Variants in myelin regulatory factor (MYRF) cause autosomal dominant and syndromic nanophthalmos in humans and retinal degeneration in mice

Sarah J Garnai, Michelle L Brinkmeier, Ben Emery, Tomas S Aleman, Louise C Pyle, Biliana Veleva-Rotse, Robert A Sisk, Frank W Rozsa, Ayse Bilge Ozel, Jun Z Li, Sayoko E Moroi, Steven M Archer, Cheng-Mao Lin, Sarah Sheskey, Laurel Wiinikka-Buesser, James Eadie, Jill E Urquhart, Graeme C M Black, Mohammad I Othman, Michael Boehnke, Scot A Sullivan, Gregory L Skuta, Hemant S Pawar, Alexander E Katz, Laryssa A Huryn, Robert B Hufnagel, Genomic Ascertainment Cohort, Sally A Camper, Julia E Richards, Lev Prasov, Sarah J Garnai, Michelle L Brinkmeier, Ben Emery, Tomas S Aleman, Louise C Pyle, Biliana Veleva-Rotse, Robert A Sisk, Frank W Rozsa, Ayse Bilge Ozel, Jun Z Li, Sayoko E Moroi, Steven M Archer, Cheng-Mao Lin, Sarah Sheskey, Laurel Wiinikka-Buesser, James Eadie, Jill E Urquhart, Graeme C M Black, Mohammad I Othman, Michael Boehnke, Scot A Sullivan, Gregory L Skuta, Hemant S Pawar, Alexander E Katz, Laryssa A Huryn, Robert B Hufnagel, Genomic Ascertainment Cohort, Sally A Camper, Julia E Richards, Lev Prasov

Abstract

Nanophthalmos is a rare, potentially devastating eye condition characterized by small eyes with relatively normal anatomy, a high hyperopic refractive error, and frequent association with angle closure glaucoma and vision loss. The condition constitutes the extreme of hyperopia or farsightedness, a common refractive error that is associated with strabismus and amblyopia in children. NNO1 was the first mapped nanophthalmos locus. We used combined pooled exome sequencing and strong linkage data in the large family used to map this locus to identify a canonical splice site alteration upstream of the last exon of the gene encoding myelin regulatory factor (MYRF c.3376-1G>A), a membrane bound transcription factor that undergoes autoproteolytic cleavage for nuclear localization. This variant produced a stable RNA transcript, leading to a frameshift mutation p.Gly1126Valfs*31 in the C-terminus of the protein. In addition, we identified an early truncating MYRF frameshift mutation, c.769dupC (p.S264QfsX74), in a patient with extreme axial hyperopia and syndromic features. Myrf conditional knockout mice (CKO) developed depigmentation of the retinal pigment epithelium (RPE) and retinal degeneration supporting a role of this gene in retinal and RPE development. Furthermore, we demonstrated the reduced expression of Tmem98, another known nanophthalmos gene, in Myrf CKO mice, and the physical interaction of MYRF with TMEM98. Our study establishes MYRF as a nanophthalmos gene and uncovers a new pathway for eye growth and development.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Clinical features of individuals in…
Fig 1. Clinical features of individuals in this study.
(A-G) NNO1 family member clinical imaging. (A) Ultrasound biomicroscopy showing shallow anterior chamber and narrow angles. (B-C) Optic disc photos of the right (B) and left (C) eye showing crowded discs with vascular tortuosity. (D-E) Wide-field 200-degree Optos autofluorescence images of right (D) and left (E) showing tortuous vasculature and highlighting small area of hyperfluorescence in the left eye below the inferiotemporal arcade. (F-G) SD-OCT images of right (F) and left (G) eye showing choroidal folds in the right eye and otherwise normal foveal structure. (H-J) Sporadic nanophthalmos case clinical images. (H-I) SD-OCT of right (H) and left eye (I) eye showing mild foveal hypoplasia. (J) Bscan ultrasound showing short axial length and reduced posterior segment dimensions (line). Scale bar, 200 μm.
Fig 2. Haplotype analysis and fine mapping…
Fig 2. Haplotype analysis and fine mapping of NNO1 interval.
(A) Updated NNO1 pedigree highlighting 4 individuals with dextrocardia (*) from different branches of the family. Blue box denotes subset of family that was subsequently chosen for segregation analysis. Black boxes indicate key regions of the pedigree with recombination events. (B) Haplotype analysis of key recombinants in the NNO1 pedigree showing minimal recombinant interval between D11S4191 and D11S1883 (highlighted in red). Disease haplotype is outlined with red boxes, and one example of shared non-disease haplotype is highlighted in green. Italics denote a deduced haplotype. (C) Genomic region encompassed by the non-recombinant interval on chromosome 11q (MYRF, ZP1, PPP1R32, and BEST1 location with the interval are highlighted in red).
Fig 3. The MYRF c.3376-1G>A variant co-segregates…
Fig 3. The MYRF c.3376-1G>A variant co-segregates with the nanophthalmos within the NNO1 family.
