1
|
Labay-Tejado S, Laguna J, Navarro-Angulo MJ, Muñoz L, Jodar M, Milla E. XEN 63 in a case of refractory glaucoma secondary to microphthalmia and aphakia. ARCHIVOS DE LA SOCIEDAD ESPANOLA DE OFTALMOLOGIA 2025:S2173-5794(25)00036-2. [PMID: 40058695 DOI: 10.1016/j.oftale.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 11/30/2024] [Indexed: 03/15/2025]
Abstract
Microphthalmia (MO) predisposes to glaucoma and congenital cataracts (CC). Cataract removal in MO is characterized by several complications such as aphakic glaucoma (AG). We report the case of a 33-year-old male with MO, aphakia, and refractory glaucoma who underwent inferonasal ab-interno XEN 63 implantation, after posterior vitrectomy. Whole Exome Sequencing (WES) analysis was performed identifying the likely pathogenic variant c.197G>A (p.Cys66Tyr) in heterozygosity in the PAX6 gene related to Peters anomaly (PA) and the variant of unclear significance c.353T>C (p.Leu118Pro) in heterozygosity in the CRYBB3 gene, potentially related to CC. Until the 12-month follow-up, the patient maintained intraocular pressure (IOP) between 16 and 18 mmHg without medication or complications. XEN 63, when combined with posterior vitrectomy may be an option in select cases, owing to its versatility. However, further investigation is required. Genetic analysis provides a pre-symptomatic diagnosis and may lead to a more personalised medicine.
Collapse
Affiliation(s)
- S Labay-Tejado
- Institut Clínic d'Oftalmologia (ICOF), Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain.
| | - J Laguna
- Department of Biochemistry and Molecular Genetics (CDB), Hospital Clínic de Barcelona, Barcelona, Spain
| | - M J Navarro-Angulo
- Department of Surgical Retina, Institut Clínic d'Oftalmologia (ICOF), Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - L Muñoz
- Department of Biochemistry and Molecular Genetics (CDB), Hospital Clínic de Barcelona, Barcelona, Spain
| | - M Jodar
- Department of Biochemistry and Molecular Genetics (CDB), Hospital Clínic de Barcelona, Barcelona, Spain; Department of Biomedicine, Faculty of Medicine and Biomedical Sciences, Universitat de Barcelona, Barcelona, Spain
| | - E Milla
- Department of Glaucoma, Institut Clínic d'Oftalmologia (ICOF), Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
2
|
Hussain A, Villalba MF, Swols DM, Khzam RA, Johnson BK, Peart L, D'Haiti S, Grajewski AL, Tekin M, Chang TC, Bademci G. Genome sequencing reveals novel variants in a diverse population with congenital anterior segment anomalies. Sci Rep 2025; 15:518. [PMID: 39747279 PMCID: PMC11695809 DOI: 10.1038/s41598-024-84205-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Congenital anterior segment anomalies are disorders that affect the development of the eye and cause severe visual impairment. The molecular basis of congenital anterior segment anomalies is not well known. In this study, genome sequencing was performed on 27 families from diverse ethnicities with congenital anterior segment anomalies and 11 variants were identified, most of which were novel and family specific. These variants included single nucleotide variants CPAMD8:c.4825 C > T, c.534 G > A, CRYBB1:c.683 C > A, NHS:c.1180 C > T, GJA3:c.176 C > T, CRYGC:c.470 G > A, COL2A1:c.2819 G > A, c.1693 C > T, EPHA2:c.2864 A > C, a splice donor variant in COL11A1:c.933 + 1del, and a copy number variant in FBN1. The observed inheritance patterns were predominantly dominant, with a few recessive cases and a single instance of X-linked inheritance. Genome sequencing identified variants in 40.74% of diverse cases, offering valuable insights for enhancing the diagnosis and management of this disorder.
Collapse
Affiliation(s)
- Ashraf Hussain
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Maria Fernanda Villalba
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Dayna Morel Swols
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Rayan Abou Khzam
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Brittney Keira Johnson
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - LéShon Peart
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Sarha D'Haiti
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alana L Grajewski
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Mustafa Tekin
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Ta Chen Chang
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Guney Bademci
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
3
|
Reis LM, Basel D, Bitoun P, Walton DS, Glaser T, Semina EV. Novel Intragenic and Genomic Variants Highlight the Phenotypic Variability in HCCS-Related Disease. Genes (Basel) 2024; 15:1636. [PMID: 39766903 PMCID: PMC11675438 DOI: 10.3390/genes15121636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Disruption of HCCS results in microphthalmia with linear skin lesions (MLS) characterized by microphthalmia/anophthalmia, corneal opacity, aplastic skin lesions, variable central nervous system and cardiac anomalies, intellectual disability, and poor growth in heterozygous females. Structural variants consisting of chromosomal rearrangements or deletions are the most common variant type, but a small number of intragenic variants have been reported. Methods: Exome sequencing identified variants affecting HCCS. Results: Three novel intragenic variants and two genomic deletions of HCCS were found in individuals with primarily ocular features of MLS. X-inactivation was highly skewed in affected individuals with all three intragenic variants. Corneal opacity was the most penetrant feature (100%). In addition, a duplication of uncertain significance including both HCCS and AMELX was identified in a male with corneal anomalies, glaucoma, an atrial septal defect, and enamel hypoplasia along with a family history of developmental ocular disorders consistent with X-linked inheritance. Conclusion: Although variable expressivity is a known feature of MLS, our findings provide additional support for including HCCS in testing for individuals with isolated ocular anomalies and provide further evidence for its association with congenital aphakia, aniridia/other iris defects, and corneal staphyloma/ectasia.
Collapse
Affiliation(s)
- Linda M. Reis
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Donald Basel
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Pierre Bitoun
- Génétique Médicale, SIDVA91/Altérité, 1 Impasse de la Cour de France, 91260 Juvisy-sur-Orge, France;
| | - David S. Walton
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 8 Hawthorne Place, Boston, MA 02114, USA;
| | - Tom Glaser
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA;
| | - Elena V. Semina
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
4
|
Vaseghi P, Habibi L, Neidich JA, Cao Y, Fattahi N, Rashidi-Nezhad R, Salehnezhad T, Dalili H, Rahimi Sharbaf F, Zarkesh MR, Malekian M, Mokhberdezfuli M, Mehrtash A, Ardeshirdavani A, Kariminejad R, Ghorbansabagh V, Sadeghimoghadam P, Naddaf A, Esmaeilnia Shirvany T, Mosayebi Z, Sahebdel B, Golshahi F, Shirazi M, Shamel S, Moeini R, Heidari A, Daneshmand MA, Ghasemi R, Akrami SM, Rashidi-Nezhad A. Towards solving the genetic diagnosis odyssey in Iranian patients with congenital anomalies. Eur J Hum Genet 2024; 32:1238-1249. [PMID: 38278869 PMCID: PMC11499880 DOI: 10.1038/s41431-024-01533-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 11/23/2023] [Accepted: 12/12/2023] [Indexed: 01/28/2024] Open
Abstract
Understanding the underlying causes of congenital anomalies (CAs) can be a complex diagnostic journey. We aimed to assess the efficiency of exome sequencing (ES) and chromosomal microarray analysis (CMA) in patients with CAs among a population with a high fraction of consanguineous marriage. Depending on the patient's symptoms and family history, karyotype/Quantitative Fluorescence- Polymerase Chain Reaction (QF-PCR) (n = 84), CMA (n = 81), ES (n = 79) or combined CMA and ES (n = 24) were performed on 168 probands (66 prenatal and 102 postnatal) with CAs. Twelve (14.28%) probands were diagnosed by karyotype/QF-PCR and seven (8.64%) others were diagnosed by CMA. ES findings were conclusive in 39 (49.36%) families, and 61.90% of them were novel variants. Also, 64.28% of these variants were identified in genes that follow recessive inheritance in CAs. The diagnostic rate (DR) of ES was significantly higher than that of CMA in children from consanguineous families (P = 0·0001). The highest DR by CMA was obtained in the non-consanguineous postnatal subgroup and by ES in the consanguineous prenatal subgroup. In a population that is highly consanguineous, our results suggest that ES may have a higher diagnostic yield than CMA and should be considered as the first-tier test in the evaluation of patients with congenital anomalies.
Collapse
Affiliation(s)
- Parisa Vaseghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Laleh Habibi
- Ronash Medical Genetics Laboratory, Tehran, Iran
| | - Julie A Neidich
- Department of Pathology & Immunology, Division of Laboratory & Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Yang Cao
- Department of Pathology & Immunology, Division of Laboratory & Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Neda Fattahi
- Ronash Medical Genetics Laboratory, Tehran, Iran
| | | | | | - Hossein Dalili
- Breastfeeding Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pediatrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Rahimi Sharbaf
- Department of Obstetrics and Gynecology, School of Medicine, Yas Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zarkesh
- Department of Neonatology, Yas Hospital Complex, Tehran university of medical sciences, Tehran, Iran
| | | | - Mahdieh Mokhberdezfuli
- Ronash Medical Genetics Laboratory, Tehran, Iran
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Vafa Ghorbansabagh
- Department of Pediatrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Maternal, Fetal and Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parvane Sadeghimoghadam
- Department of Pediatrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Maternal, Fetal and Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Naddaf
- Department of Pediatrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Maternal, Fetal and Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahereh Esmaeilnia Shirvany
- Department of Pediatrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Maternal, Fetal and Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ziba Mosayebi
- Department of Pediatrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Maternal, Fetal and Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrokh Sahebdel
- Department of Obstetrics and Gynecology, School of Medicine, Yas Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Golshahi
- Department of Obstetrics and Gynecology, School of Medicine, Yas Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboobeh Shirazi
- Department of Obstetrics and Gynecology, School of Medicine, Yas Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Shamel
- Department of Neonatology, Yas Hospital Complex, Tehran university of medical sciences, Tehran, Iran
| | - Roksana Moeini
- Department of Neonatology, Yas Hospital Complex, Tehran university of medical sciences, Tehran, Iran
| | | | | | - Reza Ghasemi
- Department of Pathology & Immunology, Division of Laboratory & Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Seyed Mohammad Akrami
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ali Rashidi-Nezhad
- Maternal, Fetal and Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Genetics Ward, Yas Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Reis LM, Seese SE, Costakos D, Semina EV. Congenital anterior segment ocular disorders: Genotype-phenotype correlations and emerging novel mechanisms. Prog Retin Eye Res 2024; 102:101288. [PMID: 39097141 PMCID: PMC11392650 DOI: 10.1016/j.preteyeres.2024.101288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Development of the anterior segment of the eye requires reciprocal sequential interactions between the arising tissues, facilitated by numerous genetic factors. Disruption of any of these processes results in congenital anomalies in the affected tissue(s) leading to anterior segment disorders (ASD) including aniridia, Axenfeld-Rieger anomaly, congenital corneal opacities (Peters anomaly, cornea plana, congenital primary aphakia), and primary congenital glaucoma. Current understanding of the genetic factors involved in ASD remains incomplete, with approximately 50% overall receiving a genetic diagnosis. While some genes are strongly associated with a specific clinical diagnosis, the majority of known factors are linked with highly variable phenotypic presentations, with pathogenic variants in FOXC1, CYP1B1, and PITX2 associated with the broadest spectrum of ASD conditions. This review discusses typical clinical presentations including associated systemic features of various forms of ASD; the latest functional data and genotype-phenotype correlations related to 25 ASD factors including newly identified genes; promising novel candidates; and current and emerging treatments for these complex conditions. Recent developments of interest in the genetics of ASD include identification of phenotypic expansions for several factors, discovery of multiple modes of inheritance for some genes, and novel mechanisms including a growing number of non-coding variants and alleles affecting specific domains/residues and requiring further studies.
Collapse
Affiliation(s)
- Linda M Reis
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Sarah E Seese
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Deborah Costakos
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Elena V Semina
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA; Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin and Children's Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
6
|
Cronemberger S, Albuquerque ALB, Silva ACSE, Zanini JLSS, da Silva AHG, Barbosa LF, da Cunha Rubião F, de Lima FL, Casimiro RF, Martins MP, Diniz-Filho A, Bastos-Rodrigues L, Friedman E, De Marco L. Bilateral Peters' anomaly, aniridia and Wilms tumour (WAGR syndrome) in monozygotic twins. Acta Paediatr 2024; 113:1420-1425. [PMID: 38363039 DOI: 10.1111/apa.17160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
AIM This study reports the bilateral association of Peters' anomaly and congenital aniridia in monozygotic twins subsequently diagnosed with Wilms tumour (WAGR syndrome). METHODS Two monozygotic female twins were referred at age 2 months with bilateral corneal opacity. A diagnosis of Peters' anomaly associated to aniridia was made in both eyes of both twins. Physical examination and ultrasonography were carried out at 12 months of age to explore the possibility of WAGR-related anomalies, specifically Wilms tumour. DNA were isolated and subjected to whole exome sequencing. RESULTS Peters' anomaly associated to aniridia in both eyes as well as bilateral Wilms tumour in both children were diagnosed. Exome analyses showed a large heterozygous deletion encompassing 6 648 473 bp in chromosome 11p13, using Integrative Genomics Viewer and AnnotSV software. CONCLUSION WAGR syndrome is a rare contiguous gene deletion syndrome with a greater risk of developing Wilms tumour associated with Peters' anomaly and congenital aniridia. However, co-occurrence of both anomalies was rarely reported in twins, and never in both eyes of monozygotic twins. Here, we report the bilateral association of Peters' anomaly and congenital aniridia in monozygotic twins with WAGR syndrome.
Collapse
Affiliation(s)
| | - Anna L B Albuquerque
- Department of Surgery, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | - Luciana F Barbosa
- Hospital São Geraldo, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Felipe L de Lima
- Hospital São Geraldo, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Alberto Diniz-Filho
- Hospital São Geraldo, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Eitan Friedman
- The Preventive Personalized Medicine Center, Assuta Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Luiz De Marco
- Department of Surgery, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
7
|
Wowra B, Dobrowolski D, Parekh M, Wylęgała E. General Treatment and Ophthalmic Management of Peters' Anomaly. J Clin Med 2024; 13:532. [PMID: 38256667 PMCID: PMC10816361 DOI: 10.3390/jcm13020532] [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: 11/19/2023] [Revised: 12/30/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Peters' anomaly (PA) is a manifestation of complex disorders in the development of the anterior segment of the eye. The most recognizable feature of the disease is a doughnut-shaped central corneal opacity and adhesions between the opacity and underlying iris. Glaucoma is observed in 30-70% of patients, with up to 50% of the patients showing concomitant vision-threatening disorders. Up to 60% of patients have systemic abnormalities or developmental delays. Being a rare malformation, PA is one of the most common congenital indications for corneal transplantation in infants. Penetrating keratoplasty is used as the primary method of treatment in cases with corneal opacification of a degree that forbids visual development in both eyes. The heterogeneity of co-occurring ophthalmic and systemic malformations in the spectrum of PA determines the wide range of success, defined by various endpoints: graft clarity or visual acuity. Although surgical advancement has made corneal grafting possible in younger children, it has a higher graft failure rate and worse visual prognosis than adult keratoplasty. Optical sector iridectomy, pupil dilation, or cornea rotation can alternatively be performed. Satisfying results of pediatric keratoprosthesis in particular cases of PA have been described. Postoperative treatment of PA aims to maintain a clear optical pathway and prevent amblyopia. This article therefore aims at reporting the ophthalmic treatment and need for multidisciplinary management of PA, including pharmacological and surgical treatment.
