1
|
Tao J, Luo J, Li K, Yang R, Lin Y, Ge J. Comprehensive genetic analysis uncovers the mutational spectrum of MFRP and its genotype-phenotype correlation in a large cohort of Chinese microphthalmia patients. Gene 2024; 926:148647. [PMID: 38848879 DOI: 10.1016/j.gene.2024.148647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
PURPOSE Microphthalmia is a severe congenital ocular disease featured by abnormal ocular development. The aim of this study was to detail the genetic and clinical characteristics of a large cohort of Chinese patients with microphthalmia related to MFRP variants, focusing on uncovering genotype-phenotype correlations. METHODS Fifty microphthalmia patients from 44 unrelated Chinese families were recruited. Whole-exome sequencing (WES) was conducted to analyze the coding regions and adjacent intronic regions of MFRP. Axial lengths (AL) were measured for all probands and available family members. Protein structures of mutations with high frequency in our cohort were predicted. The genotype-phenotype correlations were explored by statistical analysis. RESULTS Sixteen MFRP variants were detected in 17 families, accounting for 38.64 % of all microphthalmia families. There were 9 novel mutations (c.427+1G>C, c.428-2A>C, c.561_575del:p.A188_E192del, c.836G>A:p.C279Y, c.1010_1021del:p.H337_E340del:p.Y479*, c.1516_1517del:p.S506Pfs*66, c.1561T>G:p.C521G, c.1616G>A:p.R539H, and c.1735C>T:p.P579S) and six previously reported variants in MFRP, with p.E496K and p.H337_E340del being highly frequent, found in eight (47.06 %) and two families (11.76 %), respectively. Seven variants (43.75 %) were located in the C-terminal cysteine-rich frizzled-related domain (CRD) (7/16, 43.75 %). Protein prediction implicated p.E496K and p.H337_E340del mutations might lead to a destabilization of the MFRP protein. The average AL of all 42 eyes was 16.02 ± 1.05 mm, and 78.36 % of eyes with AL < 16 mm harbored p.E496K variant. Twenty-six eyes with variant variant had shorter AL than that of the other 16 eyes without this variant (p = 0.006), highlighting a novel genotype-phenotype correlation. CONCLUSIONS In this largest cohort of Chinese patients with microphthalmia, the 9 novel variants, high frequency of p.E496W, and mutation hotspots in CRD reveals unique insights into the MFRP mutation spectrum among Chinese patients, indicating ethnic variability. A new genotype-phenotype correlation that p.E496K variant associated with a shorter AL is unveiled. Our findings enhance the current knowledge of MFRP-associated microphthalmia and provide valuable information for prenatal diagnosis as well as future therapy.
Collapse
Affiliation(s)
- Jing Tao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing 100730, China
| | - Jingyi Luo
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Kaijing Li
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Runcai Yang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Yixiu Lin
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510000, China.
| |
Collapse
|
2
|
Amaral RAS, Zin OA, Moraes RT, Porto FBO, Carricondo PC, Pimentel SLG, Kestelman BP, Watanabe SES, Sallum JMF. Posterior microphthalmos with retinal involvement related to MFRP gene: a report of 10 Brazilian patients. Ophthalmic Genet 2024; 45:413-420. [PMID: 38557281 DOI: 10.1080/13816810.2024.2322650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 04/04/2024]
Abstract
BACKGROUND To describe the phenotype and genotype of 10 Brazilian patients with variants in MFRP, posterior microphthalmos and retinal findings. METHODS Complete ophthalmological evaluation was done at 4 different Brazilian centers. Genetic analysis was performed using commercial next generation sequencing panels for inherited retinal disorders. RESULTS Ages of the patients ranged from 10 to 65 years and visual acuities from 0,05 to no perception of light. All were hyperopes (+4,25 to + 17,50) with a short axial length (14,4 mm to 18 mm). Common posterior segment features, though not present in all, were optic disc drusen (5/10), foveoschisis (5/10) and retinal pigmentary changes (8/10). Isolated patients presented with macular atrophy, serous retinal detachment, and chorioretinal folds. The most common variant in MFRP found in our patients was a deletion in exon 5 (c.498delC; p.Asn267Thrfs *25), present in all except 2 patients. Other variants found were c.523C>T (p.Gln175*), c.298delG (p.Ala100Argfs *37), c.666del (p.Thr223Argfs *83) and the novel variant c.257C>A (p.Ala86Asp). CONCLUSIONS This is the first report of Brazilian patients with posterior microphthalmos and pathogenic variants in MFRP and the first describe of the variant p.Ala86Asp in literature. Our cases confirm the previously reported phenotype of high hyperopia, optic disc drusen, alterations in foveal architecture, retinal pigmentary changes with loss of photoreceptor function and visual field constriction. Report of such a rare condition is important to increase awareness to the phenotype of posterior microphthalmia with associated retinal conditions.
Collapse
Affiliation(s)
- Rebeca A S Amaral
- Department of Ophthalmology, Federal University of São Paulo, São Paulo, Brazil
- Instituto de Genética Ocular (IGO), São Paulo, Brazil
| | - Olivia A Zin
- Department of Ophthalmology, Federal University of São Paulo, São Paulo, Brazil
- Brazilian Institute of Ophthalmology (IBOL), Rio de Janeiro, Brazil
| | - Remo T Moraes
- Brazilian Institute of Ophthalmology (IBOL), Rio de Janeiro, Brazil
| | - Fernanda B O Porto
- INRET Clínica e Centro de Pesquisa, Brazil
- Centro Oftalmológico de Minas Gerais, Brazil
- Clínica de Olhos da Santa Casa de Belo Horizonte, Brazil
| | | | | | | | - Sung E S Watanabe
- Department of Ophthalmology, Federal University of São Paulo, São Paulo, Brazil
| | - Juliana M F Sallum
- Department of Ophthalmology, Federal University of São Paulo, São Paulo, Brazil
- Instituto de Genética Ocular (IGO), São Paulo, Brazil
| |
Collapse
|
3
|
Chiou C, Rajabi F, Fulton AB, Acsadi G, Waitzman D, Gaier ED. Membrane Frizzled-Related Protein-Related Disease Mimicking Idiopathic Intracranial Hypertension. J Neuroophthalmol 2024; 44:e26-e28. [PMID: 36255095 PMCID: PMC10110761 DOI: 10.1097/wno.0000000000001720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Carolina Chiou
- Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA
- Harvard Medical School, Boston, MA
| | - Farrah Rajabi
- Harvard Medical School, Boston, MA
- Department of Genetics, Boston Children’s Hospital, Boston, MA
| | - Anne B. Fulton
- Harvard Medical School, Boston, MA
- Department of Ophthalmology, Boston Children’s Hospital, Boston, MA
| | - Gyula Acsadi
- Department of Neurology, Connecticut Children’s Medical Center, Hartford, CT
| | - David Waitzman
- Departments of Neurology and Ophthalmology, University of Connecticut Health Center, Farmington, CT
| | - Eric D. Gaier
- Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Ophthalmology, Boston Children’s Hospital, Boston, MA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
4
|
Fernández-Vigo JI, Gómez-de-Liaño CN, Rodríguez-Quet O, Burgos-Blasco B, Montolío-Marzo E, de-Pablo-Gómez-de-Liaño L, González-Martin-Moro J, García-Feijóo J. Clinical update in nanophthalmos: Features, diseases and complications associated. ARCHIVOS DE LA SOCIEDAD ESPANOLA DE OFTALMOLOGIA 2023; 98:687-702. [PMID: 37813187 DOI: 10.1016/j.oftale.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/25/2023] [Indexed: 10/11/2023]
Abstract
Nanophthalmos is a rare congenital condition of the eyeball that is characterised by a smaller size of the anterior and posterior segments without associated ocular malformations. Typical features that have traditionally been described in these eyes are short axial length, thickened sclera, cornea with a smaller diameter, narrow anterior chamber, and an increased lens to globe volume ratio. However, at present, there is still a lack of recognised diagnostic criteria for nanophthalmos and a classification of its severity. Its clinical relevance stems from the increased risk of multiple ocular conditions, such as high hyperopia, amblyopia, angle-closure glaucoma, retinal detachment, and cataracts. Likewise, in relation to surgery in these eyes, there are particularities in cataract and glaucoma surgery and with a greater risk of associated intra- and postoperative complications. In this way, the treatment of nanophthalmos focuses on controlling the associated eye conditions and reducing and controlling surgical complications. This review aims to update what has been published in recent years regarding nanophthalmos.
Collapse
Affiliation(s)
- J I Fernández-Vigo
- Departamento de Oftalmología, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria (IdISSC), Madrid, Spain; Centro Internacional de Oftalmología Avanzada, Madrid, Spain.
| | - C N Gómez-de-Liaño
- Departamento de Oftalmología, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria (IdISSC), Madrid, Spain
| | - O Rodríguez-Quet
- Departamento de Oftalmología, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria (IdISSC), Madrid, Spain
| | - B Burgos-Blasco
- Departamento de Oftalmología, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria (IdISSC), Madrid, Spain
| | - E Montolío-Marzo
- Departamento de Oftalmología, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria (IdISSC), Madrid, Spain
| | | | - J González-Martin-Moro
- Departamento de Oftalmología, Hospital Universitario del Henares, Coslada, Madrid, Spain
| | - J García-Feijóo
- Departamento de Oftalmología, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria (IdISSC), Madrid, Spain
| |
Collapse
|
5
|
Ota J, Inooka T, Okado S, Maeda N, Koyanagi Y, Kominami T, Nishiguchi KM, Ueno S. Pathogenic variants of MFRP and PRSS56 genes are major causes of nanophthalmos in Japanese patients. Ophthalmic Genet 2023; 44:423-429. [PMID: 37501562 DOI: 10.1080/13816810.2023.2208220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/23/2023] [Accepted: 04/24/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Nanophthalmos (NNO) is a rare condition with significantly shorter axial length than normal. Several genes are known to cause NNO, among them the MFRP and PRSS56 genes have been reported to cause majority of NNOs. The purpose of this study was to determine the genetic basis of Japanese patients with NNO. MATERIALS AND METHODS We studied seven patients with NNO. Whole exome sequencing (WES) and Sanger sequencing were performed to determine the variants causing the NNO. We also reviewed the medical charts of the patients to determine the phenotype of these seven patients. RESULTS WES revealed that four patients from three families carried homozygous frameshift variants of the PRSS56 gene (c.1066dupC). Two novel variants of the MFRP gene were detected in the other two patients: one proband had a homozygous missense variant (c.1486 G>A) and the other had a compound heterozygous variant (c.1486 G>A and c.662_663insT). The axial length of the eight eyes with the PRSS56 variant was 15.69 ± 0.48 mm (mean ± SD) and that for the 4 eyes with the MFRP variant was 15.63 ± 0.69 mm. Three of the six cases with the PRSS56 or MFRP variant had the uveal effusion syndrome. CONCLUSIONS NNOs in Japanese patients are caused by variants of the PRSS56 and MFRP genes as in other ethnic populations. In addition, two new variants of the MFRP gene were found in our cohort. The phenotypes and anomalies in Japanese patients with NNO were similar to those reported for other ethnic populations.
Collapse
Affiliation(s)
- Junya Ota
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taiga Inooka
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Okado
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Natsuki Maeda
- Department of Ophthalmology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yoshito Koyanagi
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taro Kominami
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koji M Nishiguchi
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Ueno
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Ophthalmology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
6
|
Kovacs KD, Van Tassel SH, Gupta MP. NOVEL MFRP MUTATION WITH NANOPHTHALMOS, OPTIC DISK DRUSEN, AND PERIPHERAL RETINOSCHISIS IMAGED WITH ULTRA-WIDEFIELD OPTICAL COHERENCE TOMOGRAPHY. Retin Cases Brief Rep 2023; 17:269-272. [PMID: 34293777 DOI: 10.1097/icb.0000000000001179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE To describe with multimodal imaging including the use of ultra-widefield optical coherence tomography imaging a distinct phenotype of autosomal recessive nanophthalmos associated with a novel mutation of the MFRP gene (membrane-type frizzled-related protein). METHODS Case report of a single patient followed by the Weill Cornell Medicine Department of Ophthalmology Retina and Glaucoma Services, and review of the relevant literature. RESULTS A patient with a novel homozygous mutation in the MFRP gene (c.472C>T) presented with nanophthalmos, optic disk drusen, foveal hypoplasia, and extensive peripheral retinoschisis, which was revealed to be multilevel retinoschisis on ultra-widefield optical coherence tomography. Unlike other reported cases, the findings associated with this novel mutation did not include foveoschisis or clinically obvious retinitis pigmentosa. The patient underwent prophylactic peripheral laser iridotomy in both eyes. CONCLUSION Here, we present a patient with nanophthalmos, optic disk drusen, and foveal hypoplasia associated with extensive peripheral retinoschisis imaged by ultra-widefield optical coherence tomography, but not foveal retinoschisis or prominent retinitis pigmentosa. The findings may expand the clinical spectrum of MFRP -associated nanophthalmos.
Collapse
Affiliation(s)
- Kyle D Kovacs
- Department of Ophthalmology, Weill Cornell Medical College, New York, New York
| | | | | |
Collapse
|
7
|
Zahra S, Murphy MJ, Crewther SG, Riddell N. Flash Electroretinography as a Measure of Retinal Function in Myopia and Hyperopia: A Systematic Review. Vision (Basel) 2023; 7:vision7010015. [PMID: 36977295 PMCID: PMC10052972 DOI: 10.3390/vision7010015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
Refractive errors (myopia and hyperopia) are the most common visual disorders and are severe risk factors for secondary ocular pathologies. The development of refractive errors has been shown to be associated with changes in ocular axial length, suggested to be induced by outer retinal elements. Thus, the present study systematically reviewed the literature examining retinal function as assessed using global flash electroretinograms (gfERGs) in human clinical refractive error populations. Electronic database searching via Medline, PubMed, Web of Science, Embase, Psych INFO, and CINAHL retrieved 981 unique records (last searched on the 29 May 2022). Single case studies, samples with ocular comorbidities, drug trials, and reviews were excluded. Demographic characteristics, refractive state, gfERG protocol details, and waveform characteristics were extracted for the eight studies that met the inclusion criteria for the review and were judged to have acceptable risk of bias using the OHAT tool (total N = 552 participants; age 7 to 50). Study synthesis suggests that myopia in humans involves attenuation of gfERG photoreceptor (a-wave) and bipolar cell (b-wave) function, consistent with the animal literature. Meaningful interpretation of the overall findings for hyperopia was limited by inconsistent reporting, highlighting the need for future studies to report key aspects of gfERG research design and outcomes more consistently for myopic and hyperopic refractive errors.