(A) Schematic diagram of MYRF protein and functional domains. (B) Schematic and agarose gel electrophoresis for StyI restriction digest used to confirm variant segregation in NNO1 family within one large nuclear family branch. (C) Sequence of normal and variant MYRF with splice acceptor site (underlined) and predicted amino acids. Sequencing chromatograms confirming heterozygous c.3376-1G>A mutation. Pro-rich, proline-rich domain; nls, nuclear localization sequence; DBD, DNA binding domain; ICD, intramolecular chaperone domain; TM, transmembrane domain; CTD, C-terminal domain.
Fig 4. MYRF c.3376-1G>A variant disrupts mRNA…
Fig 4. MYRF c.3376-1G>A variant disrupts mRNA splicing and produces a stable RNA species.
(A) Minigene splicing assay. Schematic diagram showing the design of the minigene assay, and sample chromatograms showing the wild-type and mutant cDNA species isolated from this assay. A single splice product was generated for each form, wild-type (WT) and mutant (Mut) RNA. The splice product for the mutant form uses a splice acceptor site one base pair downstream of the original splice site and creates a frameshift (see Fig 3C). (B) RT-PCR from individual IV-22 blood RNA. Schematic diagram of the experiment is shown with sequence chromatograms from representative cloned RT-PCR products demonstrated. The mutant clones comprised 42% (3/7) of the spliced RNA species. (C) Predicted effect of splice site mutation on C-terminal amino acid sequence, showing frameshift and replacement of the final 26 amino acids with 30 different amino acids. A putative glycosylation site is marked with a *.
Fig 5. MYRF is expressed in the…
Fig 5. MYRF is expressed in the RPE and loss of MYRF in mouse leads to pigmentary change in the mouse RPE.
(A) qRT-PCR showing relative expression of MYRF in human ocular and adnexal tissue. MYRF expression is normalized to GAPDH and reported as expression level relative to extraocular muscle. (B-C) Low-magnification (B) and high magnification (C) RPE flatmounts from P22 control (Myrf+/fl or Myrffl/fl) and Myrf heterozygous and homozygous conditional knockout mice (RxCre;Myrf+/fl and RxCre;Myrffl/fl) showing patchy areas of hypopigmentation in the RxCre;Myrffl/fl mice. (D-F) H&E histology of RPE from the above mice at E15.5 (D), P3 (E), P14 (F), and P22 (G) showing early loss of RPE pigmentation (D) in RxCre;Myrffl/fl mice, which persists after retinal histogenesis is complete (G). TM, trabecular meshwork; CB, ciliary body, Chor, choroid; PPS, peripapillary sclera; ONH, optic nerve head; PLON, post-laminar optic nerve. Scale bar, 500 μm in B, 100 μm in C, 50 μm in D, 25 μm in E.
Fig 6. Loss of MYRF leads to…
Fig 6. Loss of MYRF leads to early retinal degeneration in mice.
(A-C) H&E histology of P3 (A), P14 (B), and P22 (C) control (Myrf+/fl or Myrffl/fl) or Myrf heterozygous and homozygous conditional knockout mice (RxCre;Myrf+/fl and RxCre;Myrffl/fl). RxCre;Myrffl/fl retinas have shortened inner and outer segments, but retinal structure during development is preserved. RxCre;Myrf+/fl are structurally indistinguishable from control. (D-E) Low magnification (D) and high magnification (E) images of photoreceptor immunolabeling in P22 animals. Mouse cone arrestin (mCar, green) and rhodopsin (Rho, red) and DAPI (blue) were used to mark cones, rods, and nuclei, respectively. (F) Quantitative analysis of inner/outer segment area compared to total retinal area. (G) Quantitative analysis of cone fraction compared to fraction of total retinal cells. There is a significant decrease in IS/OS area and cone fraction in RxCre;Myrffl/fl retinas. n = 8 eyes, 6 animals (control); n = 6 eyes, 3 animals (RxCre;Myrf+/fl), n = 4 eyes, 4 animals (RxCre;Myrffl/fl). Mean±standard deviation are plotted along with each individual eye data point. **, p<0.01. Scale bar, 50μm.
Fig 7. Loss of MYRF leads to…
Fig 7. Loss of MYRF leads to global retinal dysfunction and patchy outer retinal atrophy.
(A-C) Representative central color fundus photos (A) and central (B) and peripheral (C) SD-OCT from 10-month old eyes from control (Myrffl/fl or Myrf+/fl), RxCre;Myrf+/fl and RxCre;Myrffl/fl mice. Patchy areas of atrophy with RPE pigment changes are seen most prominently in RxCre; Myrffl/fl eyes (A), and correspond to outer retinal and RPE loss on OCT (C) with relatively preserved peripapillary retina (B). (D-I) Electroretinography of 10-month old control (Myrffl/fl or Myrf+/fl), RxCre;Myrf+/fl and RxCre;Myrffl/fl mice. (D-F) Representative electroretinogram traces from 10-month old mice under scotopic (D), photopic (E), or photopic 9.9 Hz flicker conditions with the noted intensity stimuli. Both scotopic and photopic responses are diminished in RxCre;Myrffl/fl eyes compared to controls. (G-I) Scotopic a-wave (G), scotopic B-wave, and photopic B-wave amplitudes and comparison statistics across varying intensity stimuli. Error bars indicate standard deviation and are noted by the shaded grey for the control group. Summary statistics for comparisons of RxCre;Myrf+/fl and RxCre;Myrffl/fl to control eyes are shown with stars. In RxCre;Myrffl/fl, scotopic a-wave ERG amplitudes are reduced by 45–50% and B-wave amplitudes are reduced 35–45%, while photopic B-wave amplitudes are reduced 50–60%. Control n = 4 eyes from 2 animals, RxCre;Myrf+/fl n = 4 eyes from 2 animals, RxCre;Myrffl/fl n = 12 eyes from 6 animals. Mean±standard deviation. ***, p < 0.001; **, p < 0.01; * p < 0.05.