Collapse
Affiliation(s)
- Bogumil Wowra
- Chair and Department of Ophthalmology, Faculty of Medical Sciences, Zabrze Medical University of Silesia, 40-760 Katowice, Poland
| | - Dariusz Dobrowolski
- Chair and Department of Ophthalmology, Faculty of Medical Sciences, Zabrze Medical University of Silesia, 40-760 Katowice, Poland
- Department of Ophthalmology, Saint Barbara Hospital, Trauma Center, 41-200 Sosnowiec, Poland
| | - Mohit Parekh
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Edward Wylęgała
- Chair and Department of Ophthalmology, Faculty of Medical Sciences, Zabrze Medical University of Silesia, 40-760 Katowice, Poland
| |
Collapse
|
8
|
Chacon-Camacho OF, Arce-Gonzalez R, Sanchez-de la Rosa F, Urióstegui-Rojas A, Hofmann-Blancas ME, Mata-Flores F, Zenteno JC. Genetic Aspects of Glaucoma: An Updated Review. Curr Mol Med 2024; 24:1231-1249. [PMID: 37272463 DOI: 10.2174/1566524023666230602143617] [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: 12/01/2022] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 06/06/2023]
Abstract
Glaucoma is a group of diverse diseases characterized by cupping of the optic nerve head due to the loss of retinal ganglion cells. It is the most common cause of irreversible blindness throughout the world; therefore, its timely diagnosis and early detection through an ophthalmological examination are very important. We, herein, present the information on the epidemiology, pathophysiology, clinical diagnosis, and treatment of glaucoma. We also emphasize the investigations of the last decades that have allowed identifying numerous genes and susceptibility genetic factors. We have also described in detail the genes whose mutations cause or contribute to the development of the disease.
Collapse
Affiliation(s)
- Oscar Francisco Chacon-Camacho
- Department of Genetics, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
- Laboratorio 5 Edificio A-4, Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rocio Arce-Gonzalez
- Department of Genetics, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | | | - Andrés Urióstegui-Rojas
- Department of Integral Ophthalmology, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | | | - Felipe Mata-Flores
- Department of Glaucoma, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | - Juan Carlos Zenteno
- Department of Genetics, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
- Biochemistry Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
9
|
Reis LM, Amor DJ, Haddad RA, Nowak CB, Keppler-Noreuil KM, Chisholm SA, Semina EV. Alternative Genetic Diagnoses in Axenfeld-Rieger Syndrome Spectrum. Genes (Basel) 2023; 14:1948. [PMID: 37895297 PMCID: PMC10606241 DOI: 10.3390/genes14101948] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
Axenfeld-Rieger anomaly (ARA) is a specific ocular disorder that is frequently associated with other systemic abnormalities. PITX2 and FOXC1 variants explain the majority of individuals with Axenfeld-Rieger syndrome (ARS) but leave ~30% unsolved. Here, we present pathogenic/likely pathogenic variants in nine families with ARA/ARS or similar phenotypes affecting five different genes/regions. USP9X and JAG1 explained three families each. USP9X was recently linked with syndromic cognitive impairment that includes hearing loss, dental defects, ventriculomegaly, Dandy-Walker malformation, skeletal anomalies (hip dysplasia), and other features showing a significant overlap with FOXC1-ARS. Anterior segment anomalies are not currently associated with USP9X, yet our cases demonstrate ARA, congenital glaucoma, corneal neovascularization, and cataracts. The identification of JAG1 variants, linked with Alagille syndrome, in three separate families with a clinical diagnosis of ARA/ARS highlights the overlapping features and high variability of these two phenotypes. Finally, intragenic variants in CDK13, BCOR, and an X chromosome deletion encompassing HCCS and AMELX (linked with ocular and dental anomalies, correspondingly) were identified in three additional cases with ARS. Accurate diagnosis has important implications for clinical management. We suggest that broad testing such as exome sequencing be applied as a second-tier test for individuals with ARS with normal results for PITX2/FOXC1 sequencing and copy number analysis, with attention to the described genes/regions.
Collapse
Affiliation(s)
- Linda M. Reis
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (L.M.R.); (S.A.C.)
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, WI 53226, USA
| | - David J. Amor
- Murdoch Children’s Research Institute, Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia;
| | - Raad A. Haddad
- Division of Endocrinology, Diabetes, and Metabolic Diseases, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Catherine B. Nowak
- Division of Genetics and Metabolism, MassGeneral Hospital for Children, Boston, MA 02114, USA;
| | - Kim M. Keppler-Noreuil
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA;
| | - Smith Ann Chisholm
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (L.M.R.); (S.A.C.)
| | - Elena V. Semina
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (L.M.R.); (S.A.C.)
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
10
|
Khasnavis A, Fernandes M. Peters anomaly: An overview. Taiwan J Ophthalmol 2023; 13:434-442. [PMID: 38249502 PMCID: PMC10798386 DOI: 10.4103/tjo.tjo-d-23-00065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/16/2023] [Indexed: 01/23/2024] Open
Abstract
Peters anomaly (PA) is a rare, often bilateral, congenital corneal opacity, usually with a sporadic inheritance pattern, characterized by corneal opacities and irido-corneal or lenticular-corneal adhesions with a defect in the Descemet's membrane, occurring due to anterior segment dysgenesis during fetal development. Due to other ocular and systemic comorbidities, a team comprising pediatric cornea, glaucoma, and strabismus specialists in addition to a pediatrician and geneticist is necessary for the appropriate management of these children. Since the outcome of pediatric penetrating keratoplasty is variable and has a higher chance of failure when accompanied by additional procedures, such as lensectomy and vitrectomy, minimally invasive alternatives are increasingly being offered to these patients. Of note is the recently reported novel procedure: selective endothelialectomy for PA, which avoids the need for a corneal transplant and results in gradual clearing of the corneal opacity over time. In this overview, we aimed to describe the etiology, classification, pathophysiology, histopathology, clinical features, and management of PA.
Collapse
Affiliation(s)
- Arpita Khasnavis
- Academy for Eye Care Education, L V Prasad Eye Institute, Hyderabad, Telangana, India
- Cornea and Anterior Segment Service, Shantilal Shanghvi Cornea Institute, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Merle Fernandes
- Cornea and Anterior Segment Service, Shantilal Shanghvi Cornea Institute, L V Prasad Eye Institute, Hyderabad, Telangana, India
| |
Collapse
|
11
|
Khandwala NS, Ramappa M, Edward DP, Mocan MC. Axenfeld-Rieger syndrome in the pediatric population: A review. Taiwan J Ophthalmol 2023; 13:417-424. [PMID: 38249500 PMCID: PMC10798402 DOI: 10.4103/tjo.tjo-d-23-00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/03/2023] [Indexed: 01/23/2024] Open
Abstract
Axenfeld-Rieger syndrome (ARS) is a rare autosomal-dominant neurocristopathy that presents with a variety of classical ocular and systemic findings. The pathophysiology of the disease involves anterior segment dysgenesis, and patients may present with ophthalmic complications early in life, including secondary glaucoma, high refractive errors, amblyopia, and permanent visual damage. There are a limited number of studies in the literature that focus primarily on pediatric patients with ARS. The purpose of this article was to review the current literature on clinical presentation, genetic associations, diagnosis, secondary complications, and treatment of ARS in pediatric patients. Evaluating the essential clinical aspects of the disease in children may allow for earlier diagnosis and treatment and prevent visual morbidity from amblyopia and secondary glaucoma that may result in permanent visual damage.
Collapse
Affiliation(s)
- Nikhila S. Khandwala
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Muralidhar Ramappa
- Centre for Rare Eye Diseases and Ocular Genetics, The Cornea Institute, Jasti V Ramanamma Children’s Eye Care Center, L.V. Prasad Eye Institute, Hyderabad, Telangana, India
| | - Deepak P. Edward
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mehmet C. Mocan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
12
|
Paredes DI, Capasso JE, Wyman CS, Levin AV. Genetics of the anterior segment dysgenesis. Taiwan J Ophthalmol 2023; 13:500-504. [PMID: 38249510 PMCID: PMC10798390 DOI: 10.4103/tjo.tjo-d-23-00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 01/23/2024] Open
Abstract
The anterior segment dysgeneses are a broad group of heterogeneous disorders characterized by developmental abnormalities of the anterior segment of the eye, including primary congenital aphakia, Peters sequence, aniridia, and Axenfeld-Rieger spectrum. These conditions can have overlapping phenotypes and both genotypic and phenotypic heterogeneity. This article provides a strategy for both phenotyping and then genotyping using a targeted stepwise approach.
Collapse
Affiliation(s)
- Diego I Paredes
- Department of Pediatric Ophthalmology and Ocular Genetics, Flaum Eye Institute, University of Rochester, Rochester, New York, USA
- Department of Ophthalmology, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Jenina E Capasso
- Department of Pediatric Ophthalmology and Ocular Genetics, Flaum Eye Institute, University of Rochester, Rochester, New York, USA
- Department of Clinical Genetics, Golisano Children's Hospital, University of Rochester, Rochester, New York, USA
| | - Celeste S Wyman
- Department of Pediatric Ophthalmology and Ocular Genetics, Flaum Eye Institute, University of Rochester, Rochester, New York, USA
- Department of Clinical Genetics, Golisano Children's Hospital, University of Rochester, Rochester, New York, USA
| | - Alex V Levin
- Department of Pediatric Ophthalmology and Ocular Genetics, Flaum Eye Institute, University of Rochester, Rochester, New York, USA
- Department of Clinical Genetics, Golisano Children's Hospital, University of Rochester, Rochester, New York, USA
| |
Collapse
|
13
|
Reis LM, Chassaing N, Bardakjian T, Thompson S, Schneider A, Semina EV. ARHGAP35 is a novel factor disrupted in human developmental eye phenotypes. Eur J Hum Genet 2023; 31:363-367. [PMID: 36450800 PMCID: PMC9995503 DOI: 10.1038/s41431-022-01246-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
ARHGAP35 has known roles in cell migration, invasion and division, neuronal morphogenesis, and gene/mRNA regulation; prior studies indicate a role in cancer in humans and in the developing eyes, neural tissue, and renal structures in mice. We identified damaging variants in ARHGAP35 in five individuals from four families affected with anophthalmia, microphthalmia, coloboma and/or anterior segment dysgenesis disorders, together with variable non-ocular phenotypes in some families including renal, neurological, or cardiac anomalies. Three variants affected the extreme C-terminus of the protein, with two resulting in a frameshift and C-terminal extension and the other a missense change in the Rho-GAP domain; the fourth (nonsense) variant affected the middle of the gene and is the only allele predicted to undergo nonsense-mediated decay. This study implicates ARHGAP35 in human developmental eye phenotypes. C-terminal clustering of the identified alleles indicates a possible common mechanism for ocular disease but requires further studies.
Collapse
Affiliation(s)
- Linda M Reis
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin and Children's Wisconsin, Milwaukee, WI, USA
| | - Nicolas Chassaing
- Service de Génétique Médicale, Hôpital Purpan CHU Toulouse, Toulouse, France
- Platerforme AURAGEN, Lyon, France
| | | | - Samuel Thompson
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin and Children's Wisconsin, Milwaukee, WI, USA
| | | | - Elena V Semina
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin and Children's Wisconsin, Milwaukee, WI, USA.
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
14
|
Reis LM, Atilla H, Kannu P, Schneider A, Thompson S, Bardakjian T, Semina EV. Distinct Roles of Histone Lysine Demethylases and Methyltransferases in Developmental Eye Disease. Genes (Basel) 2023; 14:216. [PMID: 36672956 PMCID: PMC9859058 DOI: 10.3390/genes14010216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Histone lysine methyltransferase and demethylase enzymes play a central role in chromatin organization and gene expression through the dynamic regulation of histone lysine methylation. Consistent with this, genes encoding for histone lysine methyltransferases (KMTs) and demethylases (KDMs) are involved in complex human syndromes, termed congenital regulopathies. In this report, we present several lines of evidence for the involvement of these genes in developmental ocular phenotypes, suggesting that individuals with structural eye defects, especially when accompanied by craniofacial, neurodevelopmental and growth abnormalities, should be examined for possible variants in these genes. We identified nine heterozygous damaging genetic variants in KMT2D (5) and four other histone lysine methyltransferases/demethylases (KMT2C, SETD1A/KMT2F, KDM6A and KDM5C) in unrelated families affected with developmental eye disease, such as Peters anomaly, sclerocornea, Axenfeld-Rieger spectrum, microphthalmia and coloboma. Two families were clinically diagnosed with Axenfeld-Rieger syndrome and two were diagnosed with Peters plus-like syndrome; others received no specific diagnosis prior to genetic testing. All nine alleles were novel and five of them occurred de novo; five variants resulted in premature truncation, three were missense changes and one was an in-frame deletion/insertion; and seven variants were categorized as pathogenic or likely pathogenic and two were variants of uncertain significance. This study expands the phenotypic spectra associated with KMT and KDM factors and highlights the importance of genetic testing for correct clinical diagnosis.
