1
|
Yap YT, Pan J, Xu J, Yuan S, Niu C, Zheng C, Li W, Zhou T, Li T, Zhang Y, Holtzman MJ, Pazour GJ, Hess RA, Kelly CV, Touré A, Brody SL, Zhang Z. Role of intraflagellar transport protein IFT140 in the formation and function of motile cilia in mammals. Cell Mol Life Sci 2025; 82:198. [PMID: 40348912 PMCID: PMC12065702 DOI: 10.1007/s00018-025-05710-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 04/05/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Cilia are microtubular structures extending from the surface of most mammalian cells. They can be categorized as motile cilia and primary sensory cilia. Both types possess intraflagellar transport (IFT) machinery, composed of unique protein complexes that travel along the microtubules to deliver proteins for ciliary and flagellar assembly, disassembly, and homeostasis. Although the role of IFT in primary cilia formation has been well studied, little is known about its role in mammalian motile cilia assembly. We generated conditional knockout mice by breeding floxed Ift140 mice with the FOXJ1-Cre transgenic mouse line to specifically delete Ift140 from cells that assemble motile cilia. Mice with Ift140 deficiency did not have laterality defects or gross; however most died prior to sexual maturity. Those mutants that survived to adulthood were completely infertile. Males demonstrated abnormal spermatogenesis associated with reduced sperm count and motility, together with short length flagella, and abnormal morphology. Cilia length was diminished in the epithelial cells of the efferent ductules and airways. Cilia from cultured tracheal epithelial cells were also short and had reduced beat frequency (CBF). Ultrastructural studies revealed the presence of inner and outer dynein arms, but an abnormal central apparatus, and the accumulation of particles within the cilia. Overall, the short length and abnormal localization of ciliary proteins in Ift140 conditional mutants resulted in inadequate cilia function despite proper localization of the dynein motor complexes. We propose a key role of Ift140 for motile cilia assembly in certain tissues and suggest that genetic alterations of IFT140 could be associated with motile ciliopathies.
Collapse
Affiliation(s)
- Yi Tian Yap
- Department of Physiology, Wayne State University, 275 E Hancock Street, Detroit, MI, 48201, USA
| | - Jiehong Pan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jian Xu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Shuo Yuan
- Department of Physiology, Wayne State University, 275 E Hancock Street, Detroit, MI, 48201, USA
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Changmin Niu
- Department of Physiology, Wayne State University, 275 E Hancock Street, Detroit, MI, 48201, USA
- School of Nursing, School of Public Health, Yangzhou University, Yangzhou, Jiangsu, China
| | - Cheng Zheng
- Department of Physiology, Wayne State University, 275 E Hancock Street, Detroit, MI, 48201, USA
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Wei Li
- Department of Physiology, Wayne State University, 275 E Hancock Street, Detroit, MI, 48201, USA
| | - Ting Zhou
- Department of Physiology, Wayne State University, 275 E Hancock Street, Detroit, MI, 48201, USA
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Tao Li
- Department of Physiology, Wayne State University, 275 E Hancock Street, Detroit, MI, 48201, USA
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yong Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael J Holtzman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, 2001S. Lincoln, Urbana, IL, USA
| | - Christopher V Kelly
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA
| | - Aminata Touré
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institut pour l'Avancée des Biosciences (IAB), Team Physiology and Pathophysiology of Sperm cells, 38000, Grenoble, France
| | - Steven L Brody
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, 275 E Hancock Street, Detroit, MI, 48201, USA.