Fig 8. TMEM98 is genetically downstream of…
Fig 8. TMEM98 is genetically downstream of MYRF.
(A-B) Taqman qRT-PCR analysis of RPE expressed genes important for nanophthalmos (Myrf, Best1, Mfrp, Tmem98) in Myrf+/fl, RxCre;Myrf+/fl and RxCre;Myrffl/fl eyes at P3 (A) and P22 (B). There is significantly reduced expression of Tmem98 in RxCre;Myrffl/fl compared to wild-type, but comparable levels of expression of Mfrp and Best1. (C) TMEM98 staining in embryonic (E15.5) mouse eyes showing loss of TMEM98 staining in RxCre;Myrffl/fl compared to controls. (D-E) Mouse RPE flat mounts from P7 (D) and P22 (E) eyes showing decreased TMEM98 staining (red) and altered localization in RxCre;Myrffl/fl compared to controls. DAPI (blue) is used to counterstain nuclei. ***, p < 0.001; **, p < 0.01; * p < 0.05.
Fig 9. MYRF physically interacts with TMEM98.
Fig 9. MYRF physically interacts with TMEM98.
(A) Schematic diagram of N-terminal Myc-tagged and C-terminal HA-tagged MYRF constructs used for co-immunoprecipitation experiments. Below each construct, the observed cleavage products are shown. Note that only the N-terminal product can be detected with anti-Myc, and the C-terminal product with anti-FLAG, and all constructs are expected to be cleaved except the one corresponding to the already-cleaved C-terminal cleavage product. (B) Co-immunoprecipitation (co-IP) experiments with HA-tagged TMEM98 and Myc-tagged MYRF constructs in HEK293T cells. Extracts from HEK293T cells were transfected with the indicated constructs, and then immunoprecipitated with anti-HA (top) or anti-Myc (bottom). Western blots against Myc or HA are shown for each immunoprecipitation experiment. HA-TMEM98 immunoprecipitates with either full-length Myc-MYRF, the uncleaved form of a Myc-MYRF1-846 construct or a Myc-C-terminal-cleavage-product (aa587-1139), mapping the region of interaction to MYRF587-846. (C) Western blots showing stabilization of uncleaved MYRF in a dose dependent manner by increasing levels of TMEM98. Extracts from HEK293T cells were transfected with myc-MYRF-FLAG and increasing doses of HA-TMEM98, and subsequently blotted against C-terminal FLAG tag (MYRF), N-terminal HA tag (TMEM98), and loading control β-III tubulin (TUBB3). MYRF, full-length MYRF; CTCP, C-terminal cleavage product; aa1-846, MYRF truncated construct (amino acids 1–846); ** corresponds to full-length MYRF band, * corresponds to cleavage product band (N-terminal in B, C-terminal in C). The N-terminal and C-terminal cleavage products run at a similar size due to post-translational modification.
Fig 10. Model of MYRF and TMEM98…
Fig 10. Model of MYRF and TMEM98 function.
MYRF interacts with TMEM98 and other factors, as yet unidentified, and upon cleavage activates transcription of specific downstream genes, including TMEM98, in various tissues including RPE. MYRF domains including the N-terminal (yellow), intramolecular chaperone (green), and C-terminal (blue) domains are highlighted.

References

    1. Flaxman SR, Bourne RRA, Resnikoff S, Ackland P, Braithwaite T, et al. (2017) Global causes of blindness and distance vision impairment 1990–2020: a systematic review and meta-analysis. Lancet Glob Health 5: e1221–e1234. 10.1016/S2214-109X(17)30393-5
    1. Day AC, Khawaja AP, Peto T, Hayat S, Luben R, et al. (2013) The small eye phenotype in the EPIC-Norfolk eye study: prevalence and visual impairment in microphthalmos and nanophthalmos. BMJ Open 3.