Collapse
Affiliation(s)
- Linda M. Reis
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, WI 53226, USA
| | - Huban Atilla
- Department of Ophthalmology, School of Medicine, Ankara University, 0600 Ankara, Turkey
| | - Peter Kannu
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Adele Schneider
- Einstein Medical Center Philadelphia, Philadelphia, PA 19141, USA
| | - Samuel Thompson
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, WI 53226, USA
| | - Tanya Bardakjian
- Einstein Medical Center Philadelphia, Philadelphia, PA 19141, USA
| | - Elena V. Semina
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, WI 53226, USA
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
15
|
Syu YM, Ma JY, Ou TH, Lee CL, Lin HY, Lin SP, Lee CJ, Chen CP. De Novo Mosaic 6p23-p25.3 Tetrasomy Caused by a Small Supernumerary Marker Chromosome Presenting Trisomy Distal 6p Phenotype: A Case Report and Literature Review. Diagnostics (Basel) 2022; 12:2306. [PMID: 36291995 PMCID: PMC9600663 DOI: 10.3390/diagnostics12102306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/13/2022] [Accepted: 09/21/2022] [Indexed: 11/26/2022] Open
Abstract
Small supernumerary marker chromosomes (sSMCs) derived from the chromosome 6 short arm are rare and their clinical significance remains unknown. No case with sSMC(6) without centromeric DNA has been reported. Partial trisomy and tetrasomy of distal 6p is a rare but clinically distinct syndrome. We report on a de novo mosaic sSMC causing partial tetrasomy for 6p23-p25.3 in a male infant with symptoms of being small for gestational age, microcephaly, facial dysmorphism, congenital eye defects, and multi-system malformation. Conventional cytogenetic analysis revealed a karyotype of 47,XY,+mar [25]/46,XY [22]. Array comparative genomic hybridization (aCGH) revealed mosaic tetrasomy of distal 6p. This is the first case of mosaic tetrasomy 6p23-p25.3 caused by an inverted duplicated neocentric sSMC with characteristic features of trisomy distal 6p. Comparison of phenotypes in cases with trisomy and tetrasomy of 6p23-p25.3 could facilitate a genotype-phenotype correlation and identification of candidate genes contributing to their presentation. The presentation of anterior segment dysgenesis and anomaly of the renal system suggest triplosensitivity of the FOXC1 gene. In patients with microcephaly growth retardation, and malformation of the cardiac and renal systems, presentation of anterior segment dysgenesis might be indicative of chromosome 6p duplication, and aCGH evaluation should be performed for associated syndromic disease.
Collapse
Affiliation(s)
- Yu-Min Syu
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City 22021, Taiwan
- Division of Genetics and Metabolism, Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Juine-Yih Ma
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City 22021, Taiwan
| | - Tzu-Hsuen Ou
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City 22021, Taiwan
| | - Chung-Lin Lee
- Division of Genetics and Metabolism, Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Hsiang-Yu Lin
- Division of Genetics and Metabolism, Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, Division of Genetics and Metabolism, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Childhood Care and Education, MacKay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| | - Shuan-Pei Lin
- Division of Genetics and Metabolism, Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Rare Disease Center, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, Division of Genetics and Metabolism, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan
| | - Chia-Jung Lee
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City 22021, Taiwan
| | - Chih-Ping Chen
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei 10449, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
- Institute of Clinical and Community Health Nursing, National Yang Ming Chiao Tung University, Taipei 11230, Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11230, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41346, Taiwan
| |
Collapse
|
16
|
Harboyan Syndrome: A Novel SLC4A11 Variant With Unique Genotype-Phenotype Correlation. Cornea 2022; 41:1053-1057. [PMID: 35439766 DOI: 10.1097/ico.0000000000003023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE The purpose of this study was to describe the genotypic and phenotypic characteristics of an infant with a SLC4A11 mutation associated with bilateral corneal edema, hearing loss, and hydronephrosis present since birth. METHODS This was a case report. Ophthalmic and systemic examination of the proband, histopathologic and ultrastructural characteristics of bilateral corneal discs, and molecular genetic evaluation by whole-exome sequencing are described. RESULTS A male infant was born with bilateral corneal opacities, sensorineural hearing loss, and hydronephrosis to healthy parents after an uneventful pregnancy. Penetrating keratoplasty of the left eye at age 10 months demonstrated minimal corneal edema with normal thickness Descemet membrane and cellular endothelium with intracytoplasmic vacuoles and degenerative changes in rare cells. Penetrating keratoplasty of the right eye 6 months later disclosed prominent corneal edema with a thickened posterior banded layer of Descemet membrane and severe endothelial atrophy. Whole-exome sequencing of the proband and parents' blood demonstrated a homozygous mutation in SLC4A11 gene (c.1735_1737delCTC,p.Leu579del). The combined clinical, histopathologic, and molecular genetic findings raised consideration of an unusual phenotype of Harboyan syndrome manifesting as congenital hereditary endothelial dystrophy with a prelingual rather than, as previously described, postlingual hearing loss. CONCLUSIONS We report a novel homozygous SLC4A11 variant with a previously undocumented phenotype of CHED in association with prelingual sensorineural hearing loss and hydronephrosis, thus broadening our understanding of the spectrum of genotypic and phenotypic findings of Harboyan syndrome.
Collapse
|
17
|
Karaconji T, Zagora S, Grigg JR. Approach to childhood glaucoma: A review. Clin Exp Ophthalmol 2022; 50:232-246. [PMID: 35023613 DOI: 10.1111/ceo.14039] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/01/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022]
Abstract
Childhood glaucoma represents a heterogenous group of rare ocular conditions that may result in significant sight threatening complications related to elevated intraocular pressure (IOP). It can be classified as either primary or secondary and the latter may have systemic associations. This review will be based on the work of the childhood glaucoma research network (CGRN) and will focus on the diagnosis and management of the most common types of childhood glaucoma. These include primary congenital glaucoma (PCG) and juvenile open angle glaucoma (JOAG) as well as secondary causes of glaucoma associated with non-acquired ocular anomalies (Axenfeld-Rieger anomaly; Peters anomaly and Aniridia), glaucoma associated with systemic disease (Sturge Weber syndrome and Neurofibromatosis), those due to acquired conditions (Uveitic glaucoma, trauma and tumours) and importantly glaucoma following cataract surgery.
Collapse
Affiliation(s)
- Tanya Karaconji
- Speciality of Ophthalmology, Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Department of Ophthalmology, The Children's Hospital, Westmead, Australia
| | - Sophia Zagora
- Speciality of Ophthalmology, Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Department of Ophthalmology, The Children's Hospital, Westmead, Australia
| | - John R Grigg
- Speciality of Ophthalmology, Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Department of Ophthalmology, The Children's Hospital, Westmead, Australia.,Eye Genetics Research Group Children's Medical Research Institute, The Children's Hospital at Westmead and Eye Genetics Clinics, The Children's Hospital at Westmead, Westmead, Australia
| |
Collapse
|
18
|
Chesneau B, Aubert-Mucca M, Fremont F, Pechmeja J, Soler V, Isidor B, Nizon M, Dollfus H, Kaplan J, Fares-Taie L, Rozet JM, Busa T, Lacombe D, Naudion S, Amiel J, Rio M, Attie-Bitach T, Lesage C, Thouvenin D, Odent S, Morel G, Vincent-Delorme C, Boute O, Vanlerberghe C, Dieux A, Boussion S, Faivre L, Pinson L, Laffargue F, Le Guyader G, Le Meur G, Prieur F, Lambert V, Laudier B, Cottereau E, Ayuso C, Corton-Pérez M, Bouneau L, Le Caignec C, Gaston V, Jeanton-Scaramouche C, Dupin-Deguine D, Calvas P, Chassaing N, Plaisancié J. First evidence of SOX2 mutations in Peters' anomaly: lessons from molecular screening of 95 patients. Clin Genet 2022; 101:494-506. [PMID: 35170016 DOI: 10.1111/cge.14123] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 11/30/2022]
Abstract
Peters' anomaly (PA) is a rare anterior segment dysgenesis characterized by central corneal opacity and irido-lenticulo-corneal adhesions. Several genes are involved in syndromic or isolated PA (B3GLCT, PAX6, PITX3, FOXE3, CYP1B1). Some Copy Number Variations (CNVs) have also been occasionally reported. Despite this genetic heterogeneity, most of patients remain without genetic diagnosis. We retrieved a cohort of 95 individuals with PA and performed genotyping using a combination of Comparative genomic hybridization, whole genome, exome and targeted sequencing of 119 genes associated with ocular development anomalies. Causative genetic defects involving 12 genes and CNVs were identified for 1/3 of patients. Unsurprisingly, B3GLCT and PAX6 were the most frequently implicated genes, respectively in syndromic and isolated PA. Unexpectedly, the third gene involved in our cohort was SOX2, the major gene of micro-anophthalmia. Four unrelated patients with PA (isolated or with microphthalmia) were carrying pathogenic variants in this gene that was never associated with PA before. Here we described the largest cohort of PA patients ever reported. The genetic bases of PA are still to be explored as genetic diagnosis was unavailable for 2/3 of patients. Nevertheless, we showed here for the first time the involvement of SOX2 in PA, offering new evidence for its role in corneal transparency and anterior segment development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bertrand Chesneau
- Génétique Médicale, Hôpital Purpan, CHU, Toulouse, France.,Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU, Toulouse, France
| | | | - Félix Fremont
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU, Toulouse, France.,Service d'ophtalmologie, Hôpital Purpan, CHU Toulouse, France
| | - Jacmine Pechmeja
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU, Toulouse, France.,Service d'ophtalmologie, Hôpital Purpan, CHU Toulouse, France
| | - Vincent Soler
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU, Toulouse, France.,Service d'ophtalmologie, Hôpital Purpan, CHU Toulouse, France
| | - Bertrand Isidor
- Génétique Médicale, Institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Mathilde Nizon
- Génétique Médicale, Institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Hélène Dollfus
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), Hôpitaux Universitaires, Strasbourg, France
| | - Josseline Kaplan
- Laboratoire de Génétique Ophtalmologique, INSERM U1163, Institut Imagine, Paris, France
| | - Lucas Fares-Taie
- Laboratoire de Génétique Ophtalmologique, INSERM U1163, Institut Imagine, Paris, France
| | - Jean-Michel Rozet
- Laboratoire de Génétique Ophtalmologique, INSERM U1163, Institut Imagine, Paris, France
| | - Tiffany Busa
- Génétique Clinique, AP- HM CHU Timone Enfants, Marseille, France
| | - Didier Lacombe
- Département de Génétique Médicale, CHU Bordeaux, Bordeaux, France
| | - Sophie Naudion
- Département de Génétique Médicale, CHU Bordeaux, Bordeaux, France
| | - Jeanne Amiel
- Service de Génétique Médicale, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Marlène Rio
- Service de Génétique Médicale, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Tania Attie-Bitach
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, AP-, HP, Paris, France
| | | | | | - Sylvie Odent
- Service de Génétique Clinique, Centre Labellisé pour les Anomalies du Développement Ouest, CHU Rennes; Institut de Génétique et Développement de Rennes, CNRS, UMR 6290, Université de Rennes, ERN ITHACA, France
| | - Godelieve Morel
- Service de Génétique Clinique, Centre Labellisé pour les Anomalies du Développement Ouest, CHU Rennes; Institut de Génétique et Développement de Rennes, CNRS, UMR 6290, Université de Rennes, ERN ITHACA, France
| | | | | | | | | | | | - Laurence Faivre
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, CHU, Dijon, France
| | - Lucile Pinson
- Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, CHU de Montpellier, France
| | | | | | | | | | - Victor Lambert
- Service d'ophtalmologie, Hôpital Nord, Saint-Etienne, France
| | | | | | - Carmen Ayuso
- Genetics & Genomics Department, Jiménez Díaz University Hospital-Universidad Autónoma de Madrid (IIS-FJD-UAM). Centre for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Marta Corton-Pérez
- Genetics & Genomics Department, Jiménez Díaz University Hospital-Universidad Autónoma de Madrid (IIS-FJD-UAM). Centre for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, Spain
| | | | | | | | | | | | - Patrick Calvas
- Génétique Médicale, Hôpital Purpan, CHU, Toulouse, France.,Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU, Toulouse, France
| | - Nicolas Chassaing
- Génétique Médicale, Hôpital Purpan, CHU, Toulouse, France.,Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU, Toulouse, France
| | - Julie Plaisancié
- Génétique Médicale, Hôpital Purpan, CHU, Toulouse, France.,Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU, Toulouse, France.,INSERM U1214, ToNIC, Université Toulouse III, France
| |
Collapse
|
19
|
Imelda E, Gunawan F. Clinical management of a rare Peters' anomaly-induced secondary childhood glaucoma: A case report. NARRA J 2021; 1:e53. [PMID: 38450209 PMCID: PMC10914085 DOI: 10.52225/narraj.v1i3.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/01/2021] [Indexed: 03/08/2024]
Abstract
Childhood glaucoma is a rare disorder that occurs from birth until teenage years caused by an abnormality of aqueous humor pathways. About 50-70% of Peters' anomaly is accompanied by secondary childhood glaucoma. The presence of glaucoma will affect the prognosis. We reported the evaluation and treatment of secondary childhood glaucoma due to Peters' anomaly. A 5 months-old boy was presented with the complaint of a enlarged left eye since 3 months old. The complaint was accompanied by a watering eye and frequently closed upon light exposure. The left eye looked opaquer than contralateral. Examination under anesthesia showed that the intraocular pressure (IOP) was 35 mmHg in the left eye and the corneal diameter was 14 mm. Other findings were keratopathy, diffuse corneal edema, buphthalmos, shallow anterior chamber, anterior synechiae, and linear slit shaped pupils in the nasal region. Patient was treated with ophthalmic timolol maleate which was later followed by trabeculectomy. After 1 week post-surgery, IOP assessment by palpation suggested the right eye within normal range while the IOP of left eye was higger than normal. Blepharospasm, epiphora, photophobia, bleb on superior, subconjunctiva bleeding, buphthalmos, keratopathy, minimal corneal edema, anterior chamber with shallow image, and posterior synechia were found in left eye anterior segment. In conclusion, trabeculotomy and trabeculectomy are recommended if there is no reduction of IOP observed after receiving timolol maleate therapy. The choice of surgical management is dependent on the feasibility of the protocol.
Collapse
Affiliation(s)
- Eva Imelda
- Department of Ophthalmology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Dr Zainoel Abidin Hospital, Banda Aceh, Indonesia
| | | |
Collapse
|
20
|
Latta L, Figueiredo FC, Ashery-Padan R, Collinson JM, Daniels J, Ferrari S, Szentmáry N, Solá S, Shalom-Feuerstein R, Lako M, Xapelli S, Aberdam D, Lagali N. Pathophysiology of aniridia-associated keratopathy: Developmental aspects and unanswered questions. Ocul Surf 2021; 22:245-266. [PMID: 34520870 DOI: 10.1016/j.jtos.2021.09.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/19/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022]
Abstract
Aniridia, a rare congenital disease, is often characterized by a progressive, pronounced limbal insufficiency and ocular surface pathology termed aniridia-associated keratopathy (AAK). Due to the characteristics of AAK and its bilateral nature, clinical management is challenging and complicated by the multiple coexisting ocular and systemic morbidities in aniridia. Although it is primarily assumed that AAK originates from a congenital limbal stem cell deficiency, in recent years AAK and its pathogenesis has been questioned in the light of new evidence and a refined understanding of ocular development and the biology of limbal stem cells (LSCs) and their niche. Here, by consolidating and comparing the latest clinical and preclinical evidence, we discuss key unanswered questions regarding ocular developmental aspects crucial to AAK. We also highlight hypotheses on the potential role of LSCs and the ocular surface microenvironment in AAK. The insights thus gained lead to a greater appreciation for the role of developmental and cellular processes in the emergence of AAK. They also highlight areas for future research to enable a deeper understanding of aniridia, and thereby the potential to develop new treatments for this rare but blinding ocular surface disease.