Collapse
Affiliation(s)
- Sania Zahra
- Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne 3083, Australia
| | - Melanie J. Murphy
- Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne 3083, Australia
| | - Sheila G. Crewther
- Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne 3083, Australia
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne 3122, Australia
| | - Nina Riddell
- Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne 3083, Australia
- Correspondence:
| |
Collapse
|
8
|
Vanden Heuvel C, Aldred B, Boulter T, Sullivan R, Ver Hoeve J, Schmitt M. MFRP variant results in nanophthalmos, retinitis pigmentosa, variability in foveal avascular zone. Ophthalmic Genet 2023; 44:83-88. [PMID: 35880649 DOI: 10.1080/13816810.2022.2103835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
BACKGROUND Membrane frizzled-related protein (MFRP) plays a critical role in ocular development. MFRP mutations are known to cause nanophthalmos and, in some cases, retinitis pigmentosa, foveoschisis, and/or optic nerve head (ONH) drusen. The broad clinical spectrum of MFRP mutations necessitates further investigation of specific genotype-phenotype relationships. MATERIALS AND METHODS We reviewed ophthalmologic and genetic medical records of two affected siblings and one unaffected sibling. RESULTS Genetic testing revealed variants MFRP c.855T>A, p.(Cys285*) and MFRP c.1235T>C, p.(Leu412Pro) in trans in the two affected siblings. In both cases, photopic and scotopic responses were markedly reduced on electroretinogram (ERG), with greater decrease in scotopic function. Optical coherence tomography for both siblings revealed non-cystoid thickening. Blunted foveal reflexes were also observed in both siblings. Notably, foveal avascular zone abnormalities were seen on fundus autofluorescence in only one affected sibling. CONCLUSIONS MFRP-related ocular disease may be underrecognized due to its presentation with high hyperopia and possibly subtle retinal findings. Presence of variants MFRP c.855T>A, p.(Cys285*) and MFRP c.1235T>C, p.(Leu412Pro) in trans resulted in nanophthalmos and retinitis pigmentosa without associated foveoschisis or ONH drusen in our patients, consistent with the incomplete phenotype previously described in Neri et al. Abnormalities in the foveal avascular zone have been noted in other case studies and were inconsistently associated with the variants described here, representing a potential area for future investigation.
Collapse
Affiliation(s)
- Claire Vanden Heuvel
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Breanna Aldred
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Tyler Boulter
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Rachel Sullivan
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - James Ver Hoeve
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Melanie Schmitt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
9
|
Hanyu T, Ueki S, Hasegawa Y, Kiyokawa M, Fukuchi T. Posterior microphthalmos with achievement of good visual acuity and disappearance of papillomacular retinal folds: a case report. BMC Ophthalmol 2022; 22:424. [PMID: 36344972 PMCID: PMC9639268 DOI: 10.1186/s12886-022-02648-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
Background Posterior microphthalmos (PM) is a rare condition with poor visual prognosis even after amblyopia treatment. We report a case of PM with achievement of good visual acuity and disappearance of papillomacular retinal folds (PFs) over a period of 7 years. Case presentation A girl aged 3 years and 5 months was referred to our hospital, after poor visual acuity was identified at a medical checkup for 3-year-olds. She had severe spherical hyperopia: + 17.25 D in the right eye (RE) and + 18 D in the left eye (LE). Her corrected visual acuity was 20/200 in the RE and 20/250 in the LE. PFs were observed in both eyes on optical coherence tomography (OCT), and the diagnosis of PM was made based on the normal corneal diameter and anterior chamber depth. During the course of the disease, a gradual decrease in the height of the PFs was observed on OCT. The corrected visual acuity at age 10 years was 20/20 in the RE and 20/25 in the LE. Conclusions The visual prognosis of PM is poor, and only one case with good visual acuity has been reported in the literature. The patient in the present case not only developed good visual acuity, but also showed improvement in macular morphology, which was not noted in previous reports. Early diagnosis of PM and early amblyopia treatment is important for the visual development in PM.
Collapse
Affiliation(s)
- Takako Hanyu
- grid.260975.f0000 0001 0671 5144Division of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi-Dori 1-757, Chuo-Ku, Niigata, 951-8510 Japan ,Hanyu Clinic, Igarashi-Higashi 1-1-15, Nishi-Ku, Niigata, 950-2045 Japan
| | - Satoshi Ueki
- grid.260975.f0000 0001 0671 5144Division of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi-Dori 1-757, Chuo-Ku, Niigata, 951-8510 Japan
| | - Yukari Hasegawa
- grid.260975.f0000 0001 0671 5144Division of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi-Dori 1-757, Chuo-Ku, Niigata, 951-8510 Japan
| | - Megumi Kiyokawa
- grid.260975.f0000 0001 0671 5144Division of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi-Dori 1-757, Chuo-Ku, Niigata, 951-8510 Japan ,Toshimi Eye Clinic, Katsubogawa 1-1-12, Sanjo City, Niigata, 955-0056 Japan
| | - Takeo Fukuchi
- grid.260975.f0000 0001 0671 5144Division of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi-Dori 1-757, Chuo-Ku, Niigata, 951-8510 Japan
| |
Collapse
|
10
|
Kumari A, Ayala-Ramirez R, Zenteno JC, Huffman K, Sasik R, Ayyagari R, Borooah S. Single cell RNA sequencing confirms retinal microglia activation associated with early onset retinal degeneration. Sci Rep 2022; 12:15273. [PMID: 36088481 PMCID: PMC9464204 DOI: 10.1038/s41598-022-19351-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Mutations in the Membrane-type frizzled related protein (Mfrp) gene results in an early-onset retinal degeneration associated with retinitis pigmentosa, microphthalmia, optic disc drusen and foveal schisis. In the current study, a previously characterized mouse model of human retinal degeneration carrying homozygous c.498_499insC mutations in Mfrp (MfrpKI/KI) was used. Patients carrying this mutation have retinal degeneration at an early age. The model demonstrates subretinal deposits and develops early-onset photoreceptor degeneration. We observed large subretinal deposits in MfrpKI/KI mice which were strongly CD68 positive and co-localized with autofluorescent spots. Single cell RNA sequencing of MfrpKI/KI mice retinal microglia showed a significantly higher number of pan-macrophage marker Iba-1 and F4/80 positive cells with increased expression of activation marker (CD68) and lowered microglial homeostatic markers (TMEM119, P2ry13, P2ry13, Siglech) compared with wild type mice confirming microglial activation as observed in retinal immunostaining showing microglia activation in subretinal region. Trajectory analysis identified a small cluster of microglial cells with activation transcriptomic signatures that could represent a subretinal microglia population in MfrpKI/KI mice expressing higher levels of APOE. We validated these findings using immunofluorescence staining of retinal cryosections and found a significantly higher number of subretinal Iba-1/ApoE positive microglia in MfrpKI/KI mice with some subretinal microglia also expressing lowered levels of microglial homeostatic marker TMEM119, confirming microglial origin. In summary, we confirm that MfrpKI/KI mice carrying the c.498_499insC mutation had a significantly higher population of activated microglia in their retina with distinct subsets of subretinal microglia. Further, studies are required to confirm whether the association of increased subretinal microglia in MfrpKI/KI mice are causal in degeneration.
Collapse
|
11
|
Drinking hydrogen water improves photoreceptor structure and function in retinal degeneration 6 mice. Sci Rep 2022; 12:13610. [PMID: 35948585 PMCID: PMC9365798 DOI: 10.1038/s41598-022-17903-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/02/2022] [Indexed: 11/08/2022] Open
Abstract
Retinitis pigmentosa (RP) is a genetically heterogeneous group of inherited retinal disorders involving the progressive dysfunction of photoreceptors and the retinal pigment epithelium, for which there is currently no treatment. The rd6 mouse is a natural model of autosomal recessive retinal degeneration. Given the known contributions of oxidative stress caused by reactive oxygen species (ROS) and selective inhibition of potent ROS peroxynitrite and OH·by H2 gas we have previously demonstrated, we hypothesized that ingestion of H2 water may delay the progression of photoreceptor death in rd6 mice. H2 mice showed significantly higher retinal thickness as compared to controls on optical coherence tomography. Histopathological and morphometric analyses revealed higher thickness of the outer nuclear layer for H2 mice than controls, as well as higher counts of opsin red/green-positive cells. RNA sequencing (RNA-seq) analysis of differentially expressed genes in the H2 group versus control group revealed 1996 genes with significantly different expressions. Gene and pathway ontology analysis showed substantial upregulation of genes responsible for phototransduction in H2 mice. Our results show that drinking water high in H2 (1.2-1.6 ppm) had neuroprotective effects and inhibited photoreceptor death in mice, and suggest the potential of H2 for the treatment of RP.
Collapse
|
12
|
Non-vasogenic cystoid maculopathies. Prog Retin Eye Res 2022; 91:101092. [PMID: 35927124 DOI: 10.1016/j.preteyeres.2022.101092] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022]
Abstract
Besides cystoid macular edema due to a blood-retinal barrier breakdown, another type of macular cystoid spaces referred to as non-vasogenic cystoid maculopathies (NVCM) may be detected on optical coherence tomography but not on fluorescein angiography. Various causes may disrupt retinal cell cohesion or impair retinal pigment epithelium (RPE) and Müller cell functions in the maintenance of retinal dehydration, resulting in cystoid spaces formation. Tractional causes include vitreomacular traction, epiretinal membranes and myopic foveoschisis. Surgical treatment does not always allow cystoid space resorption. In inherited retinal dystrophies, cystoid spaces may be part of the disease as in X-linked retinoschisis or enhanced S-cone syndrome, or occur occasionally as in bestrophinopathies, retinitis pigmentosa and allied diseases, congenital microphthalmia, choroideremia, gyrate atrophy and Bietti crystalline dystrophy. In macular telangiectasia type 2, cystoid spaces and cavitations do not depend on the fluid leakage from telangiectasia. Various causes affecting RPE function may result in NVCM such as chronic central serous chorioretinopathy and paraneoplastic syndromes. Non-exudative age macular degeneration may also be complicated by intraretinal cystoid spaces in the absence of fluorescein leakage. In these diseases, cystoid spaces occur in a context of retinal cell loss. Various causes of optic atrophy, including open-angle glaucoma, result in microcystoid spaces in the inner nuclear layer due to a retrograde transsynaptic degeneration. Lastly, drug toxicity may also induce cystoid maculopathy. Identifying NVCM on multimodal imaging, including fluorescein angiography if needed, allows guiding the diagnosis of the causative disease and choosing adequate treatment when available.
Collapse
|
13
|
Lopez-Cañizares A, Carletti P, Berrocal AM. A Young Boy With Changes in the Retinal Pigment Epithelium. JAMA Ophthalmol 2022; 140:1015-1016. [PMID: 35862055 DOI: 10.1001/jamaophthalmol.2022.1653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
14
|
Ren X, Gao Y, Lin Y, Fu X, Xiao L, Wang X, Zeng Z, Bao L, Yan N, Zhang M, Tang L. A Novel Mutation in the Membrane Frizzled-Related Protein Gene for Posterior Microphthalmia, Non-pigmented Retinitis Pigmentosa, Optic Nerve Drusen, and Retinoschisis in a Consanguineous Family. Front Med (Lausanne) 2022; 9:835621. [PMID: 35402469 PMCID: PMC8987310 DOI: 10.3389/fmed.2022.835621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Microphthalmos (MCO) is a rare developmental defect characterized by small malformed eyes. Our study aimed to describe the clinical characteristics of posterior microphthalmos syndrome caused by a novel variant in MFRP gene in a Chinese patient. Methods Complete ophthalmologic examinations were performed for the proband and proband's family members. Whole exon sequencing (WES) and Sanger sequencing were used to identify the mutated genes, and bioinformatic analysis was undertaken to predict the effect of this variant. Results Clinical analysis showed that the proband had reduced axial length (17.95 and 17.98 mm) with normal-size corneas and shallow anterior chamber depth. Fundus photography showed scattered yellowish-white spots in the whole retina with cup-to-disc ratios of 0.95 in both eyes. Retinoschisis in the inner nuclear layer and reduced outer retina thickness were apparent on OCT examination, and optic nerve drusen demonstrated increased autofluorescence in fundus autofluorescence (FAF). Perimeter examination revealed a tubular visual field for the right eye, and electroretinography (ERG) revealed a moderately reduced rod response combined with compromised cone response. Ocular examinations of the patient's family members were unremarkable. WES revealed that the proband had homozygous mutations in c.55-1 (IVS1) G>A in intron 1 for the MFRP gene. Both the proband's parents and offspring were confirmed to be heterozygous by Sanger sequencing. Bioinformatic analysis showed this mutation was deleterious. Conclusion We reported autosomal recessive posterior microphthalmia, atypical retinitis pigmentosa, and retinoschisis caused by a novel mutation in the MFRP gene in this consanguineous marriage family. Our study further broadens the mutation and phenotype spectrum of the MFRP gene in microphthalmia.