    1. Hoffman RS, Vasavada AR, Allen QB, Snyder ME, Devgan U, et al. (2015) Cataract surgery in the small eye. J Cataract Refract Surg 41: 2565–2575. 10.1016/j.jcrs.2015.10.008
    1. Carricondo PC, Andrade T, Prasov L, Ayres BM, Moroi SE (2018) Nanophthalmos: A Review of the Clinical Spectrum and Genetics. J Ophthalmol 2018: 2735465 10.1155/2018/2735465
    1. Hendriks M, Verhoeven VJM, Buitendijk GHS, Polling JR, Meester-Smoor MA, et al. (2017) Development of Refractive Errors-What Can We Learn From Inherited Retinal Dystrophies? Am J Ophthalmol 182: 81–89. 10.1016/j.ajo.2017.07.008
    1. Aleman TS, Uyhazi KE, Serrano LW, Vasireddy V, Bowman SJ, et al. (2018) RDH12 Mutations Cause a Severe Retinal Degeneration With Relatively Spared Rod Function. Invest Ophthalmol Vis Sci 59: 5225–5236. 10.1167/iovs.18-24708
    1. Aleman TS, Cideciyan AV, Aguirre GK, Huang WC, Mullins CL, et al. (2011) Human CRB1-associated retinal degeneration: comparison with the rd8 Crb1-mutant mouse model. Invest Ophthalmol Vis Sci 52: 6898–6910. 10.1167/iovs.11-7701
    1. Dinculescu A, Estreicher J, Zenteno JC, Aleman TS, Schwartz SB, et al. (2012) Gene therapy for retinitis pigmentosa caused by MFRP mutations: human phenotype and preliminary proof of concept. Hum Gene Ther 23: 367–376. 10.1089/hum.2011.169
    1. Garafalo AV, Calzetti G, Cideciyan AV, Roman AJ, Saxena S, et al. (2018) Cone Vision Changes in the Enhanced S-Cone Syndrome Caused by NR2E3 Gene Mutations. Invest Ophthalmol Vis Sci 59: 3209–3219. 10.1167/iovs.18-24518
    1. Russell-Eggitt IM, Clayton PT, Coffey R, Kriss A, Taylor DS, et al. (1998) Alstrom syndrome. Report of 22 cases and literature review. Ophthalmology 105: 1274–1280. 10.1016/S0161-6420(98)97033-6
    1. Cideciyan AV, Rachel RA, Aleman TS, Swider M, Schwartz SB, et al. (2011) Cone photoreceptors are the main targets for gene therapy of NPHP5 (IQCB1) or NPHP6 (CEP290) blindness: generation of an all-cone Nphp6 hypomorph mouse that mimics the human retinal ciliopathy. Hum Mol Genet 20: 1411–1423. 10.1093/hmg/ddr022
    1. Hammond CJ, Snieder H, Gilbert CE, Spector TD (2001) Genes and environment in refractive error: the twin eye study. Invest Ophthalmol Vis Sci 42: 1232–1236.
    1. Klein AP, Suktitipat B, Duggal P, Lee KE, Klein R, et al. (2009) Heritability analysis of spherical equivalent, axial length, corneal curvature, and anterior chamber depth in the Beaver Dam Eye Study. Arch Ophthalmol 127: 649–655. 10.1001/archophthalmol.2009.61
    1. Xu Y, Guan L, Xiao X, Zhang J, Li S, et al. (2016) Identification of MFRP Mutations in Chinese Families with High Hyperopia. Optom Vis Sci 93: 19–26. 10.1097/OPX.0000000000000751
    1. Jiang D, Yang Z, Li S, Xiao X, Jia X, et al. (2013) Evaluation of PRSS56 in Chinese subjects with high hyperopia or primary angle-closure glaucoma. Mol Vis 19: 2217–2226.
    1. Nair KS, Hmani-Aifa M, Ali Z, Kearney AL, Ben Salem S, et al. (2011) Alteration of the serine protease PRSS56 causes angle-closure glaucoma in mice and posterior microphthalmia in humans and mice. Nat Genet 43: 579–584. 10.1038/ng.813
    1. Mehalow AK, Kameya S, Smith RS, Hawes NL, Denegre JM, et al. (2003) CRB1 is essential for external limiting membrane integrity and photoreceptor morphogenesis in the mammalian retina. Hum Mol Genet 12: 2179–2189. 10.1093/hmg/ddg232
    1. Paylakhi S, Labelle-Dumais C, Tolman NG, Sellarole MA, Seymens Y, et al. (2018) Muller glia-derived PRSS56 is required to sustain ocular axial growth and prevent refractive error. PLoS Genet 14: e1007244 10.1371/journal.pgen.1007244
    1. Marmorstein AD, Marmorstein LY, Rayborn M, Wang X, Hollyfield JG, et al. (2000) Bestrophin, the product of the Best vitelliform macular dystrophy gene (VMD2), localizes to the basolateral plasma membrane of the retinal pigment epithelium. Proc Natl Acad Sci U S A 97: 12758–12763. 10.1073/pnas.220402097
    1. Kameya S, Hawes NL, Chang B, Heckenlively JR, Naggert JK, et al. (2002) Mfrp, a gene encoding a frizzled related protein, is mutated in the mouse retinal degeneration 6. Hum Mol Genet 11: 1879–1886.
    1. Sundin OH (2005) The mouse's eye and Mfrp: not quite human. Ophthalmic Genet 26: 153–155. 10.1080/13816810500374359
    1. Ayala-Ramirez R, Graue-Wiechers F, Robredo V, Amato-Almanza M, Horta-Diez I, et al. (2006) A new autosomal recessive syndrome consisting of posterior microphthalmos, retinitis pigmentosa, foveoschisis, and optic disc drusen is caused by a MFRP gene mutation. Mol Vis 12: 1483–1489.