Collapse
Affiliation(s)
- L Latta
- Dr. Rolf. M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Homburg, Saar, Germany; Department of Ophthalmology, Saarland University Medical Center, Homburg, Saar, Germany.
| | - F C Figueiredo
- Department of Ophthalmology, Royal Victoria Infirmary, Newcastle Upon Tyne, United Kingdom
| | - R Ashery-Padan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - J M Collinson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - J Daniels
- Cells for Sight, UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - S Ferrari
- The Veneto Eye Bank Foundation, Venice, Italy
| | - N Szentmáry
- Dr. Rolf. M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - S Solá
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - R Shalom-Feuerstein
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - M Lako
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - S Xapelli
- Instituto Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - D Aberdam
- Centre de Recherche des Cordeliers, INSERM U1138, Team 17, France; Université de Paris, 75006, Paris, France.
| | - N Lagali
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Ophthalmology, Sørlandet Hospital Arendal, Arendal, Norway.
| |
Collapse
|
21
|
Diel H, Ding C, Grehn F, Chronopoulos P, Bartsch O, Hoffmann EM. First observation of secondary childhood glaucoma in Coffin-Siris syndrome: a case report and literature review. BMC Ophthalmol 2021; 21:28. [PMID: 33430815 PMCID: PMC7802219 DOI: 10.1186/s12886-020-01788-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Severe congenital ophthalmological malformations and glaucoma might be an important occasional feature in patients with Coffin-Siris syndrome (CSS), especially Coffin-Siris syndrome 9 (CSS9, OMIM #615866) caused by SOX11 mutation. Recently, primary (open-angle) glaucoma was described in two children with the most common form of Coffin-Siris syndrome, CSS1 (OMIM #135900) by ARID1B (AT-rich interaction domain-containing protein 1B) gene mutation. In this article, we present the first report of glaucoma with Coffin-Siris syndrome 9 as well as the first report of secondary glaucoma with any form of Coffin-Siris syndrome. These findings indicate that secondary glaucoma is an occasional finding in patients with Coffin-Siris syndrome. CASE PRESENTATION A child with secondary childhood glaucoma and additional ocular manifestations was evaluated and treated at the childhood glaucoma centre in Mainz, Germany. Examination under general anaesthesia revealed ocular anterior segment dysgenesis (ASD) (Peters type iridocorneal dysgenesis) in combination with congenital limbal stem cell deficiency (LSCD), aniridia, and cataract. The patient also had multiple other congenital anomalies and severe developmental delay. To explain his combination of anomalies, molecular genetic analysis from peripheral blood was performed in late 2018 and early 2019. Following normal findings with a panel diagnostic of 18 genes associated with congenital glaucoma, whole exome sequencing was performed and revealed a novel likely pathogenic heterozygous variant c.251G>T, p.(Gly84Val) in the SOX11 gene (SRY-related HMG-box gene 11). The variant had occurred de novo. Thus, the multiple congenital anomalies and developmental delay of the patient represented Coffin-Siris syndrome 9 (CSS9, OMIM #615866). CONCLUSIONS When eye diseases occur in combination with other systemic features, genetic analysis can be seminal. Results indicate that glaucoma is an occasional feature of patients with Coffin-Siris syndrome. As early treatment may improve the visual outcome of patients with glaucoma, we suggest that patients with Coffin-Siris syndrome should receive specific ophthalmological screening.
Collapse
Affiliation(s)
- Heidi Diel
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, D – 55131 Mainz, Germany
| | - Can Ding
- Institute of Human Genetics, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Franz Grehn
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, D – 55131 Mainz, Germany
| | - Panagiotis Chronopoulos
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, D – 55131 Mainz, Germany
| | - Oliver Bartsch
- Institute of Human Genetics, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Esther M. Hoffmann
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, D – 55131 Mainz, Germany
| |
Collapse
|
22
|
Li Y, Zhang J, Dai Y, Fan Y, Xu J. Novel Mutations in COL6A3 That Associated With Peters' Anomaly Caused Abnormal Intracellular Protein Retention and Decreased Cellular Resistance to Oxidative Stress. Front Cell Dev Biol 2020; 8:531986. [PMID: 33304895 PMCID: PMC7693641 DOI: 10.3389/fcell.2020.531986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 09/22/2020] [Indexed: 11/13/2022] Open
Abstract
Peters' anomaly (PA) is a rare form of anterior segment dysgenesis characterized by central corneal opacity accompanied by iridocorneal or lenticulo-corneal adhesions. Although genetic mutations, particularly those affecting transcription factors that function in eye development, are known to cause PA, the etiology of this disease remains poorly understood. In this study, 23 patients with PA were recruited for panel sequencing. Four out of 23 patients were found to carry variants in known PA causal genes, PITX2 and PITX3. More importantly, two homozygous mutations (NM_057164: p.Val86Ala and p.Arg689Cys) in the COL6A3 gene (collagen type VI alpha-3 chain) that correlated with the phenotype of type I PA were identified, and then validated by following whole-exome sequencing. The expression profile of the COL6A3 gene in the cornea and the impact of the mutations on protein physiological processing and cellular function were further explored. It was shown that COL6A3 presented relatively high expression in the cornea. The mutant COL6A3 protein was relatively retained intracellularly, and its expression reduced cellular resistance to oxidative stress through an enhanced endoplasmic reticulum stress response. Taken together, our findings expanded the known genetic spectrum of PA, and provided evidence for the involvement of COL6A3 or collagen VI in ocular anterior segment development, thereby offering new insight for future investigations targeting PA.
Collapse
Affiliation(s)
- Yue Li
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jing Zhang
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yiqin Dai
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yidan Fan
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jianjiang Xu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
23
|
Morphometric analysis of the lens in human aniridia and mouse Small eye. Exp Eye Res 2020; 203:108371. [PMID: 33248069 DOI: 10.1016/j.exer.2020.108371] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/17/2020] [Accepted: 11/22/2020] [Indexed: 12/16/2022]
Abstract
Congenital aniridia is caused by heterozygous mutations in the PAX6 gene. In this disease, congenital iris and foveal hypoplasia is associated with juvenile onset cataract, glaucoma, and corneal keratopathy. In rodents, Pax6 mutations result in a congenital reduction in ocular size that is not typically described in human aniridia. Here, the ocular morphometry of aniridia patients is compared with the lens phenotype of Pax6+/tm1/Pgr mice to reveal whether there are species differences in Pax6 regulation of lens development and homeostasis. Ultrasound biometry (UBM) revealed that eleven percent of aniridia patients exhibited mild microphthalmia while the anterior chamber depth of aniridic eyes was significantly reduced from 6 months of age onward. Although aniridic lens thickness was normal from birth, it was significantly decreased in aniridic lenses older than 30. Notably, 86% of aniridic lenses exhibited cataractous changes in this cohort. In addition, a significant proportion of aniridia patients develop lens subluxation as they age associated with reduced lens diameter as measured by anterior segment optical coherence tomography (AS-OCT). Analysis of young adult Pax6+/tm1/Pgr mouse lenses by micro-computed tomography (microCT), bright field and dark field imaging revealed that they are reduced in size but did not exhibit overt cataracts at this age. Overall, this study reveals that congenital microphthalmia as assessed by axial length, or microphakia, as assessed by lens thickness, are not typical in human aniridia, although these are primary manifestations of Pax6 mutations in mice, suggesting that PAX6 regulates some aspects of lens development differently between these species.
Collapse
|
24
|
Darbari E, Zare-Abdollahi D, Alavi A, Rezaei Kanavi M, Feizi S, Hosseini SB, Baradaran-Rafii A, Ahmadieh H, Issazadeh-Navikas S, Elahi E. A mutation in DOP1B identified as a probable cause for autosomal recessive Peters anomaly in a consanguineous family. Mol Vis 2020; 26:757-765. [PMID: 33273802 PMCID: PMC7700884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 11/23/2020] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Peters anomaly (PA) is a heterogeneous developmental disorder characterized by central corneal opacity and iridocorneal or corneolenticular adhesions. Although many causative genes have been identified, most screened patients do not have mutations in the known genes. We aimed to identify the genetic cause of Peters anomaly in a pedigree with three affected individuals. METHODS Slit-lamp biomicroscopy and ultrasound biomicroscopy were performed for definitive diagnosis. Exome sequencing was conducted on the DNA of all three patients. After identification of a candidate causative gene, expression of the gene was assessed with real-time PCR in various ocular tissues of three human embryos and three adults. RESULTS The patients were affected with isolated PA. The parents of the patients were related to one another. Inheritance of PA was autosomal recessive. After appropriate filtering of the exome data, a homozygous variation in DOP1B remained as the only candidate genetic cause of PA in the pedigree. The variant segregated with disease status in the pedigree and was absent among 800 control Iranians. The variant has been reported in various databases at frequencies of 0.006 or less only in the heterozygous state in some cohorts of African origin. The p.Val1660 amino acid affected by the mutation is completely conserved in mammals and birds during evolution. Expression of DOP1B was shown in all adult and embryonic lens, iris, cornea, sclera, and retina tissues that were tested. CONCLUSIONS DOP1B that encodes DOP1 leucine zipper like protein B was identified as the putative PA-causing gene in pedigree PA-101. As DOP1B is positioned within the Down syndrome chromosomal region on chromosome 21, until now this gene has mostly been studied with respect to brain functions. However, members of the Dopey gene family have been shown to have roles in development in other organisms. Evidence of the expression of DOP1B in various PA-relevant eye tissues, which, to the best of our knowledge, is shown here for the first time, is to be noted. However, this finding does not necessarily implicate a specific role for DOP1B in eye development as the gene is expressed in many tissues. Ultimately, definitive assessment of the contribution of DOP1B to PA pathology awaits identification of mutations in the gene in unrelated patients with PA and functional studies.
Collapse
Affiliation(s)
- Ensieh Darbari
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Davood Zare-Abdollahi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Afagh Alavi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Feizi
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Alireza Baradaran-Rafii
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ahmadieh
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, Copenhagen Biocentre, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
25
|
Chang YL, Chao AS, Chou CY, Chang SD, Chiang MC, Lee YS. A donor twin discordant with Peters anomaly in a twin-twin transfusion syndrome case: a case report. BMC Pregnancy Childbirth 2020; 20:558. [PMID: 32967640 PMCID: PMC7513296 DOI: 10.1186/s12884-020-03269-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/18/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Peters anomaly is a rare form of anterior segment ocular dysgenesis, the antenatal image of Peters anomaly had not been reported. We herein showcased a discordant finding of Peters anomaly in a monozygotic twin complicated with twin-twin transfusion syndrome (TTTS) and exhibited its antenatal sonographic images, CASE PRESENTATION: A 38-year-old gravida 2 para 1 pregnant woman visited our clinic at the gestational age of 18 weeks where TTTS stage III was diagnosed and the following laser therapy was done successfully. Ten days after the surgery, the follow-up ultrasound detected the opacity of both fetal eyeballs in the donor twin and thus congenital cataract was suspected initially. Then magnetic resonance imaging (MRI) examination was arranged at the gestational age of 23 weeks, and no central nervous system or other anomaly was found. At the 29 weeks of gestation, the opacity of both fetal eyeballs of the donor twin did not clear. The pregnancy resulted in cesarean section at the gestational age of 37 weeks indicated by malpresentation where two male live births were born. Examination under anesthesia was arranged for donor twin after delivery and Peters anomaly was diagnosed based on central corneal opacity with iridocorneal and corneolenticular adhesions. CONCLUSIONS The prenatal image of Peters anomaly may present as the opacity of the fetal eyeballs similar to congenital cataract. Some cases of the Peters anomaly had been reported with a genetic abnormality, but since our case presented discordant presentation in monozygotic twin pregnancy where both twins are supposed to share the same genetic make-up, therefore other factors that are epigenetic may be held accountable. Nevertheless, a genetic origin of the anomaly in our case cannot be excluded.
Collapse
Affiliation(s)
- Yao-Lung Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan.
| | - An-Shine Chao
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Ching-Yu Chou
- Department of Obstetrics and Gynecology, Cathay General Hospital, Hsinchu, Taiwan
| | - Shuenn-Dyh Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Ming-Chou Chiang
- Department of pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Yung-Sung Lee
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taiwan
| |
Collapse
|
26
|
Kletke SN, Vincent A, Maynes JT, Elbaz U, Mireskandari K, Lam WC, Ali A. A de novo mutation in PITX2 underlies a unique form of Axenfeld-Rieger syndrome with corneal neovascularization and extensive proliferative vitreoretinopathy. Ophthalmic Genet 2020; 41:358-362. [PMID: 32429730 DOI: 10.1080/13816810.2020.1768556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/20/2020] [Accepted: 05/09/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Axenfeld-Rieger syndrome is characterized by a spectrum of anterior segment dysgenesis involving neural-crest-derived tissues, most commonly secondary to mutations in the transcription factor genes PITX2 and FOXC1. MATERIALS AND METHODS Single retrospective case report. RESULTS A full-term infant presented at 5 weeks of age with bilateral Peters anomaly and Axenfeld-Rieger syndrome, with development of atypical features of progressive corneal neovascularization and proliferative vitreoretinopathy. Despite surgical interventions, the patient progressed to bilateral phthisis bulbi by 22 months of age. Genetic testing revealed a novel de novo p.Leu212Valfs*39 mutation in PITX2, leading to loss of a C-terminal OAR domain that functions in transcriptional regulation. CONCLUSIONS It is important to consider mutations in PITX2 in atypical cases of anterior segment dysgenesis that also present with abnormalities in the angiogenesis of the anterior and posterior segments.