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
2
|
Zagorec N, Calamel A, Delaporte M, Olinger E, Orr S, Sayer JA, Pillay VG, Denommé-Pichon AS, Tran Mau-Them F, Nambot S, Faivre L, Ars E, Torra R, Ong ACM, Devuyst O, Perico N, Després AM, Lemoine H, de Fallois J, Brousse R, Hummel A, Knebelmann B, Maisonneuve N, Halbritter J, Le Meur Y, Audrézet MP, Cornec-Le Gall E. Clinical Spectrum and Prognosis of Atypical Autosomal Dominant Polycystic Kidney Disease Caused by Monoallelic Pathogenic Variants of IFT140. Am J Kidney Dis 2025; 85:465-476.e1. [PMID: 39732359 DOI: 10.1053/j.ajkd.2024.10.009] [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: 04/08/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 12/30/2024]
Abstract
RATIONALE & OBJECTIVE Monoallelic predicted loss-of-function (pLoF) variants in IFT140 have recently been associated with an autosomal dominant polycystic kidney disease (ADPKD)-like phenotype. This study enhanced the characterization of this phenotype. STUDY DESIGN Case series. SETTING & PARTICIPANTS Seventy-five among 2,797 European individuals with ADPKD-like phenotypes who underwent genetic testing that revealed pLoF IFT140-variants. FINDINGS The 75 individuals (median age 56 years, 53.3% females) were from 61 families and were found to have 41 different monoallelic pLoF IFT140-variants. The majority of individuals presented with large, exophytic kidney cysts (median total kidney volume, 688mL [IQR, 201-4,139]), and 90.2% were classified using the Mayo Imaging Classification as Mayo Class 2A. Arterial hypertension was present in 50.7% of the individuals (median age at diagnosis, 59 years [IQR, 29-73]). Only 1 patient developed kidney failure (at age 69 years). A significant difference was observed in the age-adjusted estimated glomerular filtration rate (eGFR) between the male and female patients (P<0.001), and 56.3% of the individuals over the age of 60 years had an eGFR of<60mL/min/1.73m2. The estimated genetic prevalence of monoallelic pLoF IFT140 variants was 19.76 (95% CI, 18.8-20.7) and 27.89 (95% CI, 23.8-31.9) per 10,000 in the Genome Aggregation Database and the 100,000 Genomes Project (100kG), respectively. Only cystic kidney disease (ICD-10 Q61) was associated with pLoF IFT140 variants (P = 2.9 × 10-9, odds ratio = 5.6 (95% CI, 3.3-9.2) in 100kG. STUDY LIMITATIONS Retrospective study; IFT140-related cystic kidney disease may not be diagnosed in younger patients or patients with milder forms. CONCLUSIONS Individuals with monoallelic IFT140 pLoF variants are likely to develop kidney cysts atypical of classic ADPKD and generally have a favorable kidney prognosis. PLAIN-LANGUAGE SUMMARY Monoallelic pathogenic variants in IFT140 have been linked to a spectrum of kidney disease clinically similar to autosomal dominant polycystic kidney disease (ADPKD). This article describes a case series of 75 individuals with ADPKD-IFT140. Affected individuals typically presented with an atypical imaging pattern, had fewer but larger kidney cysts compared with classic ADPKD, and rarely developed liver cysts. Although the kidney prognosis appeared better than in classic ADPKD, 56.3% of individuals over 60 years of age had stage 3 or more severe CKD. Individuals with ADPKD-IFT140 variants are likely to develop kidney cyst patterns atypical of ADPKD. Their kidney prognosis appears favorable.
Collapse
Affiliation(s)
- Nikola Zagorec
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de Référence MARHEA, Brest; Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia; Department of Nephrology and Dialysis, Dubrava University Hospital, Zagreb, Croatia
| | - Alizée Calamel
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de Référence MARHEA, Brest
| | - Margaux Delaporte
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de Référence MARHEA, Brest
| | - Eric Olinger
- Center for Human Genetics, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Sarah Orr
- Biosciences Institute, Newcastle Upon Tyne
| | - John A Sayer
- Biosciences Institute, Newcastle Upon Tyne; NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle Upon Tyne; the Newcastle upon Tyne Hospitals NHS Foundation Trust, Renal Services, Newcastle Upon Tyne
| | - Vignesh-Guru Pillay
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, CHU Dijon; INSERM UMR1231 GAD, Genetics of Developmental Disorders, Université de Bourgogne-Franche-Comté
| | - Anne-Sophie Denommé-Pichon
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, CHU Dijon; INSERM UMR1231 GAD, Genetics of Developmental Disorders, Université de Bourgogne-Franche-Comté
| | - Frederic Tran Mau-Them
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, CHU Dijon; INSERM UMR1231 GAD, Genetics of Developmental Disorders, Université de Bourgogne-Franche-Comté
| | - Sophie Nambot