    1. Crespi J, Buil JA, Bassaganyas F, Vela-Segarra JI, Diaz-Cascajosa J, et al. (2008) A novel mutation confirms MFRP as the gene causing the syndrome of nanophthalmos-renititis pigmentosa-foveoschisis-optic disk drusen. Am J Ophthalmol 146: 323–328. 10.1016/j.ajo.2008.04.029
    1. Yardley J, Leroy BP, Hart-Holden N, Lafaut BA, Loeys B, et al. (2004) Mutations of VMD2 splicing regulators cause nanophthalmos and autosomal dominant vitreoretinochoroidopathy (ADVIRC). Invest Ophthalmol Vis Sci 45: 3683–3689. 10.1167/iovs.04-0550
    1. Zenteno JC, Buentello-Volante B, Ayala-Ramirez R, Villanueva-Mendoza C (2011) Homozygosity mapping identifies the Crumbs homologue 1 (Crb1) gene as responsible for a recessive syndrome of retinitis pigmentosa and nanophthalmos. Am J Med Genet A 155A: 1001–1006. 10.1002/ajmg.a.33862
    1. Guziewicz KE, Zangerl B, Lindauer SJ, Mullins RF, Sandmeyer LS, et al. (2007) Bestrophin gene mutations cause canine multifocal retinopathy: a novel animal model for best disease. Invest Ophthalmol Vis Sci 48: 1959–1967. 10.1167/iovs.06-1374
    1. Koenning M, Jackson S, Hay CM, Faux C, Kilpatrick TJ, et al. (2012) Myelin gene regulatory factor is required for maintenance of myelin and mature oligodendrocyte identity in the adult CNS. J Neurosci 32: 12528–12542. 10.1523/JNEUROSCI.1069-12.2012
    1. Bujalka H, Koenning M, Jackson S, Perreau VM, Pope B, et al. (2013) MYRF is a membrane-associated transcription factor that autoproteolytically cleaves to directly activate myelin genes. PLoS Biol 11: e1001625 10.1371/journal.pbio.1001625
    1. Li Z, Park Y, Marcotte EM (2013) A Bacteriophage tailspike domain promotes self-cleavage of a human membrane-bound transcription factor, the myelin regulatory factor MYRF. PLoS Biol 11: e1001624 10.1371/journal.pbio.1001624
    1. Emery B, Agalliu D, Cahoy JD, Watkins TA, Dugas JC, et al. (2009) Myelin gene regulatory factor is a critical transcriptional regulator required for CNS myelination. Cell 138: 172–185. 10.1016/j.cell.2009.04.031
    1. Xiao L, Ohayon D, McKenzie IA, Sinclair-Wilson A, Wright JL, et al. (2016) Rapid production of new oligodendrocytes is required in the earliest stages of motor-skill learning. Nat Neurosci 19: 1210–1217. 10.1038/nn.4351
    1. Duncan GJ, Plemel JR, Assinck P, Manesh SB, Muir FGW, et al. (2017) Myelin regulatory factor drives remyelination in multiple sclerosis. Acta Neuropathol 134: 403–422. 10.1007/s00401-017-1741-7
    1. Pinz H, Pyle LC, Li D, Izumi K, Skraban C, et al. (2018) De novo variants in Myelin regulatory factor (MYRF) as candidates of a new syndrome of cardiac and urogenital anomalies. Am J Med Genet A 176: 969–972. 10.1002/ajmg.a.38620
    1. Chitayat D, Shannon P, Uster T, Nezarati MM, Schnur RE, et al. (2018) An Additional Individual with a De Novo Variant in Myelin Regulatory Factor (MYRF) with Cardiac and Urogenital Anomalies: Further Proof of Causality: Comments on the article by Pinz et al. (). Am J Med Genet A 176: 2041–2043. 10.1002/ajmg.a.40360
    1. Kurahashi H, Azuma Y, Masuda A, Okuno T, Nakahara E, et al. (2018) MYRF is associated with encephalopathy with reversible myelin vacuolization. Ann Neurol 83: 98–106. 10.1002/ana.25125
    1. Qi H, Yu L, Zhou X, Wynn J, Zhao H, et al. (2018) De novo variants in congenital diaphragmatic hernia identify MYRF as a new syndrome and reveal genetic overlaps with other developmental disorders. PLoS Genet 14: e1007822 10.1371/journal.pgen.1007822
    1. Othman MI, Sullivan SA, Skuta GL, Cockrell DA, Stringham HM, et al. (1998) Autosomal dominant nanophthalmos (NNO1) with high hyperopia and angle-closure glaucoma maps to chromosome 11. Am J Hum Genet 63: 1411–1418. 10.1086/302113
    1. Evans WN, Acherman RJ, Collazos JC, Castillo WJ, Rollins RC, et al. (2010) Dextrocardia: practical clinical points and comments on terminology. Pediatr Cardiol 31: 1–6. 10.1007/s00246-009-9516-0
    1. Marquardt A, Stohr H, Passmore LA, Kramer F, Rivera A, et al. (1998) Mutations in a novel gene, VMD2, encoding a protein of unknown properties cause juvenile-onset vitelliform macular dystrophy (Best's disease). Hum Mol Genet 7: 1517–1525.