Collapse
Affiliation(s)
- Stephanie N Kletke
- Department of Ophthalmology & Vision Sciences, University of Toronto , Toronto, Canada
- Department of Ophthalmology & Vision Sciences, The Hospital for Sick Children , Toronto, Canada
| | - Ajoy Vincent
- Department of Ophthalmology & Vision Sciences, University of Toronto , Toronto, Canada
- Department of Ophthalmology & Vision Sciences, The Hospital for Sick Children , Toronto, Canada
- Genetics and Genome Biology, SickKids Research Institute , Toronto, Canada
| | - Jason T Maynes
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children , Toronto, Canada
- Department of Anesthesia, University of Toronto , Toronto, Canada
- Program in Molecular Medicine, SickKids Research Institute , Toronto, Canada
| | - Uri Elbaz
- Department of Ophthalmology, Rabin Medical Center , Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv, Israel
| | - Kamiar Mireskandari
- Department of Ophthalmology & Vision Sciences, University of Toronto , Toronto, Canada
- Department of Ophthalmology & Vision Sciences, The Hospital for Sick Children , Toronto, Canada
| | - Wai-Ching Lam
- Department of Ophthalmology & Vision Sciences, The Hospital for Sick Children , Toronto, Canada
- Department of Ophthalmology, The University of Hong Kong , Hong Kong, China
| | - Asim Ali
- Department of Ophthalmology & Vision Sciences, University of Toronto , Toronto, Canada
- Department of Ophthalmology & Vision Sciences, The Hospital for Sick Children , Toronto, Canada
| |
Collapse
|
27
|
Johnson AL, Schneider JE, Mohun TJ, Williams T, Bhattacharya S, Henderson DJ, Phillips HM, Bamforth SD. Early Embryonic Expression of AP-2α Is Critical for Cardiovascular Development. J Cardiovasc Dev Dis 2020; 7:jcdd7030027. [PMID: 32717817 PMCID: PMC7570199 DOI: 10.3390/jcdd7030027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
Congenital cardiovascular malformation is a common birth defect incorporating abnormalities of the outflow tract and aortic arch arteries, and mice deficient in the transcription factor AP-2α (Tcfap2a) present with complex defects affecting these structures. AP-2α is expressed in the pharyngeal surface ectoderm and neural crest at mid-embryogenesis in the mouse, but the precise tissue compartment in which AP-2α is required for cardiovascular development has not been identified. In this study we describe the fully penetrant AP-2α deficient cardiovascular phenotype on a C57Bl/6J genetic background and show that this is associated with increased apoptosis in the pharyngeal ectoderm. Neural crest cell migration into the pharyngeal arches was not affected. Cre-expressing transgenic mice were used in conjunction with an AP-2α conditional allele to examine the effect of deleting AP-2α from the pharyngeal surface ectoderm and the neural crest, either individually or in combination, as well as the second heart field. This, surprisingly, was unable to fully recapitulate the global AP-2α deficient cardiovascular phenotype. The outflow tract and arch artery phenotype was, however, recapitulated through early embryonic Cre-mediated recombination. These findings indicate that AP-2α has a complex influence on cardiovascular development either being required very early in embryogenesis and/or having a redundant function in many tissue layers.
Collapse
Affiliation(s)
- Amy-Leigh Johnson
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
| | | | | | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anshutz Medical Campus, Aurora, CO 80045, USA;
| | - Shoumo Bhattacharya
- Department of Cardiovascular Medicine, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford OX3 7BN, UK;
| | - Deborah J. Henderson
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
| | - Helen M. Phillips
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
| | - Simon D. Bamforth
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
- Correspondence: ; Tel.: +44-191-241-8764
| |
Collapse
|
28
|
Revealing hidden genetic diagnoses in the ocular anterior segment disorders. Genet Med 2020; 22:1623-1632. [PMID: 32499604 PMCID: PMC7521990 DOI: 10.1038/s41436-020-0854-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose Ocular anterior segment disorders (ASDs) are clinically and genetically heterogeneous, and genetic diagnosis often remains elusive. In this study, we demonstrate the value of a combined analysis protocol using phenotypic, genomic, and pedigree structure data to achieve a genetic conclusion. Methods We utilized a combination of chromosome microarray, exome sequencing, and genome sequencing with structural variant and trio analysis to investigate a cohort of 41 predominantly sporadic cases. Results We identified likely causative variants in 54% (22/41) of cases, including 51% (19/37) of sporadic cases and 75% (3/4) of cases initially referred as familial ASD. Two-thirds of sporadic cases were found to have heterozygous variants, which in most cases were de novo. Approximately one-third (7/22) of genetic diagnoses were found in rarely reported or recently identified ASD genes including PXDN, GJA8, COL4A1, ITPR1, CPAMD8, as well as the new phenotypic association of Axenfeld–Rieger anomaly with a homozygous ADAMTS17 variant. The remainder of the variants were in key ASD genes including FOXC1, PITX2, CYP1B1, FOXE3, and PAX6. Conclusions We demonstrate the benefit of detailed phenotypic, genomic, variant, and segregation analysis to uncover some of the previously “hidden” heritable answers in several rarely reported and newly identified ocular ASD-related disease genes.
Collapse
|
29
|
Spectrum of Genetic Variants Associated with Anterior Segment Dysgenesis in South Florida. Genes (Basel) 2020; 11:genes11040350. [PMID: 32224865 PMCID: PMC7230952 DOI: 10.3390/genes11040350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/18/2020] [Accepted: 03/25/2020] [Indexed: 12/02/2022] Open
Abstract
Anterior segment dysgenesis (ASD) comprises a wide spectrum of developmental conditions affecting the cornea, iris, and lens, which may be associated with abnormalities of other organs. To identify disease-causing variants, we performed exome sequencing in 24 South Florida families with ASD. We identified 12 likely causative variants in 10 families (42%), including single nucleotide or small insertion–deletion variants in B3GLCT, BMP4, CYP1B1, FOXC1, FOXE3, GJA1, PXDN, and TP63, and a large copy number variant involving PAX6. Four variants were novel. Each variant was detected only in one family. Likely causative variants were detected in 1 out of 7 black and 9 out of 17 white families. In conclusion, exome sequencing for ASD allows us to identify a wide spectrum of rare DNA variants in South Florida. Further studies will explore missing variants, especially in the black communities.
Collapse
|
30
|
Thieu T, Milman T, Bhatti TR, Eagle RC. Anterior Segment Dysgenesis With Accessory Iris Membranes in an Infant With Otopalatodigital Spectrum Disorder and Mutation in the FLNA Gene. J Pediatr Ophthalmol Strabismus 2020; 57:e8-e11. [PMID: 31978233 DOI: 10.3928/01913913-20191230-02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/05/2019] [Indexed: 11/20/2022]
Abstract
A 4-month-old male infant with frontometaphyseal dysplasia and de novo FLNA gene mutation died of complications of disease. Post-mortem examination revealed accessory iris membranes. This is the first report in the literature of accessory iris membranes in a confirmed case of FLNA mutation and phenotypic anomalies consistent with frontometaphyseal dysplasia. [J Pediatr Ophthalmol Strabismus. 2020;57:e8-e11.].
Collapse
|
31
|
Contribution of a Novel B3GLCT Variant to Peters Plus Syndrome Discovered by a Combination of Next-Generation Sequencing and Automated Text Mining. Int J Mol Sci 2019; 20:ijms20236006. [PMID: 31795264 PMCID: PMC6928627 DOI: 10.3390/ijms20236006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/07/2019] [Accepted: 11/26/2019] [Indexed: 12/30/2022] Open
Abstract
Anterior segment dysgenesis (ASD) encompasses a spectrum of ocular disorders affecting the structures of the anterior eye chamber. Mutations in several genes, involved in eye development, are implicated in this disorder. ASD is often accompanied by diverse multisystemic symptoms and another genetic cause, such as variants in genes encoding collagen type IV. Thus, a wide spectrum of phenotypes and underlying genetic diversity make fast and proper diagnosis challenging. Here, we used AMELIE, an automatic text mining tool that enriches data with the most up-to-date information from literature, and wANNOVAR, which is based on well-documented databases and incorporates variant filtering strategy to identify genetic variants responsible for severely-manifested ASD in a newborn child. This strategy, applied to trio sequencing data in compliance with ACMG 2015 guidelines, helped us find two compound heterozygous variants of the B3GLCT gene, of which c.660+1G>A (rs80338851) was previously associated with the phenotype of Peters plus syndrome (PPS), while the second, NM_194318.3:c.755delC (p.T252fs), in exon 9 of the same gene was noted for the first time. PPS, a very rare subtype of ASD, is a glycosylation disorder, where the dysfunctional B3GLCT gene product, O-fucose-specific β-1,3-glucosyltransferase, is ineffective in providing a noncanonical quality control system for proper protein folding in cells. Our study expands the mutation spectrum of the B3GLCT gene related to PPS. We suggest that the implementation of automatic text mining tools in combination with careful variant filtering could help translate sequencing results into diagnosis, thus, considerably accelerating the diagnostic process and, thereby, improving patient management.
Collapse
|
32
|
Reis LM, Houssin NS, Zamora C, Abdul-Rahman O, Kalish JM, Zackai EH, Plageman TF, Semina EV. Novel variants in CDH2 are associated with a new syndrome including Peters anomaly. Clin Genet 2019; 97:502-508. [PMID: 31650526 DOI: 10.1111/cge.13660] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 11/26/2022]
Abstract
Peters anomaly (PA) is a congenital corneal opacity associated with corneo-lenticular attachments. PA can be isolated or part of a syndrome with most cases remaining genetically unsolved. Exome sequencing of a trio with syndromic PA and 145 additional unexplained probands with developmental ocular conditions identified a de novo splicing and three novel missense heterozygous CDH2 variants affecting the extracellular cadherin domains in four individuals with PA. Syndromic anomalies were seen in three individuals and included left-sided cardiac lesions, dysmorphic facial features, and decreasing height percentiles; brain magnetic resonance imaging identified agenesis of the corpus callosum and hypoplasia of the inferior cerebellar vermis. CDH2 encodes for N-cadherin, a transmembrane protein that mediates cell-cell adhesion in multiple tissues. Immunostaining in mouse embryonic eyes confirmed N-cadherin is present in the lens stalk at the time of separation from the future cornea and in the developing lens and corneal endothelium at later stages, supporting a possible role in PA. Previous studies in animal models have noted the importance of Cdh2/cdh2 in the development of the eye, heart, brain, and skeletal structures, also consistent with the patient features presented here. Examination of CDH2 in additional patients with PA is indicated to confirm this association.
Collapse
Affiliation(s)
- Linda M Reis
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Carlos Zamora
- Department of Radiology, Division of Neuroradiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Omar Abdul-Rahman
- Genetic Medicine, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jennifer M Kalish
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Elaine H Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, Philadelphia, Pennsylvania
| | | | - Elena V Semina
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Ophthalmology and Visual Sciences, Department of Cell Biology, Neurobiology and Anatomy, Children's Research Institute, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
33
|
Valensi M, Goldman G, Marchant D, Van Den Berghe L, Jonet L, Daruich A, Robert MP, Krejci E, Klein C, Mascarelli F, Versaux-Botteri C, Moulin A, Putterman M, Guimiot F, Molina T, Terris B, Brémond-Gignac D, Behar-Cohen F, Abitbol MM. Sostdc1 is expressed in all major compartments of developing and adult mammalian eyes. Graefes Arch Clin Exp Ophthalmol 2019; 257:2401-2427. [PMID: 31529323 DOI: 10.1007/s00417-019-04462-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/20/2019] [Accepted: 09/04/2019] [Indexed: 01/16/2023] Open
Abstract
PURPOSE This study was conducted in order to study Sostdc1 expression in rat and human developing and adult eyes. METHODS Using the yeast signal sequence trap screening method, we identified the Sostdc1 cDNA encoding a protein secreted by the adult rat retinal pigment epithelium. We determined by in situ hybridization, RT-PCR, immunohistochemistry, and western blot analysis Sostdc1 gene and protein expression in developing and postnatal rat ocular tissue sections. We also investigated Sostdc1 immunohistolocalization in developing and adult human ocular tissues. RESULTS We demonstrated a prominent Sostdc1 gene expression in the developing rat central nervous system (CNS) and eyes at early developmental stages from E10.5 days postconception (dpc) to E13 dpc. Specific Sostdc1 immunostaining was also detected in most adult cells of rat ocular tissue sections. We also identified the rat ocular embryonic compartments characterized by a specific Sostdc1 immunohistostaining and specific Pax6, Sox2, Otx2, and Vsx2 immunohistostaining from embryonic stages E10.5 to E13 dpc. Furthermore, we determined the localization of SOSTDC1 immunoreactivity in ocular tissue sections of developing and adult human eyes. Indeed, we detected SOSTDC1 immunostaining in developing and adult human retinal pigment epithelium (RPE) and neural retina (NR) as well as in several developing and adult human ocular compartments, including the walls of choroidal and scleral vessels. Of utmost importance, we observed a strong SOSTDC1 expression in a pathological ocular specimen of type 2 Peters' anomaly complicated by retinal neovascularization as well in the walls ofother pathological extra-ocular vessels. CONCLUSION: As rat Sostdc1 and human SOSTDC1 are dual antagonists of the Wnt/β-catenin and BMP signaling pathways, these results underscore the potential crucial roles of these pathways and their antagonists, such as Sostdc1 and SOSTDC1, in developing and adult mammalian normal eyes as well as in syndromic and nonsyndromic congenital eye diseases.
Collapse
Affiliation(s)
- Maud Valensi
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
| | - Gabrielle Goldman
- APHP, Service de Pathologie de L'Hôpital Cochin-Hôtel-Dieu, Université Paris Descartes, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Dominique Marchant
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
- Sorbonne Paris Cité, UFR SMBH, Laboratoire Hypoxie et poumons, Université Paris 13, EA 2363, 93017, Bobigny, France
| | - Loïc Van Den Berghe
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
- Inserm UMR 1037, CRCT (Cancer Research Center of Toulouse), 31037, Toulouse, France
| | - Laurent Jonet
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
| | - Alejandra Daruich
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
- AP-HP, Hôpital Universitaire Necker-Enfants-Malades, Service d'Ophtalmologie, 149 rue de Sèvres, 75015, Paris, France
| | - Matthieu P Robert
- AP-HP, Hôpital Universitaire Necker-Enfants-Malades, Service d'Ophtalmologie, 149 rue de Sèvres, 75015, Paris, France
- COGnition and Action Group, UMR 8257, CNRS, Université Paris Descartes, Paris, France
| | - Eric Krejci
- COGnition and Action Group, UMR 8257, CNRS, Université Paris Descartes, Paris, France
| | - Christophe Klein
- Centre d'Imagerie Cellulaire et de Cytométrie (CICC), Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie - Paris 6, Université Paris Descartes - Paris 5, UMR_S 1138, 75006, Paris, France
| | - Frédéric Mascarelli
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
| | - Claudine Versaux-Botteri
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
| | - Alexandre Moulin
- Département de Pathologie, Hôpital Ophtalmique Jules-Gonin , 15, avenue de France, 1004, Lausanne, Switzerland
| | - Marc Putterman
- APHP, Service de Pathologie de l'Hôpital Universitaire Necker-Enfants-Malades, Université Paris Descartes, 149 rue de Sèvres, 75015, Paris, France
| | - Fabien Guimiot
- Unité Fonctionnelle de Foeto-Pathologie, Hôpital Universitaire Robert Debré, 48 Boulevard Serrurier, 75019, Paris, France
| | - Thierry Molina
- APHP, Service de Pathologie de l'Hôpital Universitaire Necker-Enfants-Malades, Université Paris Descartes, 149 rue de Sèvres, 75015, Paris, France
| | - Benoît Terris
- APHP, Service de Pathologie de L'Hôpital Cochin-Hôtel-Dieu, Université Paris Descartes, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Dominique Brémond-Gignac
- AP-HP, Hôpital Universitaire Necker-Enfants-Malades, Service d'Ophtalmologie, 149 rue de Sèvres, 75015, Paris, France
| | - Francine Behar-Cohen
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
- AP-HP, Service d'Ophtalmologie, Hôpital Universitaire Cochin-Hôtel-Dieu, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Marc M Abitbol
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France.