Collapse
Affiliation(s)
- Xiang Ren
- Ophthalmic Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yunxia Gao
- Ophthalmic Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Lin
- Ophthalmic Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangyu Fu
- Ophthalmic Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lirong Xiao
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyue Wang
- Ophthalmic Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhibing Zeng
- Ophthalmic Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Bao
- Ophthalmic Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Naihong Yan
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Zhang
- Ophthalmic Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Ming Zhang
| | - Li Tang
- Ophthalmic Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Li Tang
| |
Collapse
|
15
|
Genetic Interaction between Mfrp and Adipor1 Mutations Affect Retinal Disease Phenotypes. Int J Mol Sci 2022; 23:ijms23031615. [PMID: 35163536 PMCID: PMC8835889 DOI: 10.3390/ijms23031615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Adipor1tm1Dgen and Mfrprd6 mutant mice share similar eye disease characteristics. Previously, studies established a functional relationship of ADIPOR1 and MFRP proteins in maintaining retinal lipidome homeostasis and visual function. However, the independent and/or interactive contribution of both genes to similar disease phenotypes, including fundus spots, decreased axial length, and photoreceptor degeneration has yet to be examined. We performed a gene-interaction study where homozygous Adipor1tm1Dgen and Mfrprd6 mice were bred together and the resulting doubly heterozygous F1 offspring were intercrossed to produce 210 F2 progeny. Four-month-old mice from all nine genotypic combinations obtained in the F2 generation were assessed for white spots by fundus photo documentation, for axial length by caliper measurements, and for photoreceptor degeneration by histology. Two-way factorial ANOVA was performed to study individual as well as gene interaction effects on each phenotype. Here, we report the first observation of reduced axial length in Adipor1tmlDgen homozygotes. We show that while Adipor1 and Mfrp interact to affect spotting and degeneration, they act independently to control axial length, highlighting the complex functional association between these two genes. Further examination of the molecular basis of this interaction may help in uncovering mechanisms by which these genes perturb ocular homeostasis.
Collapse
|
16
|
Brandt ZJ, Collery RF, Besharse JC, Link BA. Ablation of mpeg+ Macrophages Exacerbates mfrp-Related Hyperopia. Invest Ophthalmol Vis Sci 2021; 62:13. [PMID: 34913948 PMCID: PMC8684298 DOI: 10.1167/iovs.62.15.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Proper refractive development of the eye, termed emmetropization, is critical for focused vision and is impacted by both genetic determinants and several visual environment factors. Improper emmetropization caused by genetic variants can lead to congenital hyperopia, which is characterized by small eyes and relatively short ocular axial length. To date, variants in only four genes have been firmly associated with human hyperopia, one of which is MFRP. Zebrafish mfrp mutants also have hyperopia and, similar to reports in mice, exhibit increased macrophage recruitment to the retina. The goal of this research was to examine the effects of macrophage ablation on emmetropization and mfrp-related hyperopia. Methods We utilized a chemically inducible, cell-specific ablation system to deplete macrophages in both wild-type and mfrp mutant zebrafish. Spectral-domain optical coherence tomography was then used to measure components of the eye and determine relative refractive state. Histology, immunohistochemistry, and transmission electron microscopy were used to further study the eyes. Results Although macrophage ablation does not cause significant changes to the relative refractive state of wild-type zebrafish, macrophage ablation in mfrp mutants significantly exacerbates their hyperopic phenotype, resulting in a relative refractive error 1.3 times higher than that of non-ablated mfrp siblings. Conclusions Genetic inactivation of mfrp leads to hyperopia, as well as abnormal accumulation of macrophages in the retina. Ablation of the mpeg1-positive macrophage population exacerbates the hyperopia, suggesting that macrophages may be recruited in an effort help preserve emmetropization and ameliorate hyperopia.
Collapse
Affiliation(s)
- Zachary J Brandt
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Ross F Collery
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Joseph C Besharse
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Brian A Link
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
17
|
Balikov DA, Jacobson A, Prasov L. Glaucoma Syndromes: Insights into Glaucoma Genetics and Pathogenesis from Monogenic Syndromic Disorders. Genes (Basel) 2021; 12:genes12091403. [PMID: 34573386 PMCID: PMC8471311 DOI: 10.3390/genes12091403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 11/21/2022] Open
Abstract
Monogenic syndromic disorders frequently feature ocular manifestations, one of which is glaucoma. In many cases, glaucoma in children may go undetected, especially in those that have other severe systemic conditions that affect other parts of the eye and the body. Similarly, glaucoma may be the first presenting sign of a systemic syndrome. Awareness of syndromes associated with glaucoma is thus critical both for medical geneticists and ophthalmologists. In this review, we highlight six categories of disorders that feature glaucoma and other ocular or systemic manifestations: anterior segment dysgenesis syndromes, aniridia, metabolic disorders, collagen/vascular disorders, immunogenetic disorders, and nanophthalmos. The genetics, ocular and systemic features, and current and future treatment strategies are discussed. Findings from rare diseases also uncover important genes and pathways that may be involved in more common forms of glaucoma, and potential novel therapeutic strategies to target these pathways.
Collapse
Affiliation(s)
- Daniel A. Balikov
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (D.A.B.); (A.J.)
| | - Adam Jacobson
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (D.A.B.); (A.J.)
| | - Lev Prasov
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (D.A.B.); (A.J.)
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence:
| |
Collapse
|
18
|
Müller PL, Treis T, Alsaedi A, Webster AR, Khaw P, Michaelides M, Wickham L, Siriwardena D, Foster P, Moosajee M, Pavesio C, Tufail A, Egan C. Foveal structure and visual function in nanophthalmos and posterior microphthalmos. Br J Ophthalmol 2021; 106:1164-1170. [PMID: 34301612 DOI: 10.1136/bjophthalmol-2020-318717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/27/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND/AIMS The reason for visual impairment in patients with nanophthalmos and posterior microphthalmos is not completely understood. Therefore, this study aims to investigate foveal structure, and the impact of demographic, clinical and imaging parameters on best-corrected visual acuity (BCVA) in these conditions. METHODS Sixty-two eyes of 33 patients with nanophthalmos (n=40) or posterior microphthalmos (n=22), and 114 eyes of healthy controls with high-resolution retinal imaging including spectral-domain or swept-source optical coherence tomography images were included in this cross-sectional case-control study. Foveal retinal layer thickness was determined by two independent readers. A mixed-effect model was used to perform structure-function correlations and predict the BCVA based on subject-specific variables. RESULTS Most patients (28/33) had altered foveal structure associated with loss of foveal avascular zone and impaired BCVA. However, widening of outer nuclear layer, lengthening of photoreceptor outer segments, normal distribution of macular pigment and presence of Henle fibres were consistently found. Apart from the presence of choroidal effusion, which had significant impact on BCVA, the features age, refractive error, axial length and retinal layer thickness at the foveal centre explained 61.7% of the variability of BCVA. CONCLUSION This study demonstrates that choroidal effusion, age, refractive error, axial length and retinal layer thickness are responsible for the majority of interindividual variability of BCVA as well as the morphological foveal heterogeneity in patients with nanophthalmos or posterior microphthalmos. This might give further insights into the physiology of foveal development and the process of emmetropisation, and support clinicians in the assessment of these disease entities.
Collapse
Affiliation(s)
- Philipp L Müller
- Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Department of Ophthalmology, University of Bonn, Bonn, Germany.,Macula Center, Suedblick Eye Centers, Augsburg, Germany.,Institute of Ophthalmology, University College London, London, UK
| | - Tim Treis
- Institute of Pharmacy and Molekular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Abdulrahman Alsaedi
- Moorfields Eye Hospital NHS Foundation Trust, London, UK.,College of Medicine, Imam Muhammad bin Saud Islamic University, Riyadh, Saudi Arabia
| | - Andrew R Webster
- Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Institute of Ophthalmology, University College London, London, UK
| | - Peng Khaw
- Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Institute of Ophthalmology, University College London, London, UK
| | - Michel Michaelides
- Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Institute of Ophthalmology, University College London, London, UK
| | - Louisa Wickham
- Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Institute of Ophthalmology, University College London, London, UK
| | | | - Paul Foster
- Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Institute of Ophthalmology, University College London, London, UK
| | - Mariya Moosajee
- Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Great Ormond Street Hospital for Children, London, UK
| | - Carlos Pavesio
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Adnan Tufail
- Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Institute of Ophthalmology, University College London, London, UK
| | - Catherine Egan
- Moorfields Eye Hospital NHS Foundation Trust, London, UK .,Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
19
|
Retinal Stem Cell 'Retirement Plans': Growth, Regulation and Species Adaptations in the Retinal Ciliary Marginal Zone. Int J Mol Sci 2021; 22:ijms22126528. [PMID: 34207050 PMCID: PMC8234741 DOI: 10.3390/ijms22126528] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
The vertebrate retina develops from a specified group of precursor cells that adopt distinct identities and generate lineages of either the neural retina, retinal pigmented epithelium, or ciliary body. In some species, including teleost fish and amphibians, proliferative cells with stem-cell-like properties capable of continuously supplying new retinal cells post-embryonically have been characterized and extensively studied. This region, termed the ciliary or circumferential marginal zone (CMZ), possibly represents a conserved retinal stem cell niche. In this review, we highlight the research characterizing similar CMZ-like regions, or stem-like cells located at the peripheral margin, across multiple different species. We discuss the proliferative parameters, multipotency and growth mechanisms of these cells to understand how they behave in vivo and how different molecular factors and signalling networks converge at the CMZ niche to regulate their activity. The evidence suggests that the mature retina may have a conserved propensity for homeostatic growth and plasticity and that dysfunction in the regulation of CMZ activity may partially account for dystrophic eye growth diseases such as myopia and hyperopia. A better understanding of the properties of CMZ cells will enable important insight into how an endogenous generative tissue compartment can adapt to altered retinal physiology and potentially even restore vision loss caused by retinal degenerative conditions.
Collapse
|
20
|
Lang E, Koller S, Atac D, Pfäffli OA, Hanson JV, Feil S, Bähr L, Bahr A, Kottke R, Joset P, Fasler K, Barthelmes D, Steindl K, Konrad D, Wille D, Berger W, Gerth‐Kahlert C. Genotype-phenotype spectrum in isolated and syndromic nanophthalmos. Acta Ophthalmol 2021; 99:e594-e607. [PMID: 32996714 DOI: 10.1111/aos.14615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/01/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE To (i) describe a series of patients with isolated or syndromic nanophthalmos with the underlying genetic causes, including novel pathogenic variants and their functional characterization and (ii) to study the association of retinal dystrophy in patients with MFRP variants, based on a detailed literature review of genotype-phenotype correlations. METHODS Patients with nanophthalmos and available family members received a comprehensive ophthalmological examination. Genetic analysis was based on whole-exome sequencing and variant calling in core genes including MFRP, BEST1, TMEM98, PRSS56, CRB1, GJA1, C1QTNF5, MYRF and FAM111A. A minigene assay was performed for functional characterization of a splice site variant. RESULTS Seven patients, aged between three and 65 years, from five unrelated families were included. Novel pathogenic variants in MFRP (c.497C>T, c.899-3C>A, c.1180G>A), and PRSS56 (c.1202C>A), and a recurrent de novo variant in FAM111A (c.1706G>A) in a patient with Kenny-Caffey syndrome type 2, were identified. In addition, we report co-inheritance of MFRP-related nanophthalmos and ADAR-related Aicardi-Goutières syndrome. CONCLUSION Nanophthalmos is a genetically heterogeneous condition, and the severity of ocular manifestations appears not to correlate with variants in a specific gene. However, retinal dystrophy is only observed in patients harbouring pathogenic MFRP variants. Furthermore, heterozygous carriers of MFRP and PRSS56 should be screened for the presence of high hyperopia. Identifying nanophthalmos as an isolated condition or as part of a syndrome has implications for counselling and can accelerate the interdisciplinary care of patients.
Collapse
Affiliation(s)
- Elena Lang
- Department of Ophthalmology University Hospital Zurich and University of Zurich Zurich Switzerland
- Institute of Medical Molecular Genetics University of Zurich Schlieren Switzerland
| | - Samuel Koller
- Institute of Medical Molecular Genetics University of Zurich Schlieren Switzerland
| | - David Atac
- Institute of Medical Molecular Genetics University of Zurich Schlieren Switzerland
| | - Oliver A. Pfäffli
- Department of Ophthalmology University Hospital Zurich and University of Zurich Zurich Switzerland
| | - James V.M. Hanson
- Department of Ophthalmology University Hospital Zurich and University of Zurich Zurich Switzerland
| | - Silke Feil
- Institute of Medical Molecular Genetics University of Zurich Schlieren Switzerland
| | - Luzy Bähr
- Institute of Medical Molecular Genetics University of Zurich Schlieren Switzerland
| | - Angela Bahr
- Institute of Medical Genetics University of Zurich Zurich Switzerland
| | - Raimund Kottke
- Department of Diagnostic Imaging University Children's Hospital Zurich Zurich Switzerland
| | - Pascal Joset
- Institute of Medical Genetics University of Zurich Zurich Switzerland
| | - Katrin Fasler
- Department of Ophthalmology University Hospital Zurich and University of Zurich Zurich Switzerland
| | - Daniel Barthelmes
- Department of Ophthalmology University Hospital Zurich and University of Zurich Zurich Switzerland
- Save Sight Institute The University of Sydney Sydney NSW Australia
| | - Katharina Steindl
- Institute of Medical Genetics University of Zurich Zurich Switzerland
| | - Daniel Konrad
- Department of Pediatric Endocrinology and Diabetology University Children’s Hospital Zurich Switzerland
| | | | - Wolfgang Berger
- Institute of Medical Molecular Genetics University of Zurich Schlieren Switzerland
- Zurich Center for Integrative Human Physiology University of Zurich Zurich Switzerland
- Neuroscience Center Zurich, University and ETH Zurich Zurich Switzerland
| | - Christina Gerth‐Kahlert
- Department of Ophthalmology University Hospital Zurich and University of Zurich Zurich Switzerland
| |
Collapse
|
21
|
Tideman JWL, Pärssinen O, Haarman AEG, Khawaja AP, Wedenoja J, Williams KM, Biino G, Ding X, Kähönen M, Lehtimäki T, Raitakari OT, Cheng CY, Jonas JB, Young TL, Bailey-Wilson JE, Rahi J, Williams C, He M, Mackey DA, Guggenheim JA. Evaluation of Shared Genetic Susceptibility to High and Low Myopia and Hyperopia. JAMA Ophthalmol 2021; 139:601-609. [PMID: 33830181 DOI: 10.1001/jamaophthalmol.2021.0497] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Importance Uncertainty currently exists about whether the same genetic variants are associated with susceptibility to low myopia (LM) and high myopia (HM) and to myopia and hyperopia. Addressing this question is fundamental to understanding the genetics of refractive error and has clinical relevance for genotype-based prediction of children at risk for HM and for identification of new therapeutic targets. Objective To assess whether a common set of genetic variants are associated with susceptibility to HM, LM, and hyperopia. Design, Setting, and Participants This genetic association study assessed unrelated UK Biobank participants 40 to 69 years of age of European and Asian ancestry. Participants 40 to 69 years of age living in the United Kingdom were recruited from January 1, 2006, to October 31, 2010. Of the total sample of 502 682 participants, 117 279 (23.3%) underwent an ophthalmic assessment. Data analysis was performed from December 12, 2019, to June 23, 2020. Exposures Four refractive error groups were defined: HM, -6.00 diopters (D) or less; LM, -3.00 to -1.00 D; hyperopia, +2.00 D or greater; and emmetropia, 0.00 to +1.00 D. Four genome-wide association study (GWAS) analyses were performed in participants of European ancestry: (1) HM vs emmetropia, (2) LM vs emmetropia, (3) hyperopia vs emmetropia, and (4) LM vs hyperopia. Polygenic risk scores were generated from GWAS summary statistics, yielding 4 sets of polygenic risk scores. Performance was assessed in independent replication samples of European and Asian ancestry. Main Outcomes and Measures Odds ratios (ORs) of polygenic risk scores in replication samples. Results A total of 51 841 unrelated individuals of European ancestry and 2165 unrelated individuals of Asian ancestry were assigned to a specific refractive error group and included in our analyses. Polygenic risk scores derived from all 4 GWAS analyses were predictive of all categories of refractive error in both European and Asian replication samples. For example, the polygenic risk score derived from the HM vs emmetropia GWAS was predictive in the European sample of HM vs emmetropia (OR, 1.58; 95% CI, 1.41-1.77; P = 1.54 × 10-15) as well as LM vs emmetropia (OR, 1.15; 95% CI, 1.07-1.23; P = 8.14 × 10-5), hyperopia vs emmetropia (OR, 0.83; 95% CI, 0.77-0.89; P = 4.18 × 10-7), and LM vs hyperopia (OR, 1.45; 95% CI, 1.33-1.59; P = 1.43 × 10-16). Conclusions and Relevance Genetic risk variants were shared across HM, LM, and hyperopia and across European and Asian samples. Individuals with HM inherited a higher number of variants from among the same set of myopia-predisposing alleles and not different risk alleles compared with individuals with LM. These findings suggest that treatment interventions targeting common genetic risk variants associated with refractive error could be effective against both LM and HM.