    1. Kramer F, White K, Pauleikhoff D, Gehrig A, Passmore L, et al. (2000) Mutations in the VMD2 gene are associated with juvenile-onset vitelliform macular dystrophy (Best disease) and adult vitelliform macular dystrophy but not age-related macular degeneration. Eur J Hum Genet 8: 286–292. 10.1038/sj.ejhg.5200447
    1. Burgess R, Millar ID, Leroy BP, Urquhart JE, Fearon IM, et al. (2008) Biallelic mutation of BEST1 causes a distinct retinopathy in humans. Am J Hum Genet 82: 19–31. 10.1016/j.ajhg.2007.08.004
    1. Testa F, Rossi S, Passerini I, Sodi A, Di Iorio V, et al. (2008) A normal electro-oculography in a family affected by best disease with a novel spontaneous mutation of the BEST1 gene. Br J Ophthalmol 92: 1467–1470. 10.1136/bjo.2008.143776
    1. Boon CJ, Klevering BJ, Leroy BP, Hoyng CB, Keunen JE, et al. (2009) The spectrum of ocular phenotypes caused by mutations in the BEST1 gene. Prog Retin Eye Res 28: 187–205. 10.1016/j.preteyeres.2009.04.002
    1. Burgess R, MacLaren RE, Davidson AE, Urquhart JE, Holder GE, et al. (2009) ADVIRC is caused by distinct mutations in BEST1 that alter pre-mRNA splicing. J Med Genet 46: 620–625. 10.1136/jmg.2008.059881
    1. Toto L, Boon CJ, Di Antonio L, Battaglia Parodi M, Mastropasqua R, et al. (2016) BESTROPHINOPATHY: A Spectrum of Ocular Abnormalities Caused by the c.614T>C Mutation in the BEST1 Gene. Retina 36: 1586–1595. 10.1097/IAE.0000000000000950
    1. Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, et al. (2016) Analysis of protein-coding genetic variation in 60,706 humans. Nature 536: 285–291. 10.1038/nature19057
    1. Koscielny G, Yaikhom G, Iyer V, Meehan TF, Morgan H, et al. (2014) The International Mouse Phenotyping Consortium Web Portal, a unified point of access for knockout mice and related phenotyping data. Nucleic Acids Res 42: D802–809. 10.1093/nar/gkt977
    1. Kircher M, Witten DM, Jain P, O'Roak BJ, Cooper GM, et al. (2014) A general framework for estimating the relative pathogenicity of human genetic variants. Nat Genet 46: 310–315. 10.1038/ng.2892
    1. Thomas MG, Kumar A, Mohammad S, Proudlock FA, Engle EC, et al. (2011) Structural grading of foveal hypoplasia using spectral-domain optical coherence tomography a predictor of visual acuity? Ophthalmology 118: 1653–1660. 10.1016/j.ophtha.2011.01.028
    1. Biesecker LG, Mullikin JC, Facio FM, Turner C, Cherukuri PF, et al. (2009) The ClinSeq Project: piloting large-scale genome sequencing for research in genomic medicine. Genome Res 19: 1665–1674. 10.1101/gr.092841.109
    1. Guidugli L, Carreira A, Caputo SM, Ehlen A, Galli A, et al. (2014) Functional assays for analysis of variants of uncertain significance in BRCA2. Hum Mutat 35: 151–164. 10.1002/humu.22478
    1. Kim D, Choi JO, Fan C, Shearer RS, Sharif M, et al. (2017) Homo-trimerization is essential for the transcription factor function of Myrf for oligodendrocyte differentiation. Nucleic Acids Res 45: 5112–5125. 10.1093/nar/gkx080
    1. Wang M, Marin A (2006) Characterization and prediction of alternative splice sites. Gene 366: 219–227. 10.1016/j.gene.2005.07.015
    1. Turner DL, Weintraub H (1994) Expression of achaete-scute homolog 3 in Xenopus embryos converts ectodermal cells to a neural fate. Genes Dev 8: 1434–1447.
    1. Bryan JM, Fufa TD, Bharti K, Brooks BP, Hufnagel RB, et al. (2018) Identifying core biological processes distinguishing human eye tissues with precise systems-level gene expression analyses and weighted correlation networks. Hum Mol Genet 27: 3325–3339. 10.1093/hmg/ddy239
    1. Hornig J, Frob F, Vogl MR, Hermans-Borgmeyer I, Tamm ER, et al. (2013) The transcription factors Sox10 and Myrf define an essential regulatory network module in differentiating oligodendrocytes. PLoS Genet 9: e1003907 10.1371/journal.pgen.1003907
    1. Swindell EC, Bailey TJ, Loosli F, Liu C, Amaya-Manzanares F, et al. (2006) Rx-Cre, a tool for inactivation of gene expression in the developing retina. Genesis 44: 361–363. 10.1002/dvg.20225
    1. Srinivas S, Watanabe T, Lin CS, William CM, Tanabe Y, et al. (2001) Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev Biol 1: 4 10.1186/1471-213X-1-4
    1. Choi JO, Fan C, Kim D, Sharif M, An H, et al. (2018) Elucidating the transactivation domain of the pleiotropic transcription factor Myrf. Sci Rep 8: 13075 10.1038/s41598-018-31477-4
    1. Iwanami M, Hiromi Y, Okabe M (2005) Cell-type specific utilization of multiple negative feedback loops generates developmental constancy. Genes Cells 10: 743–752. 10.1111/j.1365-2443.2005.00871.x
    1. Genomes Project C, Auton A, Brooks LD, Durbin RM, Garrison EP, et al. (2015) A global reference for human genetic variation. Nature 526: 68–74. 10.1038/nature15393
    1. Sherry ST, Ward MH, Kholodov M, Baker J, Phan L, et al. (2001) dbSNP: the NCBI database of genetic variation. Nucleic Acids Res 29: 308–311.