- INSERM UMR1231 GAD, Genetics of Developmental Disorders, Université de Bourgogne-Franche-Comté; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est, Centre Hospitalier Universitaire Dijon, Dijon
| | - Laurence Faivre
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), CHU Dijon; INSERM UMR1231 GAD, Genetics of Developmental Disorders, Université de Bourgogne-Franche-Comté; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est, Centre Hospitalier Universitaire Dijon, Dijon
| | - Elisabet Ars
- Molecular Biology Laboratory, Fundació Puigvert, Instituto de Recerca Sant Pau (R Sant Pau), Medicine Department, RICORS2040, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Roser Torra
- Department of Nephrology, Fundació Puigvert, Instituto de Recerca Sant Pau (R Sant Pau), Medicine Department, RICORS2040, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Albert C M Ong
- Kidney Genetics Group, Academic Nephrology, Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom; Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Olivier Devuyst
- Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium; Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Noberto Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | | | - Hugo Lemoine
- University of Brest, Inserm, UMR 1078, Génétique, Génomique Fonctionelle et Biotechnologies, Brest
| | - Jonathan de Fallois
- Division of Nephrology, Department of Internal Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Romain Brousse
- Sorbonne Université, INSERM UMRS 1155, Nephrology Department, Assistance Publique Hopitaux De Paris, Hopital Tenon, Paris
| | - Aurélie Hummel
- Department of Nephrology, Necker Hospital, Assistance Publique-Hopitaux de Paris, Paris
| | - Bertrand Knebelmann
- Department of Nephrology, Necker Hospital, Assistance Publique-Hopitaux de Paris, Paris
| | - Nathalie Maisonneuve
- Service de Néphrologie-Dialyse, Centre Hospitalier de Valenciennes, Valenciennes, France
| | - Jan Halbritter
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedicin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universittät zu Berlin, Berlin, Germany
| | - Yannick Le Meur
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de Référence MARHEA, Brest; University of Brest, UMR 1227, B Lymphocytes, Autoimmunity and Immunotherapies, Labex IGO, Brest, France
| | - Marie-Pierre Audrézet
- Service de Génétique Moléculaire, CHRU Brest, Brest; University of Brest, Inserm, UMR 1078, Génétique, Génomique Fonctionelle et Biotechnologies, Brest
| | - Emilie Cornec-Le Gall
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de Référence MARHEA, Brest; University of Brest, Inserm, UMR 1078, Génétique, Génomique Fonctionelle et Biotechnologies, Brest.
| |
Collapse
|
3
|
Beyer T, Diwan GD, Leonhard T, Dahlke K, Klose F, Stehle IF, Seda M, Bolz S, Woerz F, Russell RB, Jenkins D, Ueffing M, Boldt K. Ciliopathy-Associated Missense Mutations in IFT140 are Tolerated by the Inherent Resilience of the IFT Machinery. Mol Cell Proteomics 2025; 24:100916. [PMID: 39880085 PMCID: PMC11907452 DOI: 10.1016/j.mcpro.2025.100916] [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: 04/25/2024] [Revised: 11/26/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
Genotype-phenotype correlations of rare diseases are complicated by low patient number, high phenotype variability, and compound heterozygosity. Mutations may cause instability of single proteins, and affect protein complex formation or overall robustness of a specific process in a given cell. Ciliopathies offer an interesting case for studying genotype-phenotype correlations as they have a spectrum of severity and include diverse phenotypes depending on different mutations in the same protein. For instance, mutations in the intraflagellar transport protein IFT140 cause a vast spectrum of ciliopathies ranging from isolated retinal dystrophy to severe skeletal abnormalities and multi-organ diseases such as Mainzer-Saldino and Jeune syndrome. Here, the quantitative effects of 23 missense mutations in IFT140, which forms part of the crucial IFT-A complex of the ciliary machinery, were analyzed using affinity purification coupled with mass spectrometry (AP-MS). A subset of 10 mutations led to a significant and domain-specific reduction in IFT140-IFT-A complex interaction indicating complex formation issues and potentially hampering its molecular function. Knockout of IFT140 led to loss of cilia, as shown before. However, phenotypically only mild effects concerning cilia assembly were observed for two out of four tested IFT140 missense mutations. Therefore, our results demonstrate the utility of AP-MS in discerning pathogenic MMs from polymorphisms, and we postulate that reduced function is tolerated by the evolutionarily highly conserved IFT-A system.
