Skip to main content
Erschienen in: BMC Ophthalmology 1/2020

Open Access 01.12.2020 | Case report

Nanophthalmos patient with a THR518MET mutation in MYRF, a case report

verfasst von: Joshua Hagedorn, Armin Avdic, Michael J. Schnieders, Benjamin R. Roos, Young H. Kwon, Arlene V. Drack, Erin A. Boese, John H. Fingert

Erschienen in: BMC Ophthalmology | Ausgabe 1/2020

Abstract

Background

Nanophthalmos has a significant genetic background and disease-causing mutations have been recently been reported in the myelin regulatory factor (MYRF) gene. We report clinical features in a patient with nanophthalmos and a Thr518Met MYRF mutation.

Case presentation

A three-year-old male was discovered to have nanophthalmos after first presenting to the emergency department for a frontal headache, eye pain, emesis, and lethargy. Imaging studies (CT and MRI) were negative except for increased posterior fossa cerebrospinal fluid. Subsequent examinations revealed nanophthalmos (short axial eye lengths 18.1 mm OD and 18.3 mm OS), microcornea, and a large crystalline lens. Peripheral chorioretinal pigment abnormalities were also observed. He experienced episodes of marked ocular hypertension (53 mmHg OD and 60 mmHg) likely due to intermittent angle closure precipitated by nanophthalmos. The ocular hypertension was responsive to topical medicines. Genetic analysis of known nanophthalmos genes MFRP and TMEM98 were negative, while a novel mutation, Thr518Met was detected in MYRF. The Thr518Met mutation was absent from 362 matched normal controls and was extremely rare in a large population database, allele frequency of 0.000024. The Thr518Met mutation altered a highly conserved amino acid in the MYRF protein and three of four algorithms suggested that this mutation is likely pathogenic. Finally, molecular modeling showed that the Thr518Met mutation is damaging to MYRF structure. Together these data suggest that the Thr518Met mutation causes nanophthalmos.

Conclusions

Nanophthalmos may present at an early age with features of angle closure glaucoma and a Thr518Met mutation in MYRF was detected in a patient with nanophthalmos. Prevalence data, homology data, mutation analysis data, and protein modeling data suggest that this variant is pathogenic and may expand the phenotypic range of syndromic nanophthalmos caused by MYRF mutations to include central nervous system abnormalities (increased posterior fossa cerebrospinal fluid).
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
THR
Threonine
MET
Methionine
MYRF
Myelin regulatory factor gene
MFRP
Membrane frizzled-related protein gene
TMEM98
Transmembrane 98 protein gene
PRSS56
Serine protease 56 gene
PRO
Proline
OD
Right eye
OS
Left eye
OU
Both eyes
IOP
Intraocular pressure

Background

The term nanophthalmos is derived from the Greek word “nano” meaning “dwarf,” and is defined by high hyperopia (> + 5 diopters) and/or short axial eye length (< 20 mm). Nanophthalmos has a strong genetic basis. Genes for autosomal recessive (MFRP and PRSS56) and autosomal dominant (TMEM98) nanophthalmos have been discovered [1]. Recently, variants in the myelin regulatory factor (MYRF) gene were identified as another cause of autosomal dominant nanophthalmos and may be responsible for 2–18% of cases of disease [25]. The MYRF gene encodes a membrane protein that is subject to autocleavage, releasing a transcription factor protein fragment that stimulates expression of genes important for myelination of CNS nerve fibers. [6] Variants in MYRF had been previously associated with cardiac defects (scimitar syndrome, dextrocardia), [79] urogenital malformations [79], diaphragmatic hernia [9], and encephalopathy. [10] In 2019, 10 unique MYRF variants were detected in nanophthalmos patients, including 9 that cause premature termination of translation via frameshifts due to insertions, deletions, splicing defects, or nonsense variants [2, 3, 5]. Although many of these nanophthalmos patients had dextrocardia and urogenital malformations [2, 4], some had isolated nanophalmos. [3] De novo MYRF variants have also been detected in three nanophthalmos patients that are the offspring of normal parents [5], suggesting that de novo variants may be a relatively common feature of nanophthalmos caused by the MYRF gene. Here we report a patient with nanophthalmos and a novel mutation in the recently discovered MYRF gene.