- AP-HP, Hôpital Universitaire Necker-Enfants-Malades, Service d'Ophtalmologie, 149 rue de Sèvres, 75015, Paris, France.
| |
Collapse
|
34
|
Reis LM, Sorokina EA, Thompson S, Muheisen S, Velinov M, Zamora C, Aylsworth AS, Semina EV. De Novo Missense Variants in WDR37 Cause a Severe Multisystemic Syndrome. Am J Hum Genet 2019; 105:425-433. [PMID: 31327510 PMCID: PMC6698968 DOI: 10.1016/j.ajhg.2019.06.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 06/14/2019] [Indexed: 01/06/2023] Open
Abstract
While genetic causes are known for many syndromes involving developmental anomalies, a large number of individuals with overlapping phenotypes remain undiagnosed. Using exome-sequencing analysis and review of matchmaker databases, we have discovered four de novo missense variants predicted to affect the N-terminal region of WDR37-p.Ser119Phe, p.Thr125Ile, p.Ser129Cys, and p.Thr130Ile-in unrelated individuals with a previously unrecognized syndrome. Features of WDR37 syndrome include the following: ocular anomalies such as corneal opacity/Peters anomaly, coloboma, and microcornea; dysmorphic facial features; significant neurological impairment with structural brain defects and seizures; poor feeding; poor post-natal growth; variable skeletal, cardiac, and genitourinary defects; and death in infancy in one individual. WDR37 encodes a protein of unknown function with seven predicted WD40 domains and no previously reported human pathogenic variants. Immunocytochemistry and western blot studies showed that wild-type WDR37 is localized predominantly in the cytoplasm and mutant proteins demonstrate similar protein levels and localization. CRISPR-Cas9-mediated genome editing generated zebrafish mutants with novel missense and frameshift alleles: p.Ser129Phe, p.Ser129Cys (which replicates one of the human variants), p.Ser129Tyr, p.Lys127Cysfs, and p.Gln95Argfs. Zebrafish carrying heterozygous missense variants demonstrated poor growth and larval lethality, while heterozygotes with frameshift alleles survived to adulthood, suggesting a potential dominant-negative mechanism for the missense variants. RNA-seq analysis of zebrafish embryos carrying a missense variant detected significant upregulation of cholesterol biosynthesis pathways. This study identifies variants in WDR37 associated with human disease and provides insight into its essential role in vertebrate development and possible molecular functions.
Collapse
Affiliation(s)
- Linda M Reis
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI 53226, USA
| | - Elena A Sorokina
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI 53226, USA
| | - Samuel Thompson
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI 53226, USA
| | - Sanaa Muheisen
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI 53226, USA
| | - Milen Velinov
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Carlos Zamora
- Department of Radiology, Division of Neuroradiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Arthur S Aylsworth
- Departments of Pediatrics and Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elena V Semina
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI 53226, USA; Departments of Ophthalmology and Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
35
|
Ohuchi H, Sato K, Habuta M, Fujita H, Bando T. Congenital eye anomalies: More mosaic than thought? Congenit Anom (Kyoto) 2019; 59:56-73. [PMID: 30039880 DOI: 10.1111/cga.12304] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022]
Abstract
The eye is a sensory organ that primarily captures light and provides the sense of sight, as well as delivering non-visual light information involving biological rhythms and neurophysiological activities to the brain. Since the early 1990s, rapid advances in molecular biology have enabled the identification of developmental genes, genes responsible for human congenital diseases, and relevant genes of mutant animals with various anomalies. In this review, we first look at the development of the eye, and we highlight seminal reports regarding archetypal gene defects underlying three developmental ocular disorders in humans: (1) holoprosencephaly (HPE), with cyclopia being exhibited in the most severe cases; (2) microphthalmia, anophthalmia, and coloboma (MAC) phenotypes; and (3) anterior segment dysgenesis (ASDG), known as Peters anomaly and its related disorders. The recently developed methods, such as next-generation sequencing and genome editing techniques, have aided the discovery of gene mutations in congenital eye diseases and gene functions in normal eye development. Finally, we discuss Pax6-genome edited mosaic eyes and propose that somatic mosaicism in developmental gene mutations should be considered a causal factor for variable phenotypes, sporadic cases, and de novo mutations in human developmental disorders.
Collapse
Affiliation(s)
- Hideyo Ohuchi
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keita Sato
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Munenori Habuta
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hirofumi Fujita
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tetsuya Bando
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
36
|
Phenotype–genotype correlations and emerging pathways in ocular anterior segment dysgenesis. Hum Genet 2018; 138:899-915. [DOI: 10.1007/s00439-018-1935-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022]
|
37
|
Plaisancié J, Ragge N, Dollfus H, Kaplan J, Lehalle D, Francannet C, Morin G, Colineaux H, Calvas P, Chassaing N. FOXE3
mutations: genotype-phenotype correlations. Clin Genet 2018; 93:837-845. [DOI: 10.1111/cge.13177] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 01/25/2023]
Affiliation(s)
- J. Plaisancié
- Service de Génétique Médicale, Hôpital Purpan, CHU Toulouse; Toulouse France
- INSERM U1056; Université Toulouse III; Toulouse France
| | - N.K. Ragge
- Faculty of Health and Life Sciences; Oxford Brookes University; Oxford UK
- West Midlands Regional Genetics Service; Birmingham Women and Children’s NHS Foundation Trust; Birmingham UK
| | - H. Dollfus
- Centre de Référence pour les affections rares en génétique ophtalmologique; CARGO, Filière SENSGENE, Hôpitaux Universitaires de Strasbourg; Strasbourg France
| | - J. Kaplan
- INSERM U1163; Génétique Ophtalmologique; Paris France
| | - D. Lehalle
- Centre de Génétique et Centre de Référence "Anomalies du Développement et Syndromes Malformatifs; Hôpital d'Enfants; Dijon France
| | - C. Francannet
- Service de Génétique Médicale; CHU Estaing; Clermont-Ferrand France
| | - G. Morin
- Service de génétique; Hôpital nord d’Amiens; Amiens France
| | - H. Colineaux
- Department of Epidemiology, Health Economics and Public Health; Toulouse University Hospital; France
- LEASP UMR1027, INSERM; Université Toulouse III; Toulouse France
| | - P. Calvas
- Service de Génétique Médicale, Hôpital Purpan, CHU Toulouse; Toulouse France
- INSERM U1056; Université Toulouse III; Toulouse France
| | - N. Chassaing
- Service de Génétique Médicale, Hôpital Purpan, CHU Toulouse; Toulouse France
- INSERM U1056; Université Toulouse III; Toulouse France
| |
Collapse
|
38
|
David D, Anand D, Araújo C, Gloss B, Fino J, Dinger M, Lindahl P, Pöyhönen M, Hannele L, Lavinha J. Identification of OAF and PVRL1 as candidate genes for an ocular anomaly characterized by Peters anomaly type 2 and ectopia lentis. Exp Eye Res 2018; 168:161-170. [PMID: 29305299 DOI: 10.1016/j.exer.2017.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 01/10/2023]
Abstract
Keratolenticular dysgenesis (KLD) and ectopia lentis are congenital eye defects. The aim of this study is the identification of molecular genetic alterations responsible for those ocular anomalies with neurologic impairment in an individual with a de novo balanced chromosome translocation t(11;18)(q23.3;q11.2)dn. Disruption of OAF, the human orthologue of the Drosophila oaf, by the 11q23.3 breakpoint results in reduced expression of this transcriptional regulator. Furthermore, four most likely nonfunctional chimeric transcripts comprising up to OAF exon 3, derived from the der(11) allele, have also been identified. This locus has been implicated by publicly available genome-wide association data in corneal disease and corneal topography. The expression of the poliovirus receptor-related 1(PVRL1) or nectin cell adhesion molecule 1 (NECTIN1), a paralogue of nectin cell adhesion molecule 3 (PVRL3) associated with congenital ocular defects, situated 500 kb upstream from 11q23.3 breakpoint, is increased. The 18q11.2 breakpoint is localized between cutaneous T-cell lymphoma-associated antigen 1(CTAGE1) and retinoblastoma binding protein 8 (RBBP8) genes. Genomic imbalance that could contribute to the observed phenotype was excluded. Analysis of gene expression datasets throughout normal murine ocular lens embryogenesis suggests that OAF expression is significantly enriched in the lens from early stages of development through adulthood, whereas PVRL1 is lens-enriched until E12.5 and then down-regulated. This contrasts with the observation that the proposita's lymphoblastoid cell lines exhibit low OAF and high PVRL1 expression as compared to control, which offers further support that the alterations described above are most likely responsible for the clinical phenotype. Finally, gene interaction topology data for PVRL1 also agree with our proposal that disruption of OAF by the translocation breakpoint and misregulation of PVRL1 due to a position effect contribute to the observed ocular and neurological phenotype.
Collapse
Affiliation(s)
- Dezső David
- Department of Human Genetics, National Health Institute Doutor Ricardo Jorge, Lisbon, Portugal.
| | - Deepti Anand
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Carlos Araújo
- Department of Human Genetics, National Health Institute Doutor Ricardo Jorge, Lisbon, Portugal
| | - Brian Gloss
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Darlinghurst, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW, Kensington, Australia
| | - Joana Fino
- Department of Human Genetics, National Health Institute Doutor Ricardo Jorge, Lisbon, Portugal
| | - Marcel Dinger
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Darlinghurst, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW, Kensington, Australia
| | - Päivi Lindahl
- Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Minna Pöyhönen
- Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Laivuori Hannele
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - João Lavinha
- Department of Human Genetics, National Health Institute Doutor Ricardo Jorge, Lisbon, Portugal
| |
Collapse
|
39
|
Zhang W, Ogando DG, Kim ET, Choi MJ, Li H, Tenessen JM, Bonanno JA. Conditionally Immortal Slc4a11-/- Mouse Corneal Endothelial Cell Line Recapitulates Disrupted Glutaminolysis Seen in Slc4a11-/- Mouse Model. Invest Ophthalmol Vis Sci 2017; 58:3723-3731. [PMID: 28738416 PMCID: PMC5525555 DOI: 10.1167/iovs.17-21781] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose To establish conditionally immortal mouse corneal endothelial cell lines with genetically matched Slc4a11+/+ and Slc4a11-/- mice as a model for investigating pathology and therapies for SLC4A11 associated congenital hereditary endothelial dystrophy (CHED) and Fuchs' endothelial corneal dystrophy. Methods We intercrossed H-2Kb-tsA58 mice (Immortomouse) expressing an IFN-γ dependent and temperature-sensitive mutant of the SV40 large T antigen (tsTAg) with Slc4a11+/+ and Slc4a11-/- C57BL/6 mice. The growth characteristics of the cell lines was assessed by doubling time. Ion transport activities (Na+/H+ exchange, bicarbonate, lactate, and Slc4a11 ammonia transport) were analyzed by intracellular pH measurement. The metabolic status of the cell lines was assessed by analyzing TCA cycle intermediates via gas chromatography mass spectrometry (GC-MS). Results The immortalized Slc4a11+/+ and Slc4a11-/- mouse corneal endothelial cells (MCECs) remained proliferative through passage 49 and maintained similar active ion transport activity. As expected, proliferation was temperature sensitive and IFN-γ dependent. Slc4a11-/- MCECs exhibited decreased proliferative capacity, reduced NH3:H+ transport, altered expression of glutaminolysis enzymes similar to the Slc4a11-/- mouse, and reduced proportion of TCA cycle intermediates derived from glutamine with compensatory increases in glucose flux compared with Slc4a11+/+ MCECs. Conclusions This is the first report of the immortalization of MCECs. Ion transport of the immortalized endothelial cells remains active, except for NH3:H+ transporter activity in Slc4a11-/- MCECs. Furthermore, Slc4a11-/- MCECs recapitulate the glutaminolysis defects observed in Slc4a11-/- mouse corneal endothelium, providing an excellent tool to study the pathogenesis of SLC4A11 mutations associated with corneal endothelial dystrophies and to screen potential therapeutic agents.
Collapse
Affiliation(s)
- Wenlin Zhang
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Diego G Ogando
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Edward T Kim
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Moon-Jung Choi
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Hongde Li
- Department of Biology, Indiana University, Bloomington, Indiana, United States
| | - Jason M Tenessen
- Department of Biology, Indiana University, Bloomington, Indiana, United States
| | - Joseph A Bonanno
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| |
Collapse
|
40
|
Zhang W, Li H, Ogando DG, Li S, Feng M, Price FW, Tennessen JM, Bonanno JA. Glutaminolysis is Essential for Energy Production and Ion Transport in Human Corneal Endothelium. EBioMedicine 2017; 16:292-301. [PMID: 28117276 PMCID: PMC5474426 DOI: 10.1016/j.ebiom.2017.01.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 12/23/2022] Open
Abstract
Corneal endothelium (CE) is among the most metabolically active tissues in the body. This elevated metabolic rate helps the CE maintain corneal transparency by its ion and fluid transport properties, which when disrupted, leads to visual impairment. Here we demonstrate that glutamine catabolism (glutaminolysis) through TCA cycle generates a large fraction of the ATP needed to maintain CE function, and this glutaminolysis is severely disrupted in cells deficient in NH3:H+ cotransporter Solute Carrier Family 4 Member 11 (SLC4A11). Considering SLC4A11 mutations leads to corneal endothelial dystrophy and sensorineural deafness, our results indicate that SLC4A11-associated developmental and degenerative disorders result from altered glutamine catabolism. Overall, our results describe an important metabolic mechanism that provides CE cells with the energy required to maintain high level transport activity, reveal a direct link between glutamine metabolism and developmental and degenerative neuronal diseases, and suggest an approach for protecting the CE during ophthalmic surgeries. Glutamine contributes half of TCA cycle intermediates in human corneal endothelium. Glutamine catabolism supplies significant ATP that fuels the endothelial pump function. SLC4A11 (NH3:2H+ cotransporter) knockout shows ammonia related oxidative damage. Loss of SLC4A11 transporter disrupts expression of glutaminolysis enzymes.