Collapse
Affiliation(s)
- Tina Beyer
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany.
| | - Gaurav D Diwan
- BioQuant, University of Heidelberg, Heidelberg, Germany; Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Tobias Leonhard
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Katrin Dahlke
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Franziska Klose
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Isabel F Stehle
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Marian Seda
- UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Sylvia Bolz
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Franziska Woerz
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Robert B Russell
- BioQuant, University of Heidelberg, Heidelberg, Germany; Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Dagan Jenkins
- UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.
| | - Marius Ueffing
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Karsten Boldt
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
4
|
Adeghate J, Goldburg SR, Bass S, Schwimmer J, Kaden TR. Novel Pathogenic Variants in IFT140 and IFT172 Genes in Three Patients with Similar Retinal Dystrophy Phenotypes. Case Rep Ophthalmol 2025; 16:323-330. [PMID: 40370963 PMCID: PMC12077866 DOI: 10.1159/000545390] [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: 11/05/2024] [Accepted: 03/05/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction The intraflagellar transport (IFT) complex plays a key role in protein transport and turnover within photoreceptors. IFT140 and IFT172 gene mutations have been associated with skeletal ciliopathies that occur concurrently with retinal dystrophy. These mutations have also been associated with non-syndromic retinal dystrophies. This phenotypic heterogeneity can make diagnosis challenging. Here, we report novel variants in IFT140 and IFT172 genes in 3 patients with similar retinal dystrophy phenotypes. Case Presentations Two siblings (a 51-year-old male and 46-year-old male) who presented with a similar retinal dystrophy, skeletal abnormalities, and kidney disease were found to have the same novel variant in the IFT140 gene, along with another, previously reported variant. An unrelated individual with a similar retinal phenotype was found to have a novel variant in the IFT172 gene, although this was noted as a variant of uncertain significance. The patients underwent testing with the Blueprint Genetics (Blueprint Genetics Oy, Keilaranta 16 A-B, 02150 Espoo, Finland) "My Retina Tracker Program Panel Plus" panel. Conclusion Novel variants in the IFT140 and IFT172 genes encoding the IFT complex may contribute to similar retinal dystrophy phenotypes, as noted in our case series.
Collapse
Affiliation(s)
- Jennifer Adeghate
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
- Northwell Health, New Hyde Park, NY, USA
- Department of Ophthalmology, Manhattan Eye, Ear and Throat Hospital, New York, NY, USA
| | - Samantha R. Goldburg
- Northwell Health, New Hyde Park, NY, USA
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Sherry Bass
- State University of New York, College of Optometry, University Eye Center, New York, NY, USA
| | - Joshua Schwimmer
- Northwell Health, New Hyde Park, NY, USA
- Department of Medicine, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Talia R. Kaden
- Northwell Health, New Hyde Park, NY, USA
- Department of Ophthalmology, Manhattan Eye, Ear and Throat Hospital, New York, NY, USA
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
5
|
Karuntu JS, Almushattat H, Nguyen XTA, Plomp AS, Wanders RJA, Hoyng CB, van Schooneveld MJ, Schalij-Delfos NE, Brands MM, Leroy BP, van Karnebeek CDM, Bergen AA, van Genderen MM, Boon CJF. Syndromic retinitis pigmentosa. Prog Retin Eye Res 2024; 107:101324. [PMID: 39733931 DOI: 10.1016/j.preteyeres.2024.101324] [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: 07/17/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024]
Abstract
Retinitis pigmentosa (RP) is a progressive inherited retinal dystrophy, characterized by the degeneration of photoreceptors, presenting as a rod-cone dystrophy. Approximately 20-30% of patients with RP also exhibit extra-ocular manifestations in the context of a syndrome. This manuscript discusses the broad spectrum of syndromes associated with RP, pathogenic mechanisms, clinical manifestations, differential diagnoses, clinical management approaches, and future perspectives. Given the diverse clinical and genetic landscape of syndromic RP, the diagnosis may be challenging. However, an accurate and timely diagnosis is essential for optimal clinical management, prognostication, and potential treatment. Broadly, the