Collapse
Affiliation(s)
- J Willem L Tideman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Olavi Pärssinen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.,Department of Ophthalmology, Central Hospital of Central Finland, Jyväskylä, Finland
| | - Annechien E G Haarman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Anthony P Khawaja
- NIHR Biomedical Research Centre, Moorfields Eye Hospital National Health Service (NHS) Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Juho Wedenoja
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Katie M Williams
- Section of Academic Ophthalmology, Faculty of Life Sciences and Medicine, King's College London School of Life Course Sciences, London, United Kingdom.,Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of Italy, Pavia, Italy
| | - Xiaohu Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Mika Kähönen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - Terho Lehtimäki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland.,Department of Clinical Chemistry, Finnish Cardiovascular Research Center, Tampere, Finland
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Finland.,Research Centre of Applied and Preventive Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Ching-Yu Cheng
- Duke-NUS Medical School, Singapore, Singapore.,Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Jost B Jonas
- Department of Ophthalmology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Beijing Institute of Ophthalmology, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Terri L Young
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison
| | - Joan E Bailey-Wilson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland
| | - Jugnoo Rahi
- UCL Great Ormond Street Institute of Child Health and Institute of Ophthalmology, University College London, London, United Kingdom
| | - Cathy Williams
- Centre for Academic Child Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Mingguang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.,Centre for Eye Research Australia; Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - David A Mackey
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Jeremy A Guggenheim
- Cardiff University School of Optometry and Vision Sciences, Cardiff, United Kingdom
| | | |
Collapse
|
22
|
Koli S, Labelle-Dumais C, Zhao Y, Paylakhi S, Nair KS. Identification of MFRP and the secreted serine proteases PRSS56 and ADAMTS19 as part of a molecular network involved in ocular growth regulation. PLoS Genet 2021; 17:e1009458. [PMID: 33755662 PMCID: PMC8018652 DOI: 10.1371/journal.pgen.1009458] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 04/02/2021] [Accepted: 03/01/2021] [Indexed: 12/24/2022] Open
Abstract
Precise regulation of ocular size is a critical determinant of normal visual acuity. Although it is generally accepted that ocular growth relies on a cascade of signaling events transmitted from the retina to the sclera, the factors and mechanism(s) involved are poorly understood. Recent studies have highlighted the importance of the retinal secreted serine protease PRSS56 and transmembrane glycoprotein MFRP, a factor predominantly expressed in the retinal pigment epithelium (RPE), in ocular size determination. Mutations in PRSS56 and MFRP constitute a major cause of nanophthalmos, a condition characterized by severe reduction in ocular axial length/extreme hyperopia. Interestingly, common variants of these genes have been implicated in myopia, a condition associated with ocular elongation. Consistent with these findings, mice with loss of function mutation in PRSS56 or MFRP exhibit a reduction in ocular axial length. However, the molecular network and cellular processes involved in PRSS56- and MFRP-mediated ocular axial growth remain elusive. Here, we show that Adamts19 expression is significantly upregulated in the retina of mice lacking either Prss56 or Mfrp. Importantly, using genetic mouse models, we demonstrate that while ADAMTS19 is not required for ocular growth during normal development, its inactivation exacerbates ocular axial length reduction in Prss56 and Mfrp mutant mice. These results suggest that the upregulation of retinal Adamts19 is part of an adaptive molecular response to counteract impaired ocular growth. Using a complementary genetic approach, we show that loss of PRSS56 or MFRP function prevents excessive ocular axial growth in a mouse model of early-onset myopia caused by a null mutation in Irbp, thus, demonstrating that PRSS56 and MFRP are also required for pathological ocular elongation. Collectively, our findings provide new insights into the molecular network involved in ocular axial growth and support a role for molecular crosstalk between the retina and RPE involved in refractive development. During ocular refractive development, the eye’s growth is modulated, such that the ocular axial length matches the optical power enabling the eyes to achieve optimal focus. Alterations in ocular growth mainly contribute to refractive errors. Mutations in human PRSS56 and MFRP are responsible for nanophthalmos that exhibit a severe reduction in ocular axial length, and high hyperopia. Importantly, mutant mouse models lacking either Müller glia expressed PRSS56, or retinal pigment epithelium (RPE) localized MFRP exhibit ocular axial length reduction. Here, we have identified Adamts19 as a factor whose levels were significantly upregulated in the retina of mice lacking either Prss56 or Mfrp. Importantly, utilizing Adamts19 knockout mice we demonstrate that upregulation of retinal Adamts19 expression constitutes a compensatory mechanism that provides partial protection against ocular axial reduction due to mutation in Prss56 and Mfrp. Next, utilizing a mouse model of early-onset myopia, we demonstrate that the mutant Irbp induced ocular axial elongation is completely dependent on Prss56 as well as Mfrp, suggesting an interplay between Müller glia and RPE in the regulation of ocular axial growth. Collectively, these findings suggest that ocular refractive development relies on complex interactions occurring between genetic factors in the retina and RPE.
Collapse
Affiliation(s)
- Swanand Koli
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Yin Zhao
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Seyyedhassan Paylakhi
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - K. Saidas Nair
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
- Department of Anatomy, University of California, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
23
|
Kondkar AA. Updates on Genes and Genetic Mechanisms Implicated in Primary Angle-Closure Glaucoma. APPLICATION OF CLINICAL GENETICS 2021; 14:89-112. [PMID: 33727852 PMCID: PMC7955727 DOI: 10.2147/tacg.s274884] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/18/2021] [Indexed: 12/29/2022]
Abstract
Primary angle-closure glaucoma (PACG) is estimated to affect over 30 million people worldwide by 2040 and is highly prevalent in the Asian population. PACG is more severe and carries three times the higher risk of blindness than primary open-angle glaucoma, thus representing a significant public health concern. High heritability and ethnic-specific predisposition to PACG suggest the involvement of genetic factors in disease development. In the recent past, genetic studies have led to the successful identification of several genes and loci associated with PACG across different ethnicities. The precise cellular and molecular roles of these multiple loci in the development and progression of PACG remains to be elucidated. Nonetheless, these studies have significantly increased our understanding of the emerging cellular processes and biological pathways that might provide more significant insights into the disease’s genetic etiology and may be valuable for future clinical applications. This review aims to summarize and update the current knowledge of PACG genetics analysis research.
Collapse
Affiliation(s)
- Altaf A Kondkar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Glaucoma Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
24
|
Prasov L, Guan B, Ullah E, Archer SM, Ayres BM, Besirli CG, Wiinikka-Buesser L, Comer GM, Del Monte MA, Elner SG, Garnai SJ, Huryn LA, Johnson K, Kamat SS, Lieu P, Mian SI, Rygiel CA, Serpen JY, Pawar HS, Brooks BP, Moroi SE, Richards JE, Hufnagel RB. Novel TMEM98, MFRP, PRSS56 variants in a large United States high hyperopia and nanophthalmos cohort. Sci Rep 2020; 10:19986. [PMID: 33203948 PMCID: PMC7672112 DOI: 10.1038/s41598-020-76725-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Nanophthalmos is a rare condition defined by a small, structurally normal eye with resultant high hyperopia. While six genes have been implicated in this hereditary condition (MFRP, PRSS56, MYRF, TMEM98, CRB1,VMD2/BEST1), the relative contribution of these to nanophthalmos or to less severe high hyperopia (≥ + 5.50 spherical equivalent) has not been fully elucidated. We collected probands and families (n = 56) with high hyperopia or nanophthalmos (≤ 21.0 mm axial length). Of 53 families that passed quality control, plausible genetic diagnoses were identified in 10/53 (18.8%) by high-throughput panel or pooled exome sequencing. These include 1 TMEM98 family (1.9%), 5 MFRP families (9.4%), and 4 PRSS56 families (7.5%), with 4 additional families having single allelic hits in MFRP or PRSS56 (7.5%). A novel deleterious TMEM98 variant (NM_015544.3, c.602G>C, p.(Arg201Pro)) segregated with disease in 4 affected members of a family. Multiple novel missense and frameshift variants in MFRP and PRSS56 were identified. PRSS56 families were more likely to have choroidal folds than other solved families, while MFRP families were more likely to have retinal degeneration. Together, this study defines the prevalence of nanophthalmos gene variants in high hyperopia and nanophthalmos and indicates that a large fraction of cases remain outside of single gene coding sequences.
Collapse
Affiliation(s)
- Lev Prasov
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA. .,Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Bin Guan
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ehsan Ullah
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Steven M Archer
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Bernadete M Ayres
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Cagri G Besirli
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Laurel Wiinikka-Buesser
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Grant M Comer
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Monte A Del Monte
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Susan G Elner
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Sarah J Garnai
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Laryssa A Huryn
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kayla Johnson
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Shivani S Kamat
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Philip Lieu
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Shahzad I Mian
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Christine A Rygiel
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Jasmine Y Serpen
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA.,Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA.,Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Hemant S Pawar
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sayoko E Moroi
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA.,Department of Ophthalmology and Visual Sciences, The Ohio State University, Columbus, OH, 43212, USA
| | - Julia E Richards
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Robert B Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
25
|
Godinho G, Madeira C, Grangeia A, Neves-Cardoso P, Santos-Silva R, Brandão E, Carneiro Â, Falcão-Reis F, Estrela-Silva S. A novel MFRP gene variant in a family with posterior microphthalmos, retinitis pigmentosa, foveoschisis, and foveal hypoplasia. Ophthalmic Genet 2020; 41:474-479. [PMID: 32703043 DOI: 10.1080/13816810.2020.1795888] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND To characterize the phenotype and genotype of a syndrome associating posterior microphthalmos (PM), retinitis pigmentosa (RP), foveoschisis, and foveal hypoplasia (FH) in a consanguineous Portuguese family. MATERIALS AND METHODS Three siblings were studied and underwent comprehensive eye examinations for best-corrected visual acuity, axial length, refractive error, B-mode ultrasound, electroretinography, retinography, fluorescein angiography (FA), kinetic visual field (VF), and optical coherence tomography (OCT). Molecular analysis was performed by Sanger sequencing of the entire coding region of the MFRP gene. RESULTS All members presented nyctalopia, decreased visual acuity, and constriction of the VF, as well as bilateral shortening of the posterior ocular segment and normal anterior segment dimensions. The fundoscopy and ERG results were compatible with RP. Macular OCT analysis revealed schisis of the outer retinal layer, FH, as well as retinal and choroidal folds. We identified a homozygous mutation in intron 9 of the membrane frizzled-related protein (MFRP) gene (c.1124 + 1 G > A). CONCLUSIONS Our study shows a family with PM and RP due to a mutation in the MFRP gene. The relationship has previously been proven, but this specific mutation has never been described. These gene mutations show wide phenotypic variability, being evident in the presence of foveoschisis, retinal and choroidal folds, and FH, other than PM and RP.