Case presentation

Clinical examination

A 3-year-old male (Nan3) presented to the emergency department with a frontal headache, eye pain, emesis, and lethargy. Physical exam was essentially normal with no focal neurologic deficits. Work up included a CT scan and MRI, which identified increased posterior fossa fluid. A lumbar puncture was also performed with normal opening pressure. Eye examination was limited due to patient agitation, but did not reveal any abnormal findings; his visual acuity was at least fix and follow OU, and the dilated eye exam was only notable for peripheral geographic chorioretinal scars OU. Toxoplasmosis serology was obtained, which was negative, and follow-up was arranged with ophthalmology. Both neurology and neurosurgery were following on an as needed basis.
At follow up eye examination, Nan3’s symptoms had resolved. His visual acuities were 2.4cy/cm (20/260) OD and 6.5cy/cm (20/94) OS by Teller cards, and intraocular pressure (IOP) was 7 mmHg OD and 8 mmHg OS by iCare tonometer. Cycloplegic refraction revealed high hyperopia (+ 7.00 + 0.50 × 090 OD and +  7.00 + 0.50 × 090 OS). On subsequent exam under anesthesia, his pachymetry readings were thick at 640 μm OD and 616 μm OS, and the horizontal white-to-white measures were small at 10.5 mm OD and 10.6 mm OS. Gonioscopy showed angles open to the scleral spur OD and with 2 clock-hours of angle closure OS. The anterior segment examination was otherwise normal. The optic nerves OU had mild pallor and cup-to-disc ratios of 0.4 OD and 0.3 OS respectively (Fig. 1a and b). As previously noted, there were chorioretinal scars in the periphery of both eyes (Fig. 1c and d). A-scan ultrasound measured short axial eye lengths of 18.1 mm OD and 18.3 mm OS), a thick sclera (2.0 mm OU), and a relatively large lens (4.3 mm OD and 4.1 mm OS). CT and MRI scans also demonstrate short axial eye lengths and thick sclera (Fig. 2). He did not show any signs of primary congenital glaucoma, including buphthalmos, Haab’s striae, or corneal edema and was diagnosed with nanophthalmos based on axial eye length measurements.
At subsequent follow-up appointments during the next year, Nan3 had several episodes of unilateral high IOPs (once in the right eye to 53 mmHg, three times in the left ranging from 42 to 60 mmHg). Topical medications (timolol-dorzolamide and latanoprost) were successful in lowering intraocular pressure during 9 months of follow-up. Based on his clinical course and measurements, Nan3 was diagnosed with nanophthalmos, microcornea, and likely intermittent angle-closure glaucoma. Close follow-up was arranged to monitor his IOP and iridocorneal angles and the need for surgical intervention. Unfortunately, he was lost to follow-up.
Nan3 has no known family history of nanophthalmos. His parents were unavailable for examination, but are not known to be highly hyperopic or have a history of angle closure glaucoma.

Genetic testing

We tested Nan3’s DNA for coding sequence variants in TMEM98 and MYRF using standard Sanger sequencing, and MFRP was also tested by a commercial laboratory (Bayler Miraca, Houston, TX). No variants were detected in TMEM98 or MFRP, however, we detected two heterozygous variants in MYRF, a synonymous change (rs149803 - Pro263Pro) and a missense change (Thr518Met - rs562652459).