syndromes associated with RP can be categorized into ciliopathies, inherited metabolic disorders, mitochondrial disorders, and miscellaneous syndromes. Among the ciliopathies associated with RP, Usher syndrome and Bardet-Biedl syndrome are the most well-known. Less common ciliopathies include Cohen syndrome, Joubert syndrome, cranioectodermal dysplasia, asphyxiating thoracic dystrophy, Mainzer-Saldino syndrome, and RHYNS syndrome. Several inherited metabolic disorders can present with RP, including Zellweger spectrum disorders, adult Refsum disease, α-methylacyl-CoA racemase deficiency, certain mucopolysaccharidoses, ataxia with vitamin E deficiency, abetalipoproteinemia, several neuronal ceroid lipofuscinoses, mevalonic aciduria, PKAN/HARP syndrome, PHARC syndrome, and methylmalonic acidaemia with homocystinuria type cobalamin (cbl) C disease. Due to the mitochondria's essential role in supplying continuous energy to the retina, disruption of mitochondrial function can lead to RP, as seen in Kearns-Sayre syndrome, NARP syndrome, primary coenzyme Q10 deficiency, SSBP1-associated disease, and long chain 3-hydroxyacyl-CoA dehydrogenase deficiency. Lastly, Cockayne syndrome and PERCHING syndrome can present with RP, but they do not fit the abovementioned hierarchy and are thus categorized as miscellaneous. Several first-in-human clinical trials are underway or in preparation for some of these syndromic forms of RP.
Collapse
Affiliation(s)
- Jessica S Karuntu
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hind Almushattat
- Department of Ophthalmology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Xuan-Thanh-An Nguyen
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - Astrid S Plomp
- Department of Human Genetics, Amsterdam Reproduction & Development, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Ronald J A Wanders
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam, the Netherlands; Department of Laboratory Medicine, Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam, the Netherlands
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mary J van Schooneveld
- Bartiméus Diagnostic Center for Complex Visual Disorders, Zeist, the Netherlands; Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Marion M Brands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Bart P Leroy
- Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium; Department of Head & Skin, Ghent University, Ghent, Belgium; Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Division of Ophthalmology and Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Clara D M van Karnebeek
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Arthur A Bergen
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands; Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Department of Human Genetics, Section Ophthalmogenetics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Maria M van Genderen
- Bartiméus Diagnostic Center for Complex Visual Disorders, Zeist, the Netherlands; Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Camiel J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Datta P, Rhee KD, Staudt RJ, Thompson JM, Hsu Y, Hassan S, Drack AV, Seo S. Delivering large genes using adeno-associated virus and the CRE-lox DNA recombination system. Hum Mol Genet 2024; 33:2094-2110. [PMID: 39393808 PMCID: PMC11630788 DOI: 10.1093/hmg/ddae144] [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: 03/27/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/13/2024] Open
Abstract
Adeno-associated virus (AAV) is a safe and efficient gene delivery vehicle for gene therapies. However, its relatively small packaging capacity limits its use as a gene transfer vector. Here, we describe a strategy to deliver large genes that exceed the AAV's packaging capacity using up to four AAV vectors and the CRE-lox DNA recombination system. We devised novel lox sites by combining non-compatible and reaction equilibrium-modifying lox site variants. These lox sites facilitate sequence-specific and near-unidirectional recombination of AAV vector genomes, enabling efficient reconstitution of up to 16 kb of therapeutic genes in a pre-determined configuration. Using this strategy, we have developed AAV gene therapy vectors to deliver IFT140, PCDH15, CEP290, and CDH23 and demonstrate efficient production of full-length proteins in cultured mammalian cells and mouse retinas. Notably, AAV-IFT140 gene therapy vectors ameliorated retinal degeneration and preserved visual functions in an IFT140-associated retinitis pigmentosa mouse model. The CRE-lox approach described here provides a simple, flexible, and effective platform for generating AAV gene therapy vectors beyond AAV's packaging capacity.
