonly deleted in B-cell chronic lymphocytic Society of Human Genetics, October 6–10, 2015, Baltimore,
leukemia. Cancer Res., 61, 2870–2877. MD.
36. Zhou, X. and Munger, K. (2010) Clld7, a candidate tumor sup- 44. Alvarez, Y., Cederlund, M.L., Cottell, D.C., Bill, B.R., Ekker, S.C.,
pressor on chromosome 13q14, regulates pathways of DNA Torres-Vazquez, J., Weinstein, B.M., Hyde, D.R., Vihtelic, T.S.
damage/repair and apoptosis. Cancer Res., 70, 9434–9443. and Kennedy, B.N. (2007) Genetic determinants of
37. Bischoff, F.R. and Ponstingl, H. (1991) Catalysis of guanine hyaloid and retinal vasculature in zebrafish. BMC Dev. Biol.,
nucleotide exchange on Ran by the mitotic regulator RCC1. 7, 114.
Nature, 354, 80–82. 45. Tinnikov, A.A. and Samuels, H.H. (2013) A novel cell lysis ap-
38. Hadjebi, O., Casas-Terradellas, E., Garcia-Gonzalo, F.R. and proach reveals that caspase-2 rapidly translocates from the
Rosa, J.L. (2008) The RCC1 superfamily: from genes, to func- nucleus to the cytoplasm in response to apoptotic stimuli.
tion, to disease. Biochim. Biophys. Acta, 1783, 1467–1479. PLoS One, 8, e61085.

You might also like