The corneal endothelium (CE) is responsible for maintaining corneal transparency through the action of active transport processes. We report that CE metabolizes the amino acid glutamine producing ATP in support of active transport. In the mouse model of CHED (Congenital Hereditary Endothelial Dystrophy), which manifests corneal edema and loss of transparency, glutamine metabolism is disrupted due to loss of SLC4A11, an NH3:2H+ transporter. This work sheds light on potential clinical therapies to facilitate CE function, the pathogenesis of CHED and Fuchs' Endothelial Corneal Dystrophy, and suggests that the ammonia handling capacity of SLC4A11 is essential for efficient metabolism of glutamine.
Collapse
Affiliation(s)
- Wenlin Zhang
- School of Optometry, Indiana University, Bloomington, IN 47405, USA.
| | - Hongde Li
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Diego G Ogando
- School of Optometry, Indiana University, Bloomington, IN 47405, USA
| | - Shimin Li
- School of Optometry, Indiana University, Bloomington, IN 47405, USA
| | | | | | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Joseph A Bonanno
- School of Optometry, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
41
|
Correia JD, da Rosa EB, Silveira DB, Correia EPE, Lorenzen MB, Travi GM, Rosa RCM, Zen PRG, Zen TD, Rosa RFM. Trisomy 18 and eye anomalies. Am J Med Genet A 2016; 173:553-555. [PMID: 27792864 DOI: 10.1002/ajmg.a.38036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/07/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Jamile D Correia
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Ernani B da Rosa
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Daniélle B Silveira
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | | | | | - Giovanni M Travi
- Pediatric Ophthalmology, Hospital da Criança Santo Antônio (HCSA)/Complexo Hospitalar Santa Casa de Porto Alegre (CHSCPA), Porto Alegre, RS, Brazil
| | - Rosana C M Rosa
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Paulo R G Zen
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.,Graduate Program in Biosciences, UFCSPA, Porto Alegre, RS, Brazil.,Clinical Genetics, UFCSPA and CHSCPA, Porto Alegre, RS, Brazil
| | - Tatiana D Zen
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.,Pharmacy, Faculdade de Ciências da Saúde, Centro Universitário Ritter dos Reis-UniRitter, Porto Alegre, RS, Brazil
| | - Rafael F M Rosa
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.,Graduate Program in Biosciences, UFCSPA, Porto Alegre, RS, Brazil.,Clinical Genetics, UFCSPA and CHSCPA, Porto Alegre, RS, Brazil
| |
Collapse
|
42
|
Impaired ADAMTS9 secretion: A potential mechanism for eye defects in Peters Plus Syndrome. Sci Rep 2016; 6:33974. [PMID: 27687499 PMCID: PMC5043182 DOI: 10.1038/srep33974] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/06/2016] [Indexed: 01/15/2023] Open
Abstract
Peters Plus syndrome (PPS), a congenital disorder of glycosylation, results from recessive mutations affecting the glucosyltransferase B3GLCT, leading to congenital corneal opacity and diverse extra-ocular manifestations. Together with the fucosyltransferase POFUT2, B3GLCT adds Glucoseβ1-3Fucose disaccharide to a consensus sequence in thrombospondin type 1 repeats (TSRs) of several proteins. Which of these target proteins is functionally compromised in PPS is unknown. We report here that haploinsufficiency of murine Adamts9, encoding a secreted metalloproteinase with 15 TSRs, leads to congenital corneal opacity and Peters anomaly (persistent lens-cornea adhesion), which is a hallmark of PPS. Mass spectrometry of recombinant ADAMTS9 showed that 9 of 12 TSRs with the O-fucosylation consensus sequence carried the Glucoseβ1-3Fucose disaccharide and B3GLCT knockdown reduced ADAMTS9 secretion in HEK293F cells. Together, the genetic and biochemical findings imply a dosage-dependent role for ADAMTS9 in ocular morphogenesis. Reduced secretion of ADAMTS9 in the absence of B3GLCT is proposed as a mechanism of Peters anomaly in PPS. The functional link between ADAMTS9 and B3GLCT established here also provides credence to their recently reported association with age-related macular degeneration.
Collapse
|
43
|
Kao L, Azimov R, Shao XM, Frausto RF, Abuladze N, Newman D, Aldave AJ, Kurtz I. Multifunctional ion transport properties of human SLC4A11: comparison of the SLC4A11-B and SLC4A11-C variants. Am J Physiol Cell Physiol 2016; 311:C820-C830. [PMID: 27581649 DOI: 10.1152/ajpcell.00233.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
Abstract
Congenital hereditary endothelial dystrophy (CHED), Harboyan syndrome (CHED with progressive sensorineural deafness), and potentially a subset of individuals with late-onset Fuchs' endothelial corneal dystrophy are caused by mutations in the SLC4A11 gene that results in corneal endothelial cell abnormalities. Originally classified as a borate transporter, the function of SLC4A11 as a transport protein remains poorly understood. Elucidating the transport function(s) of SLC4A11 is needed to better understand how its loss results in the aforementioned posterior corneal dystrophic disease processes. Quantitative PCR experiments demonstrated that, of the three known human NH2-terminal variants, SLC4A11-C is the major transcript expressed in human corneal endothelium. We studied the expression pattern of the three variants in mammalian HEK-293 cells and demonstrated that the SLC4A11-B and SLC4A11-C variants are plasma membrane proteins, whereas SLC4A11-A is localized intracellularly. SLC4A11-B and SLC4A11-C were shown to be multifunctional ion transporters capable of transporting H+ equivalents in both a Na+-independent and Na+-coupled mode. In both transport modes, SLC4A11-C H+ flux was significantly greater than SLC4A11-B. In the presence of ammonia, SLC4A11-B and SLC4A11-C generated inward currents that were comparable in magnitude. Chimera SLC4A11-C-NH2-terminus-SLC4A11-B experiments demonstrated that the SLC4A11-C NH2-terminus functions as an autoactivating domain, enhancing Na+-independent and Na+-coupled H+ flux without significantly affecting the electrogenic NH3-H(n)+ cotransport mode. All three modes of transport were significantly impaired in the presence of the CHED causing p.R109H (SLC4A11-C numbering) mutation. These complex ion transport properties need to be addressed in the context of corneal endothelial disease processes caused by mutations in SLC4A11.
Collapse
Affiliation(s)
- Liyo Kao
- Division of Nephrology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Rustam Azimov
- Division of Nephrology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Xuesi M Shao
- Department of Neurobiology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ricardo F Frausto
- Stein Eye Institute, and.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Natalia Abuladze
- Division of Nephrology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Debra Newman
- Division of Nephrology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Anthony J Aldave
- Stein Eye Institute, and.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ira Kurtz
- Division of Nephrology, .,Brain Research Institute.,David Geffen School of Medicine, University of California, Los Angeles, California
| |
Collapse
|
44
|
Happ H, Schilter KF, Weh E, Reis LM, Semina EV. 8q21.11 microdeletion in two patients with syndromic peters anomaly. Am J Med Genet A 2016; 170:2471-5. [PMID: 27378168 DOI: 10.1002/ajmg.a.37840] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/26/2016] [Indexed: 12/18/2022]
Abstract
Peters anomaly is a form of anterior segment dysgenesis characterized by central ocular opacity and corneo-lenticular adhesions. Isolated and syndromic Peters anomaly can be observed and demonstrate significant genetic heterogeneity. We report the identification of overlapping 8q21.11 deletions in two patients with syndromic Peters anomaly via whole exome sequencing and chromosomal microarray analyses. Microdeletions of 8q21.11 were recently reported in 10 patients with highly variable phenotypes involving craniofacial features, ptosis, intellectual disability, abnormalities of the hands/feet and other defects; sclerocornea and/or microphthalmia were reported in three cases. The two additional cases presented in this report expand the phenotypic spectrum of 8q21.11 microdeletions to include Peters anomaly (seen in both patients) and persistent primary dentition (seen in one patient with a larger deletion). The two novel deletions include the ZFHX4 and PEX2 genes, which were also affected in all three previous cases involving ocular anomalies. Screening of the remaining alleles of ZFHX4 and PEX2 did not identify any additional likely pathogenic variants in either patient, suggesting a dominant mechanism (haploinsufficiency) for the identified deletion. This report provides further insight into the phenotypes associated with 8q21.11 deletions and, for the first time, reports Peters anomaly as an additional ocular feature; screening for copy number variations of the 8q21.11 region should be considered in patients with Peters anomaly and related syndromic features. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hannah Happ
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kala F Schilter
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Eric Weh
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Linda M Reis
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Elena V Semina
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Ophthalmology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
45
|
Srilekha S, Rao B, Rao DM, Sudha D, Chandrasekar SP, Pandian AJ, Soumittra N, Sripriya S. Strategies for Gene Mapping in Inherited Ophthalmic Diseases. Asia Pac J Ophthalmol (Phila) 2016; 5:282-92. [PMID: 27488070 DOI: 10.1097/apo.0000000000000228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Gene mapping of inherited ophthalmic diseases such as congenital cataracts, retinal degeneration, glaucoma, age-related macular degeneration, myopia, optic atrophy, and eye malformations has shed more light on the disease pathology, identified targets for research on therapeutics, earlier detection, and treatment options for disease management and patient care. This article details the different approaches to gene identification for both Mendelian and complex eye disorders.
Collapse
Affiliation(s)
- Sundar Srilekha
- From the SNONGC Department of Genetics and Molecular Biology, Kamal Nayan Bajaj Institute for Research in Vision and Ophthalmology (KNBIRVO), Chennai, Tamil Nadu, India
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Moutton S, Fergelot P, Naudion S, Cordier MP, Solé G, Guerineau E, Hubert C, Rooryck C, Vuillaume ML, Houcinat N, Deforges J, Bouron J, Devès S, Le Merrer M, David A, Geneviève D, Giuliano F, Journel H, Megarbane A, Faivre L, Chassaing N, Francannet C, Sarrazin E, Stattin EL, Vigneron J, Leclair D, Abadie C, Sarda P, Baumann C, Delrue MA, Arveiler B, Lacombe D, Goizet C, Coupry I. Otopalatodigital spectrum disorders: refinement of the phenotypic and mutational spectrum. J Hum Genet 2016; 61:693-9. [PMID: 27193221 DOI: 10.1038/jhg.2016.37] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/15/2016] [Accepted: 03/15/2016] [Indexed: 11/09/2022]
Abstract
Otopalatodigital spectrum disorders (OPDSD) constitute a group of dominant X-linked osteochondrodysplasias including four syndromes: otopalatodigital syndromes type 1 and type 2 (OPD1 and OPD2), frontometaphyseal dysplasia, and Melnick-Needles syndrome. These syndromes variably associate specific facial and extremities features, hearing loss, cleft palate, skeletal dysplasia and several malformations, and show important clinical overlap over the different entities. FLNA gain-of-function mutations were identified in these conditions. FLNA encodes filamin A, a scaffolding actin-binding protein. Here, we report phenotypic descriptions and molecular results of FLNA analysis in a large series of 27 probands hypothesized to be affected by OPDSD. We identified 11 different missense mutations in 15 unrelated probands (n=15/27, 56%), of which seven were novel, including one of unknown significance. Segregation analyses within families made possible investigating 20 additional relatives carrying a mutation. This series allows refining the phenotypic and mutational spectrum of FLNA mutations causing OPDSD, and providing suggestions to avoid the overdiagnosis of OPD1.