Variant analysis

We evaluated the potential pathogenicity of the Thr518Met variant in the MYRF gene by assessing its prevalence in control subjects with normal eyes. No instances of the Thr518Met variant were detected in the exomes of 362 normal control subjects from the University of Iowa. Moreover, the Thr518Met variant was only rarely observed in the gnomAD database at an allele frequency of 0.000024 in a non-Finnish European population (n = 61,642, gnomad.​broadinstitute.​org) as would be expected for a variant that causes nanophthalmos. Parental samples were not available for segregation studies or to determine if Thr518Met was a de novo variant in the MYRF gene. Nan3’s parents reported no history of high hyperopia or angle closure glaucoma, which would be consistent with a de novo MYRF variant in Nan3. We further explored the pathogenicity of Thr518Met with a homology analysis. The Thr518Met variant alters an amino acid in a DNA-binding domain of MYRF that has been conserved across a diverse range of mammals and vertebrate animals (Fig. 3a). We also investigated the pathogenesis of Thr518Met with variant analysis algorithms. Three of four algorithms suggested that the Thr518Met variant is deleterious (Table 1). Finally, we modeled the tertiary structure of the MYRF protein with and without the Thr518Met variant using its solved crystal structure (PDB ID: 5YHU) [11] and refinement algorithms based on the polarizable AMOEBA force field as we have previously described [12]. Our analysis showed that the Thr518Met variant replaces a polar amino acid (threonine) with a larger, hydrophobic amino acid (methionine), which results in the loss of a hydrogen bond between two MYRF DNA-binding domain β-strands (Fig. 3b). Together these data suggest that the Thr518Met variant in MYRF may cause nanophthalmos by destabilizing the fold of its DNA-binding domain and altering its function as a transcription factor.
Table 1
Features of nanophthalmos patient (Nan3) with a Thr518Met MYRF variant. Allele frequencies of MYRF variants Pro263Pro and Thr518Met were obtained from gnomad.​broadinstitute.​org
Ocular features relevant to nanophthalmos
OD
OS
 Hyperopia
+ 7.00 + 0.50 × 90
+ 7.00 + 0.50 × 90
 Central corneal thickness
640 μm
616 μm
 Ultrasound measurements
  Axial eye length
18.1 mm
18.3 mm
  Anterior chamber depth
2.2 mm
2.4 mm
  Lens
4.3 mm
4.1 mm
  Scleral thickness
1.95 mm
1.95 mm
 Ocular hypertension
  
  Maximum IOP
53 mmHg
60 mmHg
Other ocular features
 Cornea
  Horizontal diameter
10.5 mm
10.5 mm
 Retina
Chorioretinal scars
Systemic features
 Central Nervous System
Posterior fossa cyst
Genetic testing results
MYRF: c.1553C > T, p.Thr518Met (rs562652459)
  Allele frequency
   University of Iowa database (n = 362 controls)
0
   gnomAD database (n = 61,642 Non-Finnish Europeans)
0.000024
  Variant analysis (estimation of pathogenicity)
   Blosum62
−1
   Polyphen
0.677 (Probably damaging)
   CADD
− 21.5 (1% most deleterious)
   SIFT
Tolerated

Discussion and conclusions

Syndromic cases of nanophthalmos due to MYRF variants have included cardiac and genitourinary abnormalities [2, 4]. Here we expand the phenotypic range of MYRF variants by reporting a patient with nanophthalmos along with retina abnormalities and a posterior fossa cyst.
Nan3 meets diagnostic criteria for nanophthalmos. It is likely that this patient’s presenting symptoms of headache, eye pain, emesis, and lethargy were caused by intermittent angle-closure predisposed by nanophthalmos. Nan3 also had microcornea and a relatively large lens, both exacerbating the anterior segment crowding of nanophthalmos and further increasing risk for angle closure. Hyperpigmented chorioretinal scars in the periphery of the retina resembling cobblestone degeneration, as seen in this patient, may be caused by lobular choroidal infarcts during episodes of high IOP. However, gonioscopy during episodes of high IOP did not show iris bombé or occluded angles, and there was no evidence of phacodonesis that might promote intermittent angle closure. The specific cause of elevated IOP in this patient remains unclear and in addition to angle closure, other diagnoses such as Posner-Schlossman or juvenile open angle glaucoma might be included on a differential diagnosis. The markedly early-onset of symptoms in this patient is also unusual and may be a feature of MYRF-related disease.
Nan3 is the only nanophthalmos patient reported to have a Thr518Met variant in MYRF and no segregation data is available to provide support for its pathogenicity. It remains possible that the Thr518Met is a rare, benign variation that was incidentally identified in Nan3. However, the sum of the data (prevalence data, homology data, mutation analysis data, protein modeling data) suggests that this variant may cause syndromic nanophthalmos. Although additional functional and/or animal studies of this variant will be ultimately be needed to prove its pathogenicity, our report of the Thr518Met variant provides additional evidence to support the recent association between MYRF and nanophthalmos.