    1. Khorram D, Choi M, Roos BR, Stone EM, Kopel T, et al. (2015) Novel TMEM98 mutations in pedigrees with autosomal dominant nanophthalmos. Mol Vis 21: 1017–1023.
    1. Awadalla MS, Burdon KP, Souzeau E, Landers J, Hewitt AW, et al. (2014) Mutation in TMEM98 in a large white kindred with autosomal dominant nanophthalmos linked to 17p12-q12. JAMA Ophthalmol 132: 970–977. 10.1001/jamaophthalmol.2014.946
    1. Senoo H, Araki T, Fukuzawa M, Williams JG (2013) A new kind of membrane-tethered eukaryotic transcription factor that shares an auto-proteolytic processing mechanism with bacteriophage tail-spike proteins. J Cell Sci 126: 5247–5258. 10.1242/jcs.133231
    1. Muth KN, Piefke S, Weider M, Sock E, Hermans-Borgmeyer I, et al. (2016) The Dual-specificity phosphatase Dusp15 is regulated by Sox10 and Myrf in Myelinating Oligodendrocytes. Glia 64: 2120–2132. 10.1002/glia.23044
    1. Siddiqui FM, Rubio ER, Patel VM, Aziz S, Ie S (2016) Not all right-sided hearts are the same-the importance of identifying the correct diagnosis. Ann Transl Med 4: 198 10.21037/atm.2016.05.29
    1. Tongue AC (1987) Refractive errors in children. Pediatr Clin North Am 34: 1425–1437.
    1. Wallace DK, Chandler DL, Beck RW, Arnold RW, Bacal DA, et al. (2007) Treatment of bilateral refractive amblyopia in children three to less than 10 years of age. Am J Ophthalmol 144: 487–496. 10.1016/j.ajo.2007.05.040
    1. Jones-Jordan L, Wang X, Scherer RW, Mutti DO (2014) Spectacle correction versus no spectacles for prevention of strabismus in hyperopic children. Cochrane Database Syst Rev: CD007738 10.1002/14651858.CD007738.pub2
    1. Relhan N, Jalali S, Pehre N, Rao HL, Manusani U, et al. (2016) High-hyperopia database, part I: clinical characterisation including morphometric (biometric) differentiation of posterior microphthalmos from nanophthalmos. Eye (Lond) 30: 120–126.
    1. Ritch R, Chang BM, Liebmann JM (2003) Angle closure in younger patients. Ophthalmology 110: 1880–1889. 10.1016/S0161-6420(03)00563-3
    1. Emanuel ME, Parrish RK 2nd, Gedde SJ (2014) Evidence-based management of primary angle closure glaucoma. Curr Opin Ophthalmol 25: 89–92. 10.1097/ICU.0000000000000028
    1. Kahrom M, Kahrom H (2009) Scimitar syndrome and evolution of managements. Pan Afr Med J 3: 20
    1. Huang H, Teng P, Du J, Meng J, Hu X, et al. (2018) Interactive repression of MYRF self-cleavage and activity in oligodendrocyte differentiation by TMEM98 protein. J Neurosci.
    1. Chen H, Lin H, Lin Z, Chen J, Chen W (2016) Distribution of axial length, anterior chamber depth, and corneal curvature in an aged population in South China. BMC Ophthalmol 16: 47 10.1186/s12886-016-0221-5
    1. Fotedar R, Wang JJ, Burlutsky G, Morgan IG, Rose K, et al. (2010) Distribution of axial length and ocular biometry measured using partial coherence laser interferometry (IOL Master) in an older white population. Ophthalmology 117: 417–423. 10.1016/j.ophtha.2009.07.028
    1. Wang L, Povazay B, Chen YP, Hofer B, Drexler W, et al. (2011) Heritability of ocular component dimensions in mice phenotyped using depth-enhanced swept source optical coherence tomography. Exp Eye Res 93: 482–490. 10.1016/j.exer.2011.06.008
    1. Sundin OH, Leppert GS, Silva ED, Yang JM, Dharmaraj S, et al. (2005) Extreme hyperopia is the result of null mutations in MFRP, which encodes a Frizzled-related protein. Proc Natl Acad Sci U S A 102: 9553–9558. 10.1073/pnas.0501451102
    1. Marmorstein AD, Cross HE, Peachey NS (2009) Functional roles of bestrophins in ocular epithelia. Prog Retin Eye Res 28: 206–226. 10.1016/j.preteyeres.2009.04.004
    1. Marmorstein LY, Wu J, McLaughlin P, Yocom J, Karl MO, et al. (2006) The light peak of the electroretinogram is dependent on voltage-gated calcium channels and antagonized by bestrophin (best-1). J Gen Physiol 127: 577–589. 10.1085/jgp.200509473
    1. Cross SH, Mckie L, Keighren M, West K, Thaung C, et al. (2019) Missense Mutations in the Human Nanophthalmos Gene TMEM98 Cause Retinal Defects in the Mouse. bioRxiv: 513846.