Collapse
Affiliation(s)
- Sébastien Moutton
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France.,Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France
| | - Patricia Fergelot
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France.,Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France.,Plateforme Génome Transcriptome, Centre de Génomique Fonctionnelle de Bordeaux, Université de Bordeaux, Bordeaux, France
| | - Sophie Naudion
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France
| | - Marie-Pierre Cordier
- CHU Lyon, Hôpital Femme-Mère-Enfant, Department of Medical Genetics, Bron cedex, France
| | - Guilhem Solé
- Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France.,CHU Bordeaux, Hôpital Pellegrin, Department of Neurology, Fédération des Neurosciences Cliniques, Bordeaux, France
| | - Elodie Guerineau
- Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France
| | - Christophe Hubert
- Plateforme Génome Transcriptome, Centre de Génomique Fonctionnelle de Bordeaux, Université de Bordeaux, Bordeaux, France
| | - Caroline Rooryck
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France.,Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France
| | - Marie-Laure Vuillaume
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France.,Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France
| | - Nada Houcinat
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France.,Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France
| | - Julie Deforges
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France
| | - Julie Bouron
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France
| | - Sylvie Devès
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France
| | - Martine Le Merrer
- Institut Imagine, Hôpital Necker Enfants Malades, Department of Medical Genetics, INSERM U781, Université Paris Descartes-Sorbonne Paris Cité, Paris cedex, France
| | - Albert David
- CHU Nantes, Hôpital Mère-Enfant, Department of Medical Genetics, Nantes cedex, France
| | - David Geneviève
- CHRU Montpellier, Hôpital Arnaud de Villeneuve, Department of Medical Genetics, Université Montpellier INSERM U1183, CLAD Sud Languedoc-Roussillon, Montpellier cedex, France
| | - Fabienne Giuliano
- CHU Nice, Hôpital l'Archet 2, Department of Medical Genetics, Nice cedex, France
| | - Hubert Journel
- Centre Hospitalier Bretagne Atlantique, Department of Medical Genetics and Oncogenetics, Vannes cedex, France
| | - André Megarbane
- Al-Jawhara Center, Department of Medical Genetics, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Laurence Faivre
- CHU Dijon, Department of Medical Genetics, Centre de Référence Anomalies de Développement et Syndromes Malformatifs de l'inter-région Grand-Est, Hôpital d'Enfants, Dijon, France
| | - Nicolas Chassaing
- CHU Toulouse, Hôpital Purpan, Department of Medical Genetics, UDEAR, Université de Toulouse, Inserm, UPS, CNRS, Toulouse cedex, France
| | - Christine Francannet
- CHU Clermont-Ferrand, Hôpital d'Estaing, Department of Medical Genetics, Clermont-Ferrand cedex, France
| | - Elisabeth Sarrazin
- CHU de Fort de France, Hôpital Pierre Zobda-Quitman, Department of Neuropediatrics, Centre de Référence Caribéen des Maladies Rares Neurologiques et Neuromusculaires, Martinique, France
| | - Eva-Lena Stattin
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jacqueline Vigneron
- CHU Nancy, Maternité Régionale Adolphe Pinard, Department of Medical Genetics, Nancy cedex, France
| | - Danielle Leclair
- CHU Raymond Poincaré, Department of Physical Medicine and Rehabilitation, Centre de Référence Maladies Neuromusculaires, Garches, France
| | - Caroline Abadie
- CHRU Montpellier, Hôpital Arnaud de Villeneuve, Department of Medical Genetics, Université Montpellier INSERM U1183, CLAD Sud Languedoc-Roussillon, Montpellier cedex, France
| | - Pierre Sarda
- CHRU Montpellier, Hôpital Arnaud de Villeneuve, Department of Medical Genetics, Université Montpellier INSERM U1183, CLAD Sud Languedoc-Roussillon, Montpellier cedex, France
| | - Clarisse Baumann
- AP-HP, Hôpital Robert Debré, Department of Medical Genetics, CLAD Ile de France, Paris, France
| | - Marie-Ange Delrue
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France
| | - Benoit Arveiler
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France.,Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France
| | - Didier Lacombe
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France.,Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France
| | - Cyril Goizet
- CHU Bordeaux, Hôpital Pellegrin, Department of Medical Genetics, Centre de Référence des Anomalies du Développement Embryonnaire, Bordeaux cedex, France.,Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France
| | - Isabelle Coupry
- Université de Bordeaux, INSERM U1211, Laboratoire Maladies Rares: Génétique et Métabolisme, Bordeaux, France
| |
Collapse
|
47
|
Chassaing N, Davis EE, McKnight KL, Niederriter AR, Causse A, David V, Desmaison A, Lamarre S, Vincent-Delorme C, Pasquier L, Coubes C, Lacombe D, Rossi M, Dufier JL, Dollfus H, Kaplan J, Katsanis N, Etchevers HC, Faguer S, Calvas P. Targeted resequencing identifies PTCH1 as a major contributor to ocular developmental anomalies and extends the SOX2 regulatory network. Genome Res 2016; 26:474-85. [PMID: 26893459 PMCID: PMC4817771 DOI: 10.1101/gr.196048.115] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 02/04/2016] [Indexed: 12/17/2022]
Abstract
Ocular developmental anomalies (ODA) such as anophthalmia/microphthalmia (AM) or anterior segment dysgenesis (ASD) have an estimated combined prevalence of 3.7 in 10,000 births. Mutations in SOX2 are the most frequent contributors to severe ODA, yet account for a minority of the genetic drivers. To identify novel ODA loci, we conducted targeted high-throughput sequencing of 407 candidate genes in an initial cohort of 22 sporadic ODA patients. Patched 1 (PTCH1), an inhibitor of sonic hedgehog (SHH) signaling, harbored an enrichment of rare heterozygous variants in comparison to either controls, or to the other candidate genes (four missense and one frameshift); targeted resequencing of PTCH1 in a second cohort of 48 ODA patients identified two additional rare nonsynonymous changes. Using multiple transient models and a CRISPR/Cas9-generated mutant, we show physiologically relevant phenotypes altering SHH signaling and eye development upon abrogation of ptch1 in zebrafish for which in vivo complementation assays using these models showed that all six patient missense mutations affect SHH signaling. Finally, through transcriptomic and ChIP analyses, we show that SOX2 binds to an intronic domain of the PTCH1 locus to regulate PTCH1 expression, findings that were validated both in vitro and in vivo. Together, these results demonstrate that PTCH1 mutations contribute to as much as 10% of ODA, identify the SHH signaling pathway as a novel effector of SOX2 activity during human ocular development, and indicate that ODA is likely the result of overactive SHH signaling in humans harboring mutations in either PTCH1 or SOX2.
Collapse
Affiliation(s)
- Nicolas Chassaing
- CHU Toulouse, Service de Génétique Médicale, Hôpital Purpan, 31059 Toulouse, France; Université Paul-Sabatier Toulouse III, EA-4555, 31000 Toulouse, France; Inserm U1056, 31000 Toulouse, France
| | - Erica E Davis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina 27701, USA; Department of Pediatrics and Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27701, USA
| | - Kelly L McKnight
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina 27701, USA
| | - Adrienne R Niederriter
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina 27701, USA
| | - Alexandre Causse
- Université Paul-Sabatier Toulouse III, EA-4555, 31000 Toulouse, France; CHU Toulouse, Service d'Ophtalmologie, Hôpital Purpan, 31059 Toulouse, France
| | - Véronique David
- Institut de Génétique et Développement, CNRS UMR6290, Université de Rennes 1, IFR140 GFAS, Faculté de Médecine, 35043 Rennes, France; Laboratoire de Génétique Moléculaire, CHU Pontchaillou, 35043 Rennes Cedex, France
| | - Annaïck Desmaison
- Université Paul-Sabatier Toulouse III, EA-4555, 31000 Toulouse, France
| | - Sophie Lamarre
- Université de Toulouse; INSA, UPS, INP, LISBP, F-31077 Toulouse, France; INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, F-31400 Toulouse, France; CNRS, UMR5504, F-31400 Toulouse, France; Plateforme Biopuces de la Génopole de Toulouse Midi Pyrénées, INSA/DGBA 135, 31077 Toulouse, France
| | | | - Laurent Pasquier
- Service de Génétique Clinique, Hôpital Sud, 35200 Rennes, France
| | - Christine Coubes
- Service de Génétique Médicale, Hôpital Arnaud de Villeneuve, 34295 Montpellier, France
| | - Didier Lacombe
- Service de Génétique Médicale, Hôpital Pellegrin, 33076 Bordeaux Cedex, France; Université Bordeaux Segalen, Laboratoire MRGM, 33076 Bordeaux, France
| | - Massimiliano Rossi
- Service de Génétique, Hospices Civils de Lyon, Groupement Hospitalier Est, 69677 Bron, France; INSERM U1028 UMR CNRS 5292, UCBL, CRNL TIGER Team, 69677 Bron Cedex, France
| | - Jean-Louis Dufier
- Service d'Ophtalmologie, Hôpital Necker Enfants Malades, 75015 Paris, France
| | - Helene Dollfus
- Service de Génétique Médicale, Hôpitaux Universitaires de Strasbourg, 67091 Strasbourg, France
| | - Josseline Kaplan
- INSERM U781 & Department of Genetics, Paris Descartes University, 75015 Paris, France
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina 27701, USA; Department of Pediatrics and Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27701, USA
| | - Heather C Etchevers
- Université Paul-Sabatier Toulouse III, EA-4555, 31000 Toulouse, France; INSERM, UMR_S910, Aix-Marseille University, Faculté de Médecine, 13385 Marseille, France
| | | | - Patrick Calvas
- CHU Toulouse, Service de Génétique Médicale, Hôpital Purpan, 31059 Toulouse, France; Université Paul-Sabatier Toulouse III, EA-4555, 31000 Toulouse, France; Inserm U1056, 31000 Toulouse, France
| |
Collapse
|
48
|
Filges I, Bruder E, Brandal K, Meier S, Undlien DE, Waage TR, Hoesli I, Schubach M, de Beer T, Sheng Y, Hoeller S, Schulzke S, Røsby O, Miny P, Tercanli S, Oppedal T, Meyer P, Selmer KK, Strømme P. Strømme Syndrome Is a Ciliary Disorder Caused by Mutations in CENPF. Hum Mutat 2016; 37:359-63. [PMID: 26820108 DOI: 10.1002/humu.22960] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/08/2016] [Indexed: 11/10/2022]
Abstract
Strømme syndrome was first described by Strømme et al. (1993) in siblings presenting with "apple peel" type intestinal atresia, ocular anomalies and microcephaly. The etiology remains unknown to date. We describe the long-term clinical follow-up data for the original pair of siblings as well as two previously unreported siblings with a severe phenotype overlapping that of the Strømme syndrome including fetal autopsy results. Using family-based whole-exome sequencing, we identified truncating mutations in the centrosome gene CENPF in the two nonconsanguineous Caucasian sibling pairs. Compound heterozygous inheritance was confirmed in both families. Recently, mutations in this gene were shown to cause a fetal lethal phenotype, the phenotype and functional data being compatible with a human ciliopathy [Waters et al., 2015]. We show for the first time that Strømme syndrome is an autosomal-recessive disease caused by mutations in CENPF that can result in a wide phenotypic spectrum.
Collapse
Affiliation(s)
- Isabel Filges
- Medical Genetics, University Hospital Basel, Basel, Switzerland
| | | | - Kristin Brandal
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Stephanie Meier
- Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Dag Erik Undlien
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Trine Rygvold Waage
- Section of Paediatric Neurohabilitation, Department of Clinical Neurosciences for Children, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Irene Hoesli
- Obstetrics and Gynecology, University Hospital Basel, Basel, Switzerland
| | - Max Schubach
- Institute for Medical and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tjaart de Beer
- Biozentrum and Swiss Institute of Bioinformatics, University of Basel, Basel, Switzerland
| | - Ying Sheng
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Sylvia Hoeller
- Pathology, University Hospital Basel, Basel, Switzerland
| | - Sven Schulzke
- Neonatology, University Children's Hospital Basel, Basel, Switzerland
| | - Oddveig Røsby
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Peter Miny
- Medical Genetics, University Hospital Basel, Basel, Switzerland
| | | | - Truls Oppedal
- Department of Ophthalmology, Section for Pediatric Ophthalmology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Peter Meyer
- Pathology, University Hospital Basel, Basel, Switzerland
| | - Kaja Kristine Selmer
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Petter Strømme
- Section for Clinical Neurosciences, Department of Pediatrics, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
49
|
Naudion S, Moutton S, Coupry I, Sole G, Deforges J, Guerineau E, Hubert C, Deves S, Pilliod J, Rooryck C, Abel C, Le Breton F, Collardeau-Frachon S, Cordier M, Delezoide A, Goldenberg A, Loget P, Melki J, Odent S, Patrier S, Verloes A, Viot G, Blesson S, Bessières B, Lacombe D, Arveiler B, Goizet C, Fergelot P. Fetal phenotypes in otopalatodigital spectrum disorders. Clin Genet 2015; 89:371-7. [DOI: 10.1111/cge.12679] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/12/2015] [Accepted: 09/21/2015] [Indexed: 11/27/2022]
Affiliation(s)
- S. Naudion
- CHU Bordeaux, Centre de Référence des Anomalies du Développement Embryonnaire; Service de Génétique Médicale; Bordeaux France
| | - S. Moutton
- CHU Bordeaux, Centre de Référence des Anomalies du Développement Embryonnaire; Service de Génétique Médicale; Bordeaux France
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
| | - I. Coupry
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
| | - G. Sole
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
- CHU Bordeaux; Fédération des Neurosciences Cliniques; Bordeaux France
| | - J. Deforges
- CHU Bordeaux, Centre de Référence des Anomalies du Développement Embryonnaire; Service de Génétique Médicale; Bordeaux France
| | - E. Guerineau
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
| | - C. Hubert
- Plateforme Génome Transcriptome; Centre de Génomique Fonctionnelle de Bordeaux, Université de Bordeaux; Bordeaux France
| | - S. Deves
- CHU Bordeaux, Centre de Référence des Anomalies du Développement Embryonnaire; Service de Génétique Médicale; Bordeaux France
| | - J. Pilliod
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
| | - C. Rooryck
- CHU Bordeaux, Centre de Référence des Anomalies du Développement Embryonnaire; Service de Génétique Médicale; Bordeaux France
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
| | - C. Abel
- CHU Lyon, Service de Génétique; Hôpital Femme Mère Enfant, Hospices Civils de Lyon; Lyon France
| | - F. Le Breton
- CHU Lyon, Service de Pathologie du Nord; Hôpital de la Croix-Rousse; Lyon France
| | | | - M.P. Cordier
- CHU Lyon, Service de Génétique Médicale; Hôpital Mère Enfant; Lyon France
| | - A.L. Delezoide
- APHP, Service de Biologie du Développement; Hôpital Robert Debré; Paris France
| | - A. Goldenberg
- CHU Rouen; Service de Génétique Médicale; Rouen France
| | - P. Loget
- CHU Rennes; Service d'Anatomie Cytologie Pathologique; Rennes France
| | - J. Melki
- INSERM U78, Laboratoire de Neurogénétique Moléculaire; Université de Paris XI; Paris France
| | - S. Odent
- CHU de Rennes, Service de Génétique Clinique; Centre de Référence Anomalies du Développement CLAD-Ouest, Hôpital Sud; Rennes France
| | - S. Patrier
- CHU Rouen; Service d'Anatomie Pathologique; Rouen France
| | - A. Verloes
- Département de Génétique, APHP-Hôpital universitaire Robert Debré; Université Sorbonne Paris-Cité, Faculté de Médecine Denis Diderot-Paris 7, and INSERM UMR 1141; Paris France
| | - G. Viot
- APHP, Service de Génétique Médicale; Maternité Port-Royal; Paris France
| | - S. Blesson
- CHRU Tours, Service de Génétique; Hôpital Bretonneau; Tours France
| | - B. Bessières
- APHP, Service Histo-Embryologie et Cytogénétique; Hôpital Necker; Paris France
| | - D. Lacombe
- CHU Bordeaux, Centre de Référence des Anomalies du Développement Embryonnaire; Service de Génétique Médicale; Bordeaux France
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
| | - B. Arveiler
- CHU Bordeaux, Centre de Référence des Anomalies du Développement Embryonnaire; Service de Génétique Médicale; Bordeaux France
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
| | - C. Goizet
- CHU Bordeaux, Centre de Référence des Anomalies du Développement Embryonnaire; Service de Génétique Médicale; Bordeaux France
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
| | - P. Fergelot
- CHU Bordeaux, Centre de Référence des Anomalies du Développement Embryonnaire; Service de Génétique Médicale; Bordeaux France
- University Bordeaux, Laboratoire Maladies Rares; Génétique et Métabolisme (MRGM); Bordeaux France
- Plateforme Génome Transcriptome; Centre de Génomique Fonctionnelle de Bordeaux, Université de Bordeaux; Bordeaux France
| |
Collapse
|
50
|
|