Acknowledgements

None.
This study was approved by the University of Iowa’s Internal Review Board and adhered to the tenets of the Declaration of Helsinki. Written informed consent to participate was provided by the participant’s parent.
The participant’s parent provided written consent for a research report to be published.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Carricondo PC, Andrade T, Prasov L, Ayres BM, Moroi SE. Nanophthalmos: a review of the clinical Spectrum and genetics. J Ophthalmol. 2018;2018(3):2735465–9.PubMedPubMedCentral Carricondo PC, Andrade T, Prasov L, Ayres BM, Moroi SE. Nanophthalmos: a review of the clinical Spectrum and genetics. J Ophthalmol. 2018;2018(3):2735465–9.PubMedPubMedCentral
2.
Zurück zum Zitat Garnai SJ, Brinkmeier ML, Emery B, Aleman TS, Pyle LC, Veleva-Rotse B, et al. Variants in myelin regulatory factor (MYRF) cause autosomal dominant and syndromic nanophthalmos in humans and retinal degeneration in mice. PLoS Genet. 2019;15(5):e1008130.CrossRef Garnai SJ, Brinkmeier ML, Emery B, Aleman TS, Pyle LC, Veleva-Rotse B, et al. Variants in myelin regulatory factor (MYRF) cause autosomal dominant and syndromic nanophthalmos in humans and retinal degeneration in mice. PLoS Genet. 2019;15(5):e1008130.CrossRef
3.
Zurück zum Zitat Xiao X, Sun W, Ouyang J, Li S, Jia X, Tan Z, et al. Novel truncation mutations in MYRF cause autosomal dominant high hyperopia mapped to 11p12-q13.3. Hum Genet. 2019;138(10):1077–90.CrossRef Xiao X, Sun W, Ouyang J, Li S, Jia X, Tan Z, et al. Novel truncation mutations in MYRF cause autosomal dominant high hyperopia mapped to 11p12-q13.3. Hum Genet. 2019;138(10):1077–90.CrossRef
4.
Zurück zum Zitat Siggs OM, Souzeau E, Breen J, Qassim A, Zhou T, Dubowsky A, et al. Autosomal dominant nanophthalmos and high hyperopia associated with a C-terminal frameshift variant in MYRF. Mol Vis. 2019;25:527–34.PubMedPubMedCentral Siggs OM, Souzeau E, Breen J, Qassim A, Zhou T, Dubowsky A, et al. Autosomal dominant nanophthalmos and high hyperopia associated with a C-terminal frameshift variant in MYRF. Mol Vis. 2019;25:527–34.PubMedPubMedCentral
5.
Zurück zum Zitat Guo C, Zhao Z, Chen D, He S, Sun N, Li Z, et al. Detection of clinically relevant genetic variants in Chinese patients with Nanophthalmos by trio-based whole-genome sequencing study. Invest Ophthalmol Vis Sci. 2019;60(8):2904–13.CrossRef Guo C, Zhao Z, Chen D, He S, Sun N, Li Z, et al. Detection of clinically relevant genetic variants in Chinese patients with Nanophthalmos by trio-based whole-genome sequencing study. Invest Ophthalmol Vis Sci. 2019;60(8):2904–13.CrossRef
6.
Zurück zum Zitat Emery B, Agalliu D, Cahoy JD, Watkins TA, Dugas JC, Mulinyawe SB, et al. Myelin gene regulatory factor is a critical transcriptional regulator required for CNS myelination. Cell. 2009;138(1):172–85.CrossRef Emery B, Agalliu D, Cahoy JD, Watkins TA, Dugas JC, Mulinyawe SB, et al. Myelin gene regulatory factor is a critical transcriptional regulator required for CNS myelination. Cell. 2009;138(1):172–85.CrossRef
7.
Zurück zum Zitat Pinz H, Pyle LC, Li D, Izumi K, Skraban C, Tarpinian J, et al. De novo variants in myelin regulatory factor (MYRF) as candidates of a new syndrome of cardiac and urogenital anomalies. Am J Med Genet A. 2018;176(4):969–72.CrossRef Pinz H, Pyle LC, Li D, Izumi K, Skraban C, Tarpinian J, et al. De novo variants in myelin regulatory factor (MYRF) as candidates of a new syndrome of cardiac and urogenital anomalies. Am J Med Genet A. 2018;176(4):969–72.CrossRef
8.
Zurück zum Zitat Chitayat D, Shannon P, Uster T, Nezarati MM, Schnur RE, Bhoj EJ. 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. 2018;176(9):2041–3.CrossRef Chitayat D, Shannon P, Uster T, Nezarati MM, Schnur RE, Bhoj EJ. 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. 2018;176(9):2041–3.CrossRef
9.
Zurück zum Zitat Qi H, Yu L, Zhou X, Wynn J, Zhao H, Guo Y, et al. De novo variants in congenital diaphragmatic hernia identify MYRF as a new syndrome and reveal genetic overlaps with other developmental disorders. PLoS Genet. 2018;14(12):e1007822.CrossRef Qi H, Yu L, Zhou X, Wynn J, Zhao H, Guo Y, et al. De novo variants in congenital diaphragmatic hernia identify MYRF as a new syndrome and reveal genetic overlaps with other developmental disorders. PLoS Genet. 2018;14(12):e1007822.CrossRef
10.
Zurück zum Zitat Kurahashi H, Azuma Y, Masuda A, Okuno T, Nakahara E, Imamura T, et al. MYRF is associated with encephalopathy with reversible myelin vacuolization. Ann Neurol. 2018;83(1):98–106.CrossRef Kurahashi H, Azuma Y, Masuda A, Okuno T, Nakahara E, Imamura T, et al. MYRF is associated with encephalopathy with reversible myelin vacuolization. Ann Neurol. 2018;83(1):98–106.CrossRef
11.
Zurück zum Zitat Chen B, Zhu Y, Ye S, Zhang R. Structure of the DNA-binding domain of human myelin-gene regulatory factor reveals its potential protein-DNA recognition mode. J Struct Biol. 2018;203(2):170–8.CrossRef Chen B, Zhu Y, Ye S, Zhang R. Structure of the DNA-binding domain of human myelin-gene regulatory factor reveals its potential protein-DNA recognition mode. J Struct Biol. 2018;203(2):170–8.CrossRef
12.
Zurück zum Zitat Tollefson MR, Litman JM, Qi G, O’Connell CE, Wipfler MJ, Marini RJ, et al. Structural insights into hearing loss genetics from polarizable protein repacking. Biophys J. 2019;117(3):602–12.CrossRef Tollefson MR, Litman JM, Qi G, O’Connell CE, Wipfler MJ, Marini RJ, et al. Structural insights into hearing loss genetics from polarizable protein repacking. Biophys J. 2019;117(3):602–12.CrossRef
Metadaten
Titel
Nanophthalmos patient with a THR518MET mutation in MYRF, a case report
verfasst von
Joshua Hagedorn
Armin Avdic
Michael J. Schnieders
Benjamin R. Roos
Young H. Kwon
Arlene V. Drack
Erin A. Boese
John H. Fingert
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Ophthalmology / Ausgabe 1/2020
Elektronische ISSN: 1471-2415
DOI
https://doi.org/10.1186/s12886-020-01659-8