Collapse
Affiliation(s)
- Gonçalo Godinho
- Department of Ophthalmology, Centro Hospitalar e Universitário São João , Porto, Portugal
| | - Carolina Madeira
- Department of Ophthalmology, Centro Hospitalar e Universitário São João , Porto, Portugal
| | - Ana Grangeia
- Department of Genetic, Centro Hospitalar e Universitário São João , Porto, Portugal
| | - Pedro Neves-Cardoso
- Department of Ophthalmology, Centro Hospitalar e Universitário São João , Porto, Portugal
| | - Renato Santos-Silva
- Department of Ophthalmology, Centro Hospitalar e Universitário São João , Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine, University of Porto , Porto, Portugal
| | - Elisete Brandão
- Department of Ophthalmology, Centro Hospitalar e Universitário São João , Porto, Portugal
| | - Ângela Carneiro
- Department of Ophthalmology, Centro Hospitalar e Universitário São João , Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine, University of Porto , Porto, Portugal
| | - Fernando Falcão-Reis
- Department of Ophthalmology, Centro Hospitalar e Universitário São João , Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine, University of Porto , Porto, Portugal
| | - Sérgio Estrela-Silva
- Department of Ophthalmology, Centro Hospitalar e Universitário São João , Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine, University of Porto , Porto, Portugal
| |
Collapse
|
26
|
Pseudodominant Nanophthalmos in a Roma Family Caused by a Novel PRSS56 Variant. J Ophthalmol 2020; 2020:6807809. [PMID: 32454992 PMCID: PMC7212339 DOI: 10.1155/2020/6807809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/29/2020] [Accepted: 02/14/2020] [Indexed: 12/25/2022] Open
Abstract
Background The aim of the study was to identify the molecular genetic cause of two different Mendelian traits with ocular involvement present in the members of a single consanguineous Czech Roma family. Methods We have performed ocular examination and review of medical records in two individuals diagnosed with nanophthalmos (proband and her father) and one individual followed for bilateral congenital cataract and microcornea (uncle of the proband). DNA of subjects with nanophthalmos was analysed by exome sequencing. Sanger sequencing was applied for targeted screening of potentially pathogenic variants and to follow segregation of identified variants within the family. Results A homozygous variant c.1509G>C; p.(Met503Ile), in PRSS56 was found in the two individuals affected with nanophthalmos. The change was absent from the gnomAD dataset, but two out of 118 control Roma individuals were also shown to be heterozygous carriers. Analysis of single nucleotide polymorphisms in linkage disequilibrium with the c.1509G>C in PRSS56 suggested a shared chromosomal segment. The nanophthalmos phenotype, characterized in detail in the younger individual, encompassed bilateral corneal steepening, retinal folds, buried optic head drusen, and restricted visual fields, but no signs of retinal dystrophy. A known pathogenic founder CTDP1 variant c.863+389C>T in a homozygous state was identified in the other family member confirming the suspected diagnosis of congenital cataracts, facial dysmorphism, and demyelinating neuropathy syndrome. Conclusions Herein, we report the first occurrence of nanophthalmos in the Roma population. We have identified pseudodominant inheritance for this phenotype caused by a novel variant in PRSS56, representing a possible founder effect. Despite advances in genetic technologies such as exome sequencing, careful phenotype evaluation in patients from an isolated population, along with an awareness of population-specific founder effects, is necessary to ensure that accurate molecular diagnoses are made.
Collapse
|
27
|
Collin GB, Gogna N, Chang B, Damkham N, Pinkney J, Hyde LF, Stone L, Naggert JK, Nishina PM, Krebs MP. Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss. Cells 2020; 9:cells9040931. [PMID: 32290105 PMCID: PMC7227028 DOI: 10.3390/cells9040931] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal degeneration (RD) leads to the impairment or loss of vision in millions of individuals worldwide, most frequently due to the loss of photoreceptor (PR) cells. Animal models, particularly the laboratory mouse, have been used to understand the pathogenic mechanisms that underlie PR cell loss and to explore therapies that may prevent, delay, or reverse RD. Here, we reviewed entries in the Mouse Genome Informatics and PubMed databases to compile a comprehensive list of monogenic mouse models in which PR cell loss is demonstrated. The progression of PR cell loss with postnatal age was documented in mutant alleles of genes grouped by biological function. As anticipated, a wide range in the onset and rate of cell loss was observed among the reported models. The analysis underscored relationships between RD genes and ciliary function, transcription-coupled DNA damage repair, and cellular chloride homeostasis. Comparing the mouse gene list to human RD genes identified in the RetNet database revealed that mouse models are available for 40% of the known human diseases, suggesting opportunities for future research. This work may provide insight into the molecular players and pathways through which PR degenerative disease occurs and may be useful for planning translational studies.
Collapse
Affiliation(s)
- Gayle B. Collin
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Navdeep Gogna
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Nattaya Damkham
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Jai Pinkney
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lillian F. Hyde
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lisa Stone
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Jürgen K. Naggert
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Patsy M. Nishina
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Correspondence: (P.M.N.); (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.)
| | - Mark P. Krebs
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Correspondence: (P.M.N.); (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.)
| |
Collapse
|
28
|
Siggs OM, Awadalla MS, Souzeau E, Staffieri SE, Kearns LS, Laurie K, Kuot A, Qassim A, Edwards TL, Coote MA, Mancel E, Walland MJ, Dondey J, Galanopoulous A, Casson RJ, Mills RA, MacArthur DG, Ruddle JB, Burdon KP, Craig JE. The genetic and clinical landscape of nanophthalmos and posterior microphthalmos in an Australian cohort. Clin Genet 2020; 97:764-769. [PMID: 32052405 DOI: 10.1111/cge.13722] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 11/30/2022]
Abstract
Nanophthalmos and posterior microphthalmos are ocular abnormalities in which both eyes are abnormally small, and typically associated with extreme hyperopia. We recruited 40 individuals from 13 kindreds with nanophthalmos or posterior microphthalmos, with 12 probands subjected to exome sequencing. Nine probands (69.2%) were assigned a genetic diagnosis, with variants in MYRF, TMEM98, MFRP, and PRSS56. Two of four PRSS56 families harbored the previously described c.1066dupC variant implicated in over half of all reported PRSS56 kindreds, with different surrounding haplotypes in each family suggesting a mutational hotspot. Individuals with a genetic diagnosis had shorter mean axial lengths and higher hyperopia than those without, with recessive forms associated with the most extreme phenotypes. These findings detail the genetic architecture of nanophthalmos and posterior microphthalmos in a cohort of predominantly European ancestry, their relative clinical phenotypes, and highlight the shared genetic architecture of rare and common disorders of refractive error.
Collapse
Affiliation(s)
- Owen M Siggs
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| | - Mona S Awadalla
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| | | | - Sandra E Staffieri
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Ophthalmology, University of Melbourne, Melbourne, Australia.,Department of Ophthalmology, Royal Children's Hospital, Melbourne, Australia
| | - Lisa S Kearns
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Kate Laurie
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| | - Abraham Kuot
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| | - Ayub Qassim
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| | - Thomas L Edwards
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Michael A Coote
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Erica Mancel
- Centre Hospitalier Territorial de Nouvelle-Calédonie, Noumea, New Caledonia
| | - Mark J Walland
- Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Joanne Dondey
- Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Anna Galanopoulous
- Discipline of Ophthalmology & Visual Sciences, University of Adelaide, Adelaide, Australia
| | - Robert J Casson
- Discipline of Ophthalmology & Visual Sciences, University of Adelaide, Adelaide, Australia
| | - Richard A Mills
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| | - Daniel G MacArthur
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts.,Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Jonathan B Ruddle
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Ophthalmology, University of Melbourne, Melbourne, Australia.,Department of Ophthalmology, Royal Children's Hospital, Melbourne, Australia
| | - Kathryn P Burdon
- Department of Ophthalmology, Flinders University, Adelaide, Australia.,Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Jamie E Craig
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| |
Collapse
|
29
|
Bacci GM, Bargiacchi S, Fortunato P, Pisaneschi E, Peluso F, Marziali E, Magli A, Giglio SR, Caputo R. Novel mutations in MFRP and PRSS56 are associated with posterior microphthalmos. Ophthalmic Genet 2020; 41:49-56. [PMID: 32118495 DOI: 10.1080/13816810.2020.1731835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: Biallelic pathogenic variants in MFRP and PRSS56 genes can be responsible for nanophthalmos (NO) or posterior microphthalmos (PM). This study describes detailed clinical and molecular findings in a series of five patients affected by PM from four unrelated families.Materials and Methods: All patients underwent a complete ophthalmological and genetic evaluation. For proper and deep phenotyping a multimodal instrumental approach was used for all cases: B-scan ultrasound, spectral domain optical coherence tomography (SD-OCT), fundus retinal imaging and anterior segment data were obtained. Molecular analysis of PRSS56 and MFRP genes was performed with Next-Generation Sequencing (NGS) methodology and segregation analysis on parents and one affected sibling was performed with Sanger sequencing.Results: A very high hyperopia of +14.00D or more was the main refractive error and macular abnormalities were identified in all patients. Axial length ranged from 15.3 mm to 17.86 mm (mean 16.58 mm) and age at first presentation ranged from 6 to 36 months (mean 18 months). Anterior chamber depth was within normal values, according to age, while total axial length was severely reduced in all patients. All our patients met the diagnostic criteria for PM. Three patients, including a pair of siblings, carried compound heterozygous mutations in the PRSS56 gene; in the other two patients, one homozygous or two compound heterozygous mutations in the MFRP gene were detected.Conclusion: Our study describes four novel mutations in the PRSS56 gene and one in the MFRP gene in patients with non-syndromic posterior microphthalmos. Proper genotype-phenotype correlation and early diagnosis could lead to good functional results.
Collapse
Affiliation(s)
| | - Sara Bargiacchi
- Genetics and Molecular Medicine Unit, Ospedale Pediatrico Meyer, Firenze, Italy
| | - Pina Fortunato
- Pediatric Ophthalmology Unit, A. Meyer Children's Hospital, Firenze, Italy
| | - Elisa Pisaneschi
- Medical Genetics Laboratory, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Francesca Peluso
- Genetics and Molecular Medicine Unit, Ospedale Pediatrico Meyer, Firenze, Italy
| | - Elisa Marziali
- Pediatric Ophthalmology Unit, A. Meyer Children's Hospital, Firenze, Italy
| | - Adriano Magli
- Department of Ophthalmology, Orthoptic and Pediatric Ophthalmology, Università degli Studi di Salerno Facoltà di Medicina e Chirurgia, Baronissi, Italy
| | - Sabrina Rita Giglio
- Genetics and Molecular Medicine Unit, Ospedale Pediatrico Meyer, Firenze, Italy
| | - Roberto Caputo
- Pediatric Ophthalmology Unit, A. Meyer Children's Hospital, Firenze, Italy
| |
Collapse
|
30
|
|
31
|
The majority of autosomal recessive nanophthalmos and posterior microphthalmia can be attributed to biallelic sequence and structural variants in MFRP and PRSS56. Sci Rep 2020; 10:1289. [PMID: 31992737 PMCID: PMC6987234 DOI: 10.1038/s41598-019-57338-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 12/09/2019] [Indexed: 01/07/2023] Open
Abstract
This study aimed to genetically and clinically characterize a unique cohort of 25 individuals from 21 unrelated families with autosomal recessive nanophthalmos (NNO) and posterior microphthalmia (MCOP) from different ethnicities. An ophthalmological assessment in all families was followed by targeted MFRP and PRSS56 testing in 20 families and whole-genome sequencing in one family. Three families underwent homozygosity mapping using SNP arrays. Eight distinct MFRP mutations were found in 10/21 families (47.6%), five of which are novel including a deletion spanning the 5' untranslated region and the first coding part of exon 1. Most cases harbored homozygous mutations (8/10), while a compound heterozygous and a monoallelic genotype were identified in the remaining ones (2/10). Six distinct PRSS56 mutations were found in 9/21 (42.9%) families, three of which are novel. Similarly, homozygous mutations were found in all but one, leaving 2/21 families (9.5%) without a molecular diagnosis. Clinically, all patients had reduced visual acuity, hyperopia, short axial length and crowded optic discs. Retinitis pigmentosa was observed in 5/10 (50%) of the MFRP group, papillomacular folds in 12/19 (63.2%) of MCOP and in 3/6 (50%) of NNO cases. A considerable phenotypic variability was observed, with no clear genotype-phenotype correlations. Overall, our study represents the largest NNO and MCOP cohort reported to date and provides a genetic diagnosis in 19/21 families (90.5%), including the first MFRP genomic rearrangement, offering opportunities for gene-based therapies in MFRP-associated disease. Finally, our study underscores the importance of sequence and copy number analysis of the MFRP and PRSS56 genes in MCOP and NNO.
Collapse
|
32
|
Guo C, Zhao Z, Chen D, He S, Sun N, Li Z, Liu J, Zhang D, Zhang J, Li J, Zhang M, Ge J, Liu X, Zhang X, Fan Z. Detection of Clinically Relevant Genetic Variants in Chinese Patients With Nanophthalmos by Trio-Based Whole-Genome Sequencing Study. Invest Ophthalmol Vis Sci 2019; 60:2904-2913. [PMID: 31266062 DOI: 10.1167/iovs.18-26275] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Nanophthalmos is a rare genetic disorder commonly characterized by a short axial length (AL) and severe hyperopia. Mutations that have been identified through Mendelian genetic analysis can only explain a fraction of nanophthalmic cases. We investigate the clinically relevant genetic variants in nanophthalmos by whole-genome sequencing (WGS), including de novo mutations (DNMs) and inherited mutations. Methods Clinically relevant genetic variants of 11 trios (11 nanophthalmic probands and their unaffected parents) from the Zhongshan Ophthalmic Center, China, were analyzed by WGS. We further screened three trios and 10 sporadic cases to identify the MYRF mutations. Results In two of 11 trios, without evidence of the presence of deleterious inherited autosomal variants, two DNMs of MYRF (c.789delC, p.S264fs and c.789dupC, p.S264fs) were identified in the probands. These loss-of-function DNMs were predicted to result in premature stop codons and protein structure damage in both probands. In addition, deleterious inherited genetic variants in PRSS56 and MFRP were found in eight probands of the other nine trios. Expanded screening found an additional MYRF DNM (c.1433G>C, p.R478P) in one trio and a stop-gain MYRF mutation (c.2956C>T, p.R986X) in one sporadic case, suggesting the recurrence of MYRF mutations in nanophthalmic patients. Conclusions This is the first trio-based WGS study for nanophthalmos, revealing the potential role of DNMs in MYRF and rare inherited genetic variants in PRSS56 and MFRP. The underlying mechanism of MYRF in the development of nanophthalmos needs to be further investigated.