Collapse
Affiliation(s)
- Poppy Datta
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
- Institute for Vision Research, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
| | - Kun-Do Rhee
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
- Institute for Vision Research, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
| | - Rylee J Staudt
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
- Institute for Vision Research, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
| | - Jacob M Thompson
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
- Institute for Vision Research, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
| | - Ying Hsu
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
- Institute for Vision Research, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
| | - Salma Hassan
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
- Institute for Vision Research, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
| | - Arlene V Drack
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
- Institute for Vision Research, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
- Department of Pediatrics, The University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, United States
| | - Seongjin Seo
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
- Institute for Vision Research, The University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, United States
| |
Collapse
|
7
|
Gupta M, Pazour GJ. Intraflagellar transport: A critical player in photoreceptor development and the pathogenesis of retinal degenerative diseases. Cytoskeleton (Hoboken) 2024; 81:556-568. [PMID: 38140908 PMCID: PMC11193844 DOI: 10.1002/cm.21823] [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: 11/04/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
In vertebrate vision, photons are detected by highly specialized sensory cilia called outer segments. Photoreceptor outer segments form by remodeling the membrane of a primary cilium into a stack of flattened disks. Intraflagellar transport (IFT) is critical to the formation of most types of eukaryotic cilia including the outer segments. This review covers the state of knowledge of the role of IFT in the formation and maintenance of outer segments and the human diseases that result from mutations in genes encoding the IFT complex and associated motors.
Collapse
Affiliation(s)
- Mohona Gupta
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Morningside Graduate School of Biological Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
8
|
Danish E, Alhashem A, Naaman N, Almhmoudi F, Mushiba A, Almatrafi A, Raggam A, Bukhari R. Mutations in the ciliary transport gene IFT140 cause syndromic congenital retinal dystrophy. J AAPOS 2024; 28:104007. [PMID: 39304031 DOI: 10.1016/j.jaapos.2024.104007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 09/22/2024]
Abstract
Early-onset, severe retinal dystrophy can be isolated or syndromic, presenting as part of an underlying systemic disease. Mainzer-Saldino syndrome, a rare systemic ciliopathy characterized by skeletal and renal disease, is caused by recessive mutations in the intraflagellar transport 140 chlamydomonas homologue (IFT140) gene. We present a series of 13 cases of early-onset retinal dysfunction with confirmed IFT140 mutations from 8 unrelated Saudi families belonging to 3 well-known tribes. All carried the same homozygous missense IFT140 mutation (c.1990G>A; p.Glu664Lys) except for a single family, which included 4 affected subjects, 3 of whom were aborted fetuses, with compound heterozygous pathogenic IFT140 variants (c.1525-1G>A and c.1990G>A; p.Glu664Lys). Severe retinal dystrophy was present in all living subjects, phenotypically apparent as hyperopia, nystagmus, nyctalopia, poor vision and nonrecordable full-field electroretinography. All affected individuals had skeletal abnormalities, and neurological abnormalities were common, but there was no evidence of chronic renal failure.
Collapse
Affiliation(s)
- Enam Danish
- Department of Ophthalmology, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Amal Alhashem
- Department of Pediatric, Division of Genetic and Metabolic Medicine, Prince Sultan Military Medical City, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Nada Naaman
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia; King Abdullah International Medical Research Center, Jeddah, Saudi Arabia; Department of Ophthalmology, Ministry of National Guard - Health Affairs, Jeddah, Saudi Arabia.
| | - Faeeqah Almhmoudi
- Department of Ophthalmology, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Aziza Mushiba
- Section of Medical Genetics, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Abdulaziz Raggam
- Department of Medicine, King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | - Raghad Bukhari
- College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
9
|
Huang L, Kellom E, Stepien K. Male infertility may be associated with IFT140-related autosomal recessive retinitis pigmentosa. Ophthalmic Genet 2024; 45:267-270. [PMID: 38084016 DOI: 10.1080/13816810.2023.2291672] [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: 05/19/2023] [Revised: 11/06/2023] [Accepted: 11/30/2023] [Indexed: 08/09/2024]
Abstract
BACKGROUND Pathogenic variants in IFT140 have been reported in cases of both syndromic and nonsyndromic retinitis pigmentosa (RP). Syndromic forms of IFT140-related RP have been associated with short-rib thoracic dysplasia. IFT140 variants have also been shown to cause spermatogenic dysfunction leading to infertility. However, variants in IFT140 have not been reported in patients with concurrent RP (including nonsyndromic RP) and infertility. METHODS A chart review was performed in a 42 year old male with RP and male factor infertility. RESULTS Genetic testing confirmed this patient's RP was related to variants in IFT140. Chart review and exam confirmed no findings consistent with short-rib thoracic dysplasia, leading to the conclusion that this was a form of nonsyndromic RP as has been previously reported. However, the patient had undergone an infertility workup with findings of spermatogenic dysfunction as found in other males with IFT140-related infertility. This has led us to speculate this patient may have a syndromic form of IFT140 -related RP associated with infertility and abnormal spermatogenesis. CONCLUSIONS A potential association between IFT140-related RP and male factor infertility may exist.