Weitere Artikel der Ausgabe 1/2020

BMC Ophthalmology 1/2020 Zur Ausgabe

Neu im Fachgebiet Augenheilkunde

Ophthalmika in der Schwangerschaft

Die Verwendung von Ophthalmika in der Schwangerschaft und Stillzeit stellt immer eine Off-label-Anwendung dar. Ein Einsatz von Arzneimitteln muss daher besonders sorgfältig auf sein Risiko-Nutzen-Verhältnis bewertet werden. In der vorliegenden …

Operative Therapie und Keimnachweis bei endogener Endophthalmitis

Vitrektomie Originalie

Die endogene Endophthalmitis ist eine hämatogen fortgeleitete, bakterielle oder fungale Infektion, die über choroidale oder retinale Gefäße in den Augapfel eingeschwemmt wird [ 1 – 3 ]. Von dort infiltrieren die Keime in die Netzhaut, den …

Bakterielle endogene Endophthalmitis

Vitrektomie Leitthema

Eine endogene Endophthalmitis stellt einen ophthalmologischen Notfall dar, der umgehender Diagnostik und Therapie bedarf. Es sollte mit geeigneten Methoden, wie beispielsweise dem Freiburger Endophthalmitis-Set, ein Keimnachweis erfolgen. Bei der …

So erreichen Sie eine bestmögliche Wundheilung der Kornea

Die bestmögliche Wundheilung der Kornea, insbesondere ohne die Ausbildung von lichtstreuenden Narben, ist oberstes Gebot, um einer dauerhaften Schädigung der Hornhaut frühzeitig entgegenzuwirken und die Funktion des Auges zu erhalten.   

Update Augenheilkunde

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.