Collapse
Affiliation(s)
- Congcong Guo
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhenni Zhao
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Denghui Chen
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shuxiang He
- Guangzhou KingMed Diagnostics, Guangzhou, China
| | - Nannan Sun
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhongwen Li
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jiafan Liu
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Dandan Zhang
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jiamin Zhang
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jianlong Li
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Miao Zhang
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xing Liu
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaoling Zhang
- Section of Biomedical Genetics, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States
| | - Zhigang Fan
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Siggs OM, Souzeau E, Breen J, Qassim A, Zhou T, Dubowsky A, Ruddle JB, Craig JE. Autosomal dominant nanophthalmos and high hyperopia associated with a C-terminal frameshift variant in MYRF. Mol Vis 2019; 25:527-534. [PMID: 31700225 PMCID: PMC6817736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 09/19/2019] [Indexed: 10/27/2022] Open
Abstract
Purpose Nanophthalmos is a rare subtype of microphthalmia associated with high hyperopia and an increased risk of angle-closure glaucoma. We investigated the genetic cause of nanophthalmos and high hyperopia in an autosomal dominant kindred. Methods A proband with short axial length, high hyperopia, and dextrocardia was subjected to exome sequencing. Human and rodent gene expression data sets were used to investigate the expression of relevant genes. Results We identified a segregating heterozygous frameshift variant at the 3' end of the penultimate exon of MYRF. Using Myc-MYRF chromatin immunoprecipitation data from rat oligodendrocytes, MYRF was found to bind immediately upstream of the transcriptional start site of Tmem98, a gene that itself has been implicated in autosomal dominant nanophthalmos. MYRF and TMEM98 were found to be expressed in the human retina, with a similar pattern of expression across several dissected human eye tissues. Conclusions C-terminal variants in MYRF, which are expected to escape nonsense-mediated decay, represent a rare cause of autosomal dominant nanophthalmos with or without dextrocardia or congenital diaphragmatic hernia.
Collapse
Affiliation(s)
- Owen M. Siggs
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| | - Emmanuelle Souzeau
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| | - James Breen
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Ayub Qassim
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| | - Tiger Zhou
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| | | | - Jonathan B. Ruddle
- Department of Ophthalmology, University of Melbourne, Melbourne, Victoria, Australia
| | - Jamie E. Craig
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
34
|
Cross SH, Mckie L, Keighren M, West K, Thaung C, Davey T, Soares DC, Sanchez-Pulido L, Jackson IJ. Missense Mutations in the Human Nanophthalmos Gene TMEM98 Cause Retinal Defects in the Mouse. Invest Ophthalmol Vis Sci 2019; 60:2875-2887. [PMID: 31266059 PMCID: PMC6986908 DOI: 10.1167/iovs.18-25954] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose We previously found a dominant mutation, Rwhs, causing white spots on the retina accompanied by retinal folds. Here we identify the mutant gene to be Tmem98. In humans, mutations in the orthologous gene cause nanophthalmos. We modeled these mutations in mice and characterized the mutant eye phenotypes of these and Rwhs. Methods The Rwhs mutation was identified to be a missense mutation in Tmem98 by genetic mapping and sequencing. The human TMEM98 nanophthalmos missense mutations were made in the mouse gene by CRISPR-Cas9. Eyes were examined by indirect ophthalmoscopy and the retinas imaged using a retinal camera. Electroretinography was used to study retinal function. Histology, immunohistochemistry, and electron microscopy techniques were used to study adult eyes. Results An I135T mutation of Tmem98 causes the dominant Rwhs phenotype and is perinatally lethal when homozygous. Two dominant missense mutations of TMEM98, A193P and H196P, are associated with human nanophthalmos. In the mouse these mutations cause recessive retinal defects similar to the Rwhs phenotype, either alone or in combination with each other, but do not cause nanophthalmos. The retinal folds did not affect retinal function as assessed by electroretinography. Within the folds there was accumulation of disorganized outer segment material as demonstrated by immunohistochemistry and electron microscopy, and macrophages had infiltrated into these regions. Conclusions Mutations in the mouse orthologue of the human nanophthalmos gene TMEM98 do not result in small eyes. Rather, there is localized disruption of the laminar structure of the photoreceptors.
Collapse
Affiliation(s)
- Sally H. Cross
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Lisa Mckie
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Margaret Keighren
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Katrine West
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Caroline Thaung
- Moorfields Eye Hospital NHS Foundation Trust, 162 City Road, London EC1V 2PD, United Kingdom
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, United Kingdom
| | - Tracey Davey
- Electron Microscopy Research Services, Newcastle University, Newcastle NE2 4HH, United Kingdom
| | - Dinesh C. Soares
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Luis Sanchez-Pulido
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Ian J. Jackson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| |
Collapse
|
35
|
Novel truncation mutations in MYRF cause autosomal dominant high hyperopia mapped to 11p12-q13.3. Hum Genet 2019; 138:1077-1090. [PMID: 31172260 PMCID: PMC6745028 DOI: 10.1007/s00439-019-02039-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/04/2019] [Indexed: 12/15/2022]
Abstract
High hyperopia is a common and severe form of refractive error. Genetic factors play important roles in the development of high hyperopia but the exact gene responsible for this condition is mostly unknown. We identified a large Chinese family with autosomal dominant high hyperopia. A genome-wide linkage scan mapped the high hyperopia to chromosome 11p12-q13.3, with maximum log of the odds scores of 4.68 at theta = 0 for D11S987. Parallel whole-exome sequencing detected a novel c.3377delG (p.Gly1126Valfs*31) heterozygous mutation in the MYRF gene within the linkage interval. Whole-exome sequencing in other 121 probands with high hyperopia identified additional novel mutations in MYRF within two other families: a de novo c.3274_3275delAG (p.Leu1093Profs*22) heterozygous mutation and a c.3194+2T>C heterozygous mutation. All three mutations are located in the C-terminal region of MYRF and are predicted to result in truncation of that portion. Two patients from two of the three families developed angle-closure glaucoma. These three mutations were present in neither the ExAC database nor our in-house whole-exome sequencing data from 3280 individuals. No other truncation mutations in MYRF were detected in the 3280 individuals. Knockdown of myrf resulted in small eye size in zebrafish. These evidence all support that truncation mutations in the C-terminal region of MYRF are responsible for autosomal dominant high hyperopia in these families. Our results may provide useful clues for further understanding the functional role of the C-terminal region of this critical myelin regulatory factor, as well as the molecular pathogenesis of high hyperopia and its associated angle-closure glaucoma.
Collapse
|
36
|
Liu B, Calton MA, Abell NS, Benchorin G, Gloudemans MJ, Chen M, Hu J, Li X, Balliu B, Bok D, Montgomery SB, Vollrath D. Genetic analyses of human fetal retinal pigment epithelium gene expression suggest ocular disease mechanisms. Commun Biol 2019; 2:186. [PMID: 31123710 PMCID: PMC6527609 DOI: 10.1038/s42003-019-0430-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
The retinal pigment epithelium (RPE) serves vital roles in ocular development and retinal homeostasis but has limited representation in large-scale functional genomics datasets. Understanding how common human genetic variants affect RPE gene expression could elucidate the sources of phenotypic variability in selected monogenic ocular diseases and pinpoint causal genes at genome-wide association study (GWAS) loci. We interrogated the genetics of gene expression of cultured human fetal RPE (fRPE) cells under two metabolic conditions and discovered hundreds of shared or condition-specific expression or splice quantitative trait loci (e/sQTLs). Co-localizations of fRPE e/sQTLs with age-related macular degeneration (AMD) and myopia GWAS data suggest new candidate genes, and mechanisms by which a common RDH5 allele contributes to both increased AMD risk and decreased myopia risk. Our study highlights the unique transcriptomic characteristics of fRPE and provides a resource to connect e/sQTLs in a critical ocular cell type to monogenic and complex eye disorders.
Collapse
Affiliation(s)
- Boxiang Liu
- Department of Biology, Stanford University, Stanford, CA 94305 USA
| | - Melissa A. Calton
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Nathan S. Abell
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Gillie Benchorin
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Michael J. Gloudemans
- Program in Biomedical Informatics, Stanford University School of Medicine, Stanford, 94305 CA USA
| | - Ming Chen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Jane Hu
- Department of Ophthalmology, Jules Stein Eye Institute, UCLA, Los Angeles, 90095 CA USA
| | - Xin Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Brunilda Balliu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Dean Bok
- Department of Ophthalmology, Jules Stein Eye Institute, UCLA, Los Angeles, 90095 CA USA
| | - Stephen B. Montgomery
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Douglas Vollrath
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| |
Collapse
|
37
|
Garnai SJ, Brinkmeier ML, Emery B, Aleman TS, Pyle LC, Veleva-Rotse B, Sisk RA, Rozsa FW, Ozel AB, Li JZ, Moroi SE, Archer SM, Lin CM, Sheskey S, Wiinikka-Buesser L, Eadie J, Urquhart JE, Black GC, Othman MI, Boehnke M, Sullivan SA, Skuta GL, Pawar HS, Katz AE, Huryn LA, Hufnagel RB, Camper SA, Richards JE, Prasov L. Variants in myelin regulatory factor (MYRF) cause autosomal dominant and syndromic nanophthalmos in humans and retinal degeneration in mice. PLoS Genet 2019; 15:e1008130. [PMID: 31048900 PMCID: PMC6527243 DOI: 10.1371/journal.pgen.1008130] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/20/2019] [Accepted: 04/09/2019] [Indexed: 01/11/2023] Open
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.
Collapse
Affiliation(s)
- Sarah J. Garnai
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Michelle L. Brinkmeier
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Tomas S. Aleman
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- Scheie Eye Institute, Department of Ophthalmology, Philadelphia, PA, United States of America
| | - Louise C. Pyle
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Biliana Veleva-Rotse
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Robert A. Sisk
- Cincinnati Eye Institute, Cincinnati, Ohio, United States of America
| | - Frank W. Rozsa
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Molecular and Behavior Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Ayse Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Jun Z. Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Sayoko E. Moroi
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Steven M. Archer
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Cheng-mao Lin
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Sarah Sheskey
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Laurel Wiinikka-Buesser
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - James Eadie
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Jill E. Urquhart
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, St Mary’s Hospital, Manchester, United Kingdom
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Graeme C.M. Black
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, St Mary’s Hospital, Manchester, United Kingdom
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Mohammad I. Othman
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Scot A. Sullivan
- Dean McGee Eye Institute, Department of Ophthalmology, University of Oklahoma, Oklahoma City, OK
| | - Gregory L. Skuta
- Dean McGee Eye Institute, Department of Ophthalmology, University of Oklahoma, Oklahoma City, OK
| | - Hemant S. Pawar
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Alexander E. Katz
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Laryssa A. Huryn
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Robert B. Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States of America
| | | | - Sally A. Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Julia E. Richards
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Lev Prasov
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
38
|
Genetics of anophthalmia and microphthalmia. Part 1: Non-syndromic anophthalmia/microphthalmia. Hum Genet 2019; 138:799-830. [PMID: 30762128 DOI: 10.1007/s00439-019-01977-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/30/2019] [Indexed: 12/22/2022]
Abstract
Eye formation is the result of coordinated induction and differentiation processes during embryogenesis. Disruption of any one of these events has the potential to cause ocular growth and structural defects, such as anophthalmia and microphthalmia (A/M). A/M can be isolated or occur with systemic anomalies, when they may form part of a recognizable syndrome. Their etiology includes genetic and environmental factors; several hundred genes involved in ocular development have been identified in humans or animal models. In humans, around 30 genes have been repeatedly implicated in A/M families, although many other genes have been described in single cases or families, and some genetic syndromes include eye anomalies occasionally as part of a wider phenotype. As a result of this broad genetic heterogeneity, with one or two notable exceptions, each gene explains only a small percentage of cases. Given the overlapping phenotypes, these genes can be most efficiently tested on panels or by whole exome/genome sequencing for the purposes of molecular diagnosis. However, despite whole exome/genome testing more than half of patients currently remain without a molecular diagnosis. The proportion of undiagnosed cases is even higher in those individuals with unilateral or milder phenotypes. Furthermore, even when a strong gene candidate is available for a patient, issues of incomplete penetrance and germinal mosaicism make diagnosis and genetic counseling challenging. In this review, we present the main genes implicated in non-syndromic human A/M phenotypes and, for practical purposes, classify them according to the most frequent or predominant phenotype each is associated with. Our intention is that this will allow clinicians to rank and prioritize their molecular analyses and interpretations according to the phenotypes of their patients.
Collapse
|
39
|
Chekuri A, Sahu B, Chavali VRM, Voronchikhina M, Soto-Hermida A, Suk JJ, Alapati AN, Bartsch DU, Ayala-Ramirez R, Zenteno JC, Dinculescu A, Jablonski MM, Borooah S, Ayyagari R. Long-Term Effects of Gene Therapy in a Novel Mouse Model of Human MFRP-Associated Retinopathy. Hum Gene Ther 2019; 30:632-650. [PMID: 30499344 DOI: 10.1089/hum.2018.192] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Patients harboring homozygous c.498_499insC mutations in MFRP demonstrate hyperopia, microphthalmia, retinitis pigmentosa, retinal pigment epithelial atrophy, variable degrees of foveal edema, and optic disc drusen. The disease phenotype is variable, however, with some patients maintaining good central vision and cone function till late in the disease. A knock-in mouse model with the c.498_499insC mutation in Mfrp (Mfrp KI/KI) was developed to understand the effects of these mutations in the retina. The model shares many of the features of human clinical disease, including reduced axial length, hyperopia, retinal degeneration, retinal pigment epithelial atrophy, and decreased electrophysiological responses. In addition, the eyes of these mice had a significantly greater refractive error (p < 0.01) when compared to age-matched wild-type control animals. Administration of recombinant adeno-associated virus-mediated Mfrp gene therapy significantly prevented thinning from retinal neurodegeneration (p < 0.005) and preserved retinal electrophysiology (p < 0.001) when treated eyes were compared to contralateral sham-treated control eyes. The Mfrp KI/KI mice will serve as a useful tool to model human disease and point to a potential gene therapeutic approach for patients with preserved vision and electrophysiological responses in MFRP-related retinopathy.