    1. Velez G, Tsang SH, Tsai YT, Hsu CW, Gore A, et al. (2017) Gene Therapy Restores Mfrp and Corrects Axial Eye Length. Sci Rep 7: 16151 10.1038/s41598-017-16275-8
    1. Gal A, Rau I, El Matri L, Kreienkamp HJ, Fehr S, et al. (2011) Autosomal-recessive posterior microphthalmos is caused by mutations in PRSS56, a gene encoding a trypsin-like serine protease. Am J Hum Genet 88: 382–390. 10.1016/j.ajhg.2011.02.006
    1. Sundin OH, Dharmaraj S, Bhutto IA, Hasegawa T, McLeod DS, et al. (2008) Developmental basis of nanophthalmos: MFRP Is required for both prenatal ocular growth and postnatal emmetropization. Ophthalmic Genet 29: 1–9. 10.1080/13816810701651241
    1. Chakraborty R, Park HN, Tan CC, Weiss P, Prunty MC, et al. (2017) Association of Body Length with Ocular Parameters in Mice. Optom Vis Sci 94: 387–394. 10.1097/OPX.0000000000001036
    1. Jeon CJ, Strettoi E, Masland RH (1998) The major cell populations of the mouse retina. J Neurosci 18: 8936–8946.
    1. Schmucker C, Schaeffel F (2004) A paraxial schematic eye model for the growing C57BL/6 mouse. Vision Res 44: 1857–1867. 10.1016/j.visres.2004.03.011
    1. Park H, Qazi Y, Tan C, Jabbar SB, Cao Y, et al. (2012) Assessment of axial length measurements in mouse eyes. Optom Vis Sci 89: 296–303. 10.1097/OPX.0b013e31824529e5
    1. Teare MD, Santibanez Koref MF (2014) Linkage analysis and the study of Mendelian disease in the era of whole exome and genome sequencing. Brief Funct Genomics 13: 378–383. 10.1093/bfgp/elu024
    1. Golan D, Erlich Y, Rosset S (2012) Weighted pooling—practical and cost-effective techniques for pooled high-throughput sequencing. Bioinformatics 28: i197–206. 10.1093/bioinformatics/bts208
    1. Bamshad MJ, Ng SB, Bigham AW, Tabor HK, Emond MJ, et al. (2011) Exome sequencing as a tool for Mendelian disease gene discovery. Nat Rev Genet 12: 745–755. 10.1038/nrg3031
    1. Kalia SS, Adelman K, Bale SJ, Chung WK, Eng C, et al. (2017) Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics. Genet Med 19: 249–255. 10.1038/gim.2016.190
    1. Weiner C (2014) Anticipate and communicate: Ethical management of incidental and secondary findings in the clinical, research, and direct-to-consumer contexts (December 2013 report of the Presidential Commission for the Study of Bioethical Issues). Am J Epidemiol 180: 562–564. 10.1093/aje/kwu217
    1. Wu W, Dawson DG, Sugar A, Elner SG, Meyer KA, et al. (2004) Cataract surgery in patients with nanophthalmos: results and complications. J Cataract Refract Surg 30: 584–590. 10.1016/j.jcrs.2003.07.009
    1. Li H, Durbin R (2009) Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25: 1754–1760. 10.1093/bioinformatics/btp324
    1. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, et al. (2010) The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 20: 1297–1303. 10.1101/gr.107524.110
    1. Danecek P, Auton A, Abecasis G, Albers CA, Banks E, et al. (2011) The variant call format and VCFtools. Bioinformatics 27: 2156–2158. 10.1093/bioinformatics/btr330
    1. Ng SB, Turner EH, Robertson PD, Flygare SD, Bigham AW, et al. (2009) Targeted capture and massively parallel sequencing of 12 human exomes. Nature 461: 272–276. 10.1038/nature08250
    1. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408. 10.1006/meth.2001.1262
    1. Rashid K, Dannhausen K, Langmann T (2019) Testing for Known Retinal Degeneration Mutants in Mouse Strains. Methods Mol Biol 1834: 45–58. 10.1007/978-1-4939-8669-9_3
    1. Cardiff RD, Miller CH, Munn RJ (2014) Manual hematoxylin and eosin staining of mouse tissue sections. Cold Spring Harb Protoc 2014: 655–658. 10.1101/pdb.prot073411
    1. Schindelin J, Rueden CT, Hiner MC, Eliceiri KW (2015) The ImageJ ecosystem: An open platform for biomedical image analysis. Mol Reprod Dev 82: 518–529. 10.1002/mrd.22489

Source: PubMed

3
Abonner