Collapse
Affiliation(s)
- Anil Chekuri
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California
| | - Bhubanananda Sahu
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California.,2 Department of Ophthalmology and Visual sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Venkata Ramana Murthy Chavali
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California.,3 Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marina Voronchikhina
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California
| | - Angel Soto-Hermida
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California
| | - John J Suk
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California
| | - Akhila N Alapati
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California
| | - Dirk-Uwe Bartsch
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California
| | - Raul Ayala-Ramirez
- 4 Department of Genetics-Research Unit, Institute of Ophthalmology, Conde de Valenciana, Mexico City, Mexico
| | - Juan C Zenteno
- 4 Department of Genetics-Research Unit, Institute of Ophthalmology, Conde de Valenciana, Mexico City, Mexico.,5 Department of Biochemistry, Faculty of Medicine, UNAM, Mexico City, Mexico
| | - Astra Dinculescu
- 6 Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida
| | - Monica M Jablonski
- 7 Department of Ophthalmology, The University of Tennessee Health Science Center, Hamilton Eye Institute, University of Tennessee, Memphis, Tennessee
| | - Shyamanga Borooah
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California
| | - Radha Ayyagari
- 1 Shiley Eye Institute, University of California San Diego, La Jolla, California
| |
Collapse
|
40
|
Abstract
The term nanophthalmos refers to a clinically small eye that appears morphologically normal. A nanophthalmos is characterized by hyperopia but can also be associated with various secondary pathologies, such as angle-closure glaucoma. In particular, the perioperative risks associated with a nanophthalmic eye necessitate examination of the anatomical characteristics, which can result from the disproportional size of intraocular tissues despite structural normality. These include a small anterior chamber depth, scleral thickening and anomalies of the vein plexus, which are predisposing factors for the formation of angle-closure glaucoma. The resulting therapeutic challenges in the nanophthalmic eye can be countered with iridectomy, lensectomy, vitrectomy and cyclophotocoagulation. The definition, genetics and clinical findings of nanophthalmos are discussed with a focus on the complication of glaucoma and its treatment.
Collapse
Affiliation(s)
- A Lappas
- Zentrum für Augenheilkunde, Uniklinik Köln, Joseph-Stelzmann Str. 9, 50931, Köln, Deutschland.
| | - A Rosentreter
- Klinik für Augenheilkunde, Helios Universitätsklinikum Wuppertal, Wuppertal, Deutschland
| | - A Hedergott
- Zentrum für Augenheilkunde, Uniklinik Köln, Joseph-Stelzmann Str. 9, 50931, Köln, Deutschland
| | - T S Dietlein
- Zentrum für Augenheilkunde, Uniklinik Köln, Joseph-Stelzmann Str. 9, 50931, Köln, Deutschland
| |
Collapse
|
41
|
Association of Genes implicated in primary angle-closure Glaucoma and the ocular biometric parameters of anterior chamber depth and axial length in a northern Chinese population. BMC Ophthalmol 2018; 18:271. [PMID: 30348125 PMCID: PMC6198425 DOI: 10.1186/s12886-018-0934-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/05/2018] [Indexed: 11/18/2022] Open
Abstract
Background The membrane frizzled-related protein (MFRP) gene is involved in axial length (AL) regulation and MFRP mutations cause nanophthalmos; also, the hepatocyte growth factor (HGF) gene is reported to result in morphologic changes of the anterior segment and abnormal aqueous regulation that increases the risk of primary angle-closure glaucoma (PACG), while the zinc ring finger 3 (ZNRF3) gene is associated with AL. The present study investigated the association of single nucleotide polymorphisms (SNPs) in ZNRF3, HGF and MFRP with PACG in a northern Chinese population, as well as the association of these SNPs with the ocular biometric parameters of anterior chamber depth (ACD) and AL. Methods A total of 500 PACG patients and 720 controls were recruited. All individuals were genotyped for 12 SNPs in three genes (rs7290117, rs2179129, rs4823006 and rs3178915 in ZNRF3; rs5745718, rs12536657, rs12540393, rs17427817 and rs3735520 in HGF, rs2510143, rs36015759 and rs3814762 in MFRP) using an improved multiplex ligation detection reaction (iMLDR) technique. Genotypic distribution was analyzed for Hardy-Weinberg equilibrium. Differences in the allelic and genotypic frequencies were evaluated and adjusted by age and sex. Linkage disequilibrium (LD) patterns were tested and haplotype analysis was conducted by a logistic regression model. Generalized estimation equation (GEE) analysis was conducted using SPSS for primary association testing between genotypes and ocular biometric parameters. Bonferroni corrections for multiple comparisons were performed, and the statistical power was calculated by power and sample size calculations. Results The rs7290117 SNP in ZNRF3 was significantly associated with the AL, with a p-value of 0.002. We did not observe any significant associations between the SNPs and PACG or ACD. In a stratification analysis by ethnicity, rs12540393 and rs17427817 in HGF showed a nominal association with PACG in the Hui cohort, although significance was lost after correction. Conclusions The present study suggests rs7290117 in ZNRF3 may be involved in the regulation of AL, though our results do not support a contribution of the SNPs we tested in ZNRF3, HGF and MFRP to PACG in northern Chinese people. Further studies in a larger population are warranted to confirm this conclusion.
Collapse
|
42
|
Saffren BD, Capasso JE, Zanolli M, Levin AV. Ocular manifestations of Emanuel syndrome. Am J Med Genet A 2018; 176:1964-1967. [PMID: 30178914 DOI: 10.1002/ajmg.a.40361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/24/2018] [Accepted: 05/21/2018] [Indexed: 11/07/2022]
Abstract
Emanuel syndrome is caused by a supernumerary der(22)t(11;22) and typically manifests with intellectual disability and craniofacial dysmorphism. Ocular abnormalities have infrequently been described. We report a 36-year-old man with severe intellectual disability, aphasia, and facial dysmorphism, with high myopia and juvenile open angle glaucoma (JOAG). Microarray analysis results included 47,XY,+der(22)t(11;22)(q23;q11.2), and a 269 kb deletion of 7q31.33(125,898,014-126,166,829). Two candidate genes were identified as possible etiologies for the ocular pathologies in our patient: a MFRP duplication on chromosome 11, which may play a role in high myopia and dysregulation of emmetropization, and a GRM8 deletion on chromosome 7, which may cause glutamate-induced excitotoxicity and therefore have a role in the development of JOAG, unrelated to the Emanuel syndrome genotype. We provide the first detailed description these ocular abnormalities in a patient with Emmanuel syndrome.
Collapse
Affiliation(s)
- Brooke D Saffren
- Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | | | | | - Alex V Levin
- Wills Eye Hospital, Philadelphia, Pennsylvania.,Sydney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
Matías-Pérez D, García-Montaño LA, Cruz-Aguilar M, García-Montalvo IA, Nava-Valdéz J, Barragán-Arevalo T, Villanueva-Mendoza C, Villarroel CE, Guadarrama-Vallejo C, la Cruz RVD, Chacón-Camacho O, Zenteno JC. Identification of novel pathogenic variants and novel gene-phenotype correlations in Mexican subjects with microphthalmia and/or anophthalmia by next-generation sequencing. J Hum Genet 2018; 63:1169-1180. [PMID: 30181649 DOI: 10.1038/s10038-018-0504-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/21/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023]
Abstract
Severe congenital eye malformations, particularly microphthalmia and anophthalmia, are one of the main causes of visual handicap worldwide. They can arise from multifactorial, chromosomal, or monogenic factors and can be associated with extensive clinical variability. Genetic analysis of individuals with these defects has allowed the recognition of dozens of genes whose mutations lead to disruption of normal ocular embryonic development. Recent application of next generation sequencing (NGS) techniques for genetic screening of patients with congenital eye defects has greatly improved the recognition of monogenic cases. In this study, we applied clinical exome NGS to a group of 14 Mexican patients (including 7 familial and 7 sporadic cases) with microphthalmia and/or anophthalmia. Causal or likely causal pathogenic variants were demonstrated in ~60% (8 out of 14 patients) individuals. Seven out of 8 different identified mutations occurred in well-known microphthalmia/anophthalmia genes (OTX2, VSX2, MFRP, VSX1) or in genes associated with syndromes that include ocular defects (CHD7, COL4A1) (including two instances of CHD7 pathogenic variants). A single pathogenic variant was identified in PIEZO2, a gene that was not previously associated with isolated ocular defects. NGS efficiently identified the genetic etiology of microphthalmia/anophthalmia in ~60% of cases included in this cohort, the first from Mexican origin analyzed to date. The molecular defects identified through clinical exome sequencing in this study expands the phenotypic spectra of CHD7-associated disorders and implicate PIEZO2 as a candidate gene for major eye developmental defects.
Collapse
Affiliation(s)
| | - Leopoldo A García-Montaño
- Department of Genetics-Research Unit, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | - Marisa Cruz-Aguilar
- Department of Genetics-Research Unit, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | | | - Jessica Nava-Valdéz
- Department of Genetics-Research Unit, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | - Tania Barragán-Arevalo
- Department of Human Genetics, National Institute of Pediatrics of Mexico, Mexico City, Mexico
| | - Cristina Villanueva-Mendoza
- Department of Genetics, Hospital "Dr. Luis Sanchez Bulnes", Asociación Para Evitar la Ceguera en México, Mexico City, Mexico
| | - Camilo E Villarroel
- Department of Human Genetics, National Institute of Pediatrics of Mexico, Mexico City, Mexico
| | - Clavel Guadarrama-Vallejo
- Department of Genetics-Research Unit, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | - Rocío Villafuerte-de la Cruz
- Ciencias Basicas, Escuela de Medicina, Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, NL, Mexico
| | - Oscar Chacón-Camacho
- Department of Genetics-Research Unit, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | - Juan C Zenteno
- Department of Genetics-Research Unit, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico. .,Department of Biochemistry, Faculty of Medicine, UNAM, Mexico City, Mexico.
| |
Collapse
|
44
|
Genome-wide association study of intraocular pressure uncovers new pathways to glaucoma. Nat Genet 2018; 50:1067-1071. [PMID: 30054594 DOI: 10.1038/s41588-018-0176-y] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 06/13/2018] [Indexed: 12/14/2022]
Abstract
Intraocular pressure (IOP) is currently the sole modifiable risk factor for primary open-angle glaucoma (POAG), one of the leading causes of blindness worldwide1. Both IOP and POAG are highly heritable2. We report a combined analysis of participants from the UK Biobank (n = 103,914) and previously published data from the International Glaucoma Genetic Consortium (n = 29,578)3,4 that identified 101 statistically independent genome-wide-significant SNPs for IOP, 85 of which have not been previously reported4-12. We examined these SNPs in 11,018 glaucoma cases and 126,069 controls, and 53 SNPs showed evidence of association. Gene-based tests implicated an additional 22 independent genes associated with IOP. We derived an allele score based on the IOP loci and loci influencing optic nerve head morphology. In 1,734 people with advanced glaucoma and 2,938 controls, participants in the top decile of the allele score were at increased risk (odds ratio (OR) = 5.6; 95% confidence interval (CI): 4.1-7.6) of glaucoma relative to the bottom decile.
Collapse
|
45
|
Nanophthalmos: A Review of the Clinical Spectrum and Genetics. J Ophthalmol 2018; 2018:2735465. [PMID: 29862063 PMCID: PMC5971257 DOI: 10.1155/2018/2735465] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/20/2018] [Accepted: 04/08/2018] [Indexed: 11/28/2022] Open
Abstract
Nanophthalmos is a clinical spectrum of disorders with a phenotypically small but structurally normal eye. These disorders present significant clinical challenges to ophthalmologists due to a high rate of secondary angle-closure glaucoma, spontaneous choroidal effusions, and perioperative complications with cataract and retinal surgeries. Nanophthalmos may present as a sporadic or familial disorder, with autosomal-dominant or recessive inheritance. To date, five genes (i.e., MFRP, TMEM98, PRSS56, BEST1, and CRB1) and two loci have been implicated in familial forms of nanophthalmos. Here, we review the definition of nanophthalmos, the clinical and pathogenic features of the condition, and the genetics of this disorder.
Collapse
|
46
|
Gräf S, Haimel M, Bleda M, Hadinnapola C, Southgate L, Li W, Hodgson J, Liu B, Salmon RM, Southwood M, Machado RD, Martin JM, Treacy CM, Yates K, Daugherty LC, Shamardina O, Whitehorn D, Holden S, Aldred M, Bogaard HJ, Church C, Coghlan G, Condliffe R, Corris PA, Danesino C, Eyries M, Gall H, Ghio S, Ghofrani HA, Gibbs JSR, Girerd B, Houweling AC, Howard L, Humbert M, Kiely DG, Kovacs G, MacKenzie Ross RV, Moledina S, Montani D, Newnham M, Olschewski A, Olschewski H, Peacock AJ, Pepke-Zaba J, Prokopenko I, Rhodes CJ, Scelsi L, Seeger W, Soubrier F, Stein DF, Suntharalingam J, Swietlik EM, Toshner MR, van Heel DA, Vonk Noordegraaf A, Waisfisz Q, Wharton J, Wort SJ, Ouwehand WH, Soranzo N, Lawrie A, Upton PD, Wilkins MR, Trembath RC, Morrell NW. Identification of rare sequence variation underlying heritable pulmonary arterial hypertension. Nat Commun 2018; 9:1416. [PMID: 29650961 PMCID: PMC5897357 DOI: 10.1038/s41467-018-03672-4] [Citation(s) in RCA: 241] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 03/02/2018] [Indexed: 12/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disorder with a poor prognosis. Deleterious variation within components of the transforming growth factor-β pathway, particularly the bone morphogenetic protein type 2 receptor (BMPR2), underlies most heritable forms of PAH. To identify the missing heritability we perform whole-genome sequencing in 1038 PAH index cases and 6385 PAH-negative control subjects. Case-control analyses reveal significant overrepresentation of rare variants in ATP13A3, AQP1 and SOX17, and provide independent validation of a critical role for GDF2 in PAH. We demonstrate familial segregation of mutations in SOX17 and AQP1 with PAH. Mutations in GDF2, encoding a BMPR2 ligand, lead to reduced secretion from transfected cells. In addition, we identify pathogenic mutations in the majority of previously reported PAH genes, and provide evidence for further putative genes. Taken together these findings contribute new insights into the molecular basis of PAH and indicate unexplored pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom.
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, United Kingdom.
- NIHR BioResource-Rare Diseases, Cambridge, CB2 0PT, United Kingdom.
| | - Matthias Haimel
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, United Kingdom
- NIHR BioResource-Rare Diseases, Cambridge, CB2 0PT, United Kingdom
| | - Marta Bleda
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Charaka Hadinnapola
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Laura Southgate
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, SW17 0RE, United Kingdom
- Division of Genetics & Molecular Medicine, King's College London, London, WC2R 2LS, United Kingdom
| | - Wei Li
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Joshua Hodgson
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Bin Liu
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Richard M Salmon
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Mark Southwood
- Royal Papworth Hospital, Papworth Everard, Cambridge, CB23 3RE, United Kingdom
| | - Rajiv D Machado
- Institute of Medical and Biomedical Education, St George's University of London, London, SW17 0RE, United Kingdom
| | - Jennifer M Martin
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, United Kingdom
- NIHR BioResource-Rare Diseases, Cambridge, CB2 0PT, United Kingdom
| | - Carmen M Treacy
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
- Royal Papworth Hospital, Papworth Everard, Cambridge, CB23 3RE, United Kingdom
| | - Katherine Yates
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, United Kingdom
- NIHR BioResource-Rare Diseases, Cambridge, CB2 0PT, United Kingdom
| | - Louise C Daugherty
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, United Kingdom
- NIHR BioResource-Rare Diseases, Cambridge, CB2 0PT, United Kingdom
| | - Olga Shamardina
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, United Kingdom
- NIHR BioResource-Rare Diseases, Cambridge, CB2 0PT, United Kingdom
| | - Deborah Whitehorn
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, United Kingdom
- NIHR BioResource-Rare Diseases, Cambridge, CB2 0PT, United Kingdom
| | - Simon Holden
- Addenbrooke's Hospital, Cambridge, CB2 0QQ, United Kingdom
| | | | - Harm J Bogaard
- VU University Medical Center, Amsterdam, 1007 MB, The Netherlands
| | - Colin Church
- Golden Jubilee National Hospital, Glasgow, G81 4DY, United Kingdom
| | - Gerry Coghlan
- Royal Free Hospital, London, NW3 2QG, United Kingdom
| | - Robin Condliffe
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, S10 2JF, United Kingdom
| | - Paul A Corris
- University of Newcastle, Newcastle, NE1 7RU, United Kingdom
| | - Cesare Danesino
- Department of Molecular Medicine, University of Pavia, Pavia, 27100, Italy
- Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy
| | - Mélanie Eyries
- Département de génétique, hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, and UMR_S 1166-ICAN, INSERM, UPMC Sorbonne Universités, Paris, 75252, France
| | - Henning Gall
- University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary System (ECCCPS), Giessen, 35392, Germany
| | - Stefano Ghio
- Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy
| | - Hossein-Ardeschir Ghofrani
- University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary System (ECCCPS), Giessen, 35392, Germany
- Imperial College London, London, SW7 2AZ, United Kingdom
| | - J Simon R Gibbs
- National Heart & Lung Institute, Imperial College London, London, SW3 6LY, United Kingdom
| | - Barbara Girerd
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay; AP-HP, Service de Pneumologie, Centre de référence de l'hypertension pulmonaire; INSERM UMR_S 999, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, 94270, France
| | | | - Luke Howard
- Imperial College London, London, SW7 2AZ, United Kingdom
| | - Marc Humbert
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay; AP-HP, Service de Pneumologie, Centre de référence de l'hypertension pulmonaire; INSERM UMR_S 999, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, 94270, France
| | - David G Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, S10 2JF, United Kingdom
| | - Gabor Kovacs
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, 8010, Austria
- Medical University of Graz, Graz, 8036, Austria
| | | | - Shahin Moledina
- Great Ormond Street Hospital, London, WC1N 3JH, United Kingdom
| | - David Montani
- Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay; AP-HP, Service de Pneumologie, Centre de référence de l'hypertension pulmonaire; INSERM UMR_S 999, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, 94270, France
| | - Michael Newnham
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, 8010, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, 8010, Austria
- Medical University of Graz, Graz, 8036, Austria
| | - Andrew J Peacock
- Golden Jubilee National Hospital, Glasgow, G81 4DY, United Kingdom
| | - Joanna Pepke-Zaba
- Royal Papworth Hospital, Papworth Everard, Cambridge, CB23 3RE, United Kingdom
| | | | | | - Laura Scelsi
- Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy
| | - Werner Seeger
- University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL) and of the Excellence Cluster Cardio-Pulmonary System (ECCCPS), Giessen, 35392, Germany
| | - Florent Soubrier
- Département de génétique, hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, and UMR_S 1166-ICAN, INSERM, UPMC Sorbonne Universités, Paris, 75252, France
| | - Dan F Stein
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Jay Suntharalingam
- Royal United Hospitals Bath NHS Foundation Trust, Bath, BA1 3NG, United Kingdom
| | - Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Mark R Toshner
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - David A van Heel
- Blizard Institute, Queen Mary University of London, London, E1 2AT, United Kingdom
| | | | - Quinten Waisfisz
- VU University Medical Center, Amsterdam, 1007 MB, The Netherlands
| | - John Wharton
- Imperial College London, London, SW7 2AZ, United Kingdom
| | - Stephen J Wort
- Imperial College London, London, SW7 2AZ, United Kingdom
- Royal Brompton Hospital, London, SW3 6NP, United Kingdom
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, United Kingdom
- NIHR BioResource-Rare Diseases, Cambridge, CB2 0PT, United Kingdom
| | - Nicole Soranzo
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, United Kingdom
| | - Allan Lawrie
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, United Kingdom
| | - Paul D Upton
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | | | - Richard C Trembath
- Division of Genetics & Molecular Medicine, King's College London, London, WC2R 2LS, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom.
- NIHR BioResource-Rare Diseases, Cambridge, CB2 0PT, United Kingdom.
| |
Collapse
|
47
|
Paylakhi S, Labelle-Dumais C, Tolman NG, Sellarole MA, Seymens Y, Saunders J, Lakosha H, deVries WN, Orr AC, Topilko P, John SWM, Nair KS. Müller glia-derived PRSS56 is required to sustain ocular axial growth and prevent refractive error. PLoS Genet 2018. [PMID: 29529029 PMCID: PMC5864079 DOI: 10.1371/journal.pgen.1007244] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A mismatch between optical power and ocular axial length results in refractive errors. Uncorrected refractive errors constitute the most common cause of vision loss and second leading cause of blindness worldwide. Although the retina is known to play a critical role in regulating ocular growth and refractive development, the precise factors and mechanisms involved are poorly defined. We have previously identified a role for the secreted serine protease PRSS56 in ocular size determination and PRSS56 variants have been implicated in the etiology of both hyperopia and myopia, highlighting its importance in refractive development. Here, we use a combination of genetic mouse models to demonstrate that Prss56 mutations leading to reduced ocular size and hyperopia act via a loss of function mechanism. Using a conditional gene targeting strategy, we show that PRSS56 derived from Müller glia contributes to ocular growth, implicating a new retinal cell type in ocular size determination. Importantly, we demonstrate that persistent activity of PRSS56 is required during distinct developmental stages spanning the pre- and post-eye opening periods to ensure optimal ocular growth. Thus, our mouse data provide evidence for the existence of a molecule contributing to both the prenatal and postnatal stages of human ocular growth. Finally, we demonstrate that genetic inactivation of Prss56 rescues axial elongation in a mouse model of myopia caused by a null mutation in Egr1. Overall, our findings identify PRSS56 as a potential therapeutic target for modulating ocular growth aimed at preventing or slowing down myopia, which is reaching epidemic proportions.
Collapse
Affiliation(s)
- Seyyedhassan Paylakhi
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Nicholas G Tolman
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, United States of America
| | - Michael A. Sellarole
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Yusef Seymens
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Joseph Saunders
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Hesham Lakosha
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Wilhelmine N. deVries
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, United States of America
| | - Andrew C. Orr
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Piotr Topilko
- Ecole Normale Supérieure, Institut de Biologie de l’ENS (IBENS), and Inserm U1024, and CNRS UMR 8197, Paris, France
| | - Simon WM. John
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, United States of America
- Department of Ophthalmology, Tufts University School of Medicine Boston, MA, United States of America
| | - K. Saidas Nair
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
- Department of Anatomy, University of California, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
48
|
Wiggs JL, Pasquale LR. Genetics of glaucoma. Hum Mol Genet 2017; 26:R21-R27. [PMID: 28505344 DOI: 10.1093/hmg/ddx184] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022] Open
Abstract
Genetic and genomic studies, including genome-wide association studies (GWAS) have accelerated the discovery of genes contributing to glaucoma, the leading cause of irreversible blindness world-wide. Glaucoma can occur at all ages, with Mendelian inheritance typical for the rare early onset disease (before age 40) and complex inheritance evident in common adult-onset forms of disease. Recent studies have suggested possible therapeutic targets for some patients with early-onset glaucoma based on the molecular and cellular events caused by MYOC, OPTN and TBK1 mutations. Diagnostic genetic tests using early-onset glaucoma genes are also proving useful for pre-symptomatic disease detection and genetic counseling. Recent GWAS completed for three types of common adult-onset glaucoma have identified novel loci for POAG (primary-open-angle glaucoma) (ABCA1, AFAP1, GMDS, PMM2, TGFBR3, FNDC3B, ARHGEF12, GAS7, FOXC1, ATXN2, TXNRD2); PACG (primary angle-closure glaucoma (EPDR1, CHAT, GLIS3, FERMT2, DPM2-FAM102); and exfoliation syndrome (XFS) glaucoma (CACNA1A). In total sixteen genomic regions have been associated with POAG (including the normal tension glaucoma (NTG) subgroup), 8 with PACG and 2 with XFS. These studies are defining important biological pathways and processes that contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Janey L Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Louis R Pasquale
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, MA 02114, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
49
|
Velez G, Tsang SH, Tsai YT, Hsu CW, Gore A, Abdelhakim AH, Mahajan M, Silverman RH, Sparrow JR, Bassuk AG, Mahajan VB. Gene Therapy Restores Mfrp and Corrects Axial Eye Length. Sci Rep 2017; 7:16151. [PMID: 29170418 PMCID: PMC5701072 DOI: 10.1038/s41598-017-16275-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 11/09/2017] [Indexed: 01/07/2023] Open
Abstract
Hyperopia (farsightedness) is a common and significant cause of visual impairment, and extreme hyperopia (nanophthalmos) is a consequence of loss-of-function MFRP mutations. MFRP deficiency causes abnormal eye growth along the visual axis and significant visual comorbidities, such as angle closure glaucoma, cystic macular edema, and exudative retinal detachment. The Mfrp rd6 /Mfrp rd6 mouse is used as a pre-clinical animal model of retinal degeneration, and we found it was also hyperopic. To test the effect of restoring Mfrp expression, we delivered a wild-type Mfrp to the retinal pigmented epithelium (RPE) of Mfrp rd6 /Mfrp rd6 mice via adeno-associated viral (AAV) gene therapy. Phenotypic rescue was evaluated using non-invasive, human clinical testing, including fundus auto-fluorescence, optical coherence tomography, electroretinography, and ultrasound. These analyses showed gene therapy restored retinal function and normalized axial length. Proteomic analysis of RPE tissue revealed rescue of specific proteins associated with eye growth and normal retinal and RPE function. The favorable response to gene therapy in Mfrp rd6 /Mfrp rd6 mice suggests hyperopia and associated refractive errors may be amenable to AAV gene therapy.
Collapse
Affiliation(s)
- Gabriel Velez
- Omics Laboratory, Stanford University, Palo Alto, CA, USA
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA, USA
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
| | - Stephen H Tsang
- Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Columbia University, New York, NY, USA.
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.
| | - Yi-Ting Tsai
- Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Chun-Wei Hsu
- Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Anuradha Gore
- Omics Laboratory, Stanford University, Palo Alto, CA, USA
| | - Aliaa H Abdelhakim
- Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | | | - Ronald H Silverman
- Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Janet R Sparrow
- Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Alexander G Bassuk
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA.
- Department of Neurology, University of Iowa, Iowa City, IA, USA.
- Palo Alto Veterans Administration, Palo Alto, CA, USA.
| | - Vinit B Mahajan
- Omics Laboratory, Stanford University, Palo Alto, CA, USA.
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA, USA.
- Department of Neurology, University of Iowa, Iowa City, IA, USA.
- Palo Alto Veterans Administration, Palo Alto, CA, USA.
| |
Collapse
|
50
|
Novel pathogenic mutations in C1QTNF5 support a dominant negative disease mechanism in late-onset retinal degeneration. Sci Rep 2017; 7:12147. [PMID: 28939808 PMCID: PMC5610255 DOI: 10.1038/s41598-017-11898-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/31/2017] [Indexed: 11/18/2022] Open
Abstract
Late-onset retinal degeneration (L-ORD) is a rare autosomal dominant retinal dystrophy, characterised by extensive sub-retinal pigment epithelium (RPE) deposits, RPE atrophy, choroidal neovascularisation and photoreceptor cell death associated with severe visual loss. L-ORD shows striking phenotypic similarities to age-related macular degeneration (AMD), a common and genetically complex disorder, which can lead to misdiagnosis in the early stages. To date, a single missense mutation (S163R) in the C1QTNF5 gene, encoding C1q And Tumor Necrosis Factor Related Protein 5 (C1QTNF5) has been shown to cause L-ORD in a subset of affected families. Here, we describe the identification and characterisation of three novel pathogenic mutations in C1QTNF5 in order to elucidate disease mechanisms. In silico and in vitro characterisation show that these mutations perturb protein folding, assembly or polarity of secretion of C1QTNF5 and, importantly, all appear to destabilise the wildtype protein in co-transfection experiments in a human RPE cell line. This suggests that the heterozygous mutations in L-ORD show a dominant negative, rather than a haploinsufficient, disease mechanism. The function of C1QTNF5 remains unclear but this new insight into the pathogenetic basis of L-ORD has implications for future therapeutic strategies such as gene augmentation therapy.
Collapse
|