Collapse
Affiliation(s)
- Leslie Huang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Elizabeth Kellom
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Genetics, University of Wisconsin Madison - Waisman Center, Madison, Wisconsin, USA
| | - Kimberly Stepien
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
10
|
Datta P, Rhee KD, Staudt RJ, Thompson JM, Hsu Y, Hassan S, Drack AV, Seo S. Delivering large genes using adeno-associated virus and the CRE-lox DNA recombination system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588864. [PMID: 38645107 PMCID: PMC11030439 DOI: 10.1101/2024.04.10.588864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Adeno-associated virus (AAV) is a safe and efficient gene delivery vehicle for gene therapies. However, its relatively small packaging capacity limits its use as a gene transfer vector. Here, we describe a strategy to deliver large genes that exceed the AAV's packaging capacity using up to four AAV vectors and the CRE-lox DNA recombination system. We devised novel lox sites by combining non-compatible and reaction equilibrium-modifying lox site variants. These lox sites facilitate sequence-specific and near-unidirectional recombination of AAV vector genomes, enabling efficient reconstitution of up to 16 kb of therapeutic genes in a pre-determined configuration. Using this strategy, we have developed AAV gene therapy vectors to deliver IFT140 , PCDH15 , CEP290 , and CDH23 and demonstrate efficient production of full-length proteins in cultured mammalian cells and mouse retinas. Notably, this approach significantly surpasses the trans-splicing and split-intein-based reconstitution methods in efficiency, requiring lower doses, minimizing or eliminating the production of truncated protein products, and offering flexibility in selecting splitting positions. The CRE-lox approach described here provides a simple and effective platform for producing AAV gene therapy vectors beyond AAV's packaging capacity.
Collapse
|
11
|
Pazour GJ. Cilia Structure and Function in Human Disease. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2024; 34:100509. [PMID: 38836197 PMCID: PMC11147146 DOI: 10.1016/j.coemr.2024.100509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Ciliary dysfunction causes a large group of developmental and degenerative human diseases known as ciliopathies. These diseases reflect the critical roles that cilia play in sensing the environment and in force generation for motility. Sensory functions include our senses of vision and olfaction. In addition, primary and motile cilia throughout our body monitor the environment allowing cells to coordinate their biology with the cells around them. This coordination is critical to organ development and maintenance, and ciliary dysfunction causes diverse structural birth defects and degenerative diseases. Defects in motility cause lung disease due to the failure of mucociliary clearance, male infertility due to the failure of sperm motility and the ability of sperm to move through the efferent ducts, and disturbances of the left-right axis due to a failure of nodal cilia to establish proper left-right cues.
Collapse
Affiliation(s)
- Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Biotech II, Worcester, Massachusetts, USA
| |
Collapse
|
12
|
Cevik S, Zhao P, Zorluer A, Pir MS, Bian W, Kaplan OI. Matching variants for functional characterization of genetic variants. G3 (BETHESDA, MD.) 2023; 13:jkad227. [PMID: 37933433 PMCID: PMC10700107 DOI: 10.1093/g3journal/jkad227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 09/06/2023] [Indexed: 11/08/2023]
Abstract
Rapid and low-cost sequencing, as well as computer analysis, have facilitated the diagnosis of many genetic diseases, resulting in a substantial rise in the number of disease-associated genes. However, genetic diagnosis of many disorders remains problematic due to the lack of interpretation for many genetic variants, especially missenses, the infeasibility of high-throughput experiments on mammals, and the shortcomings of computational prediction technologies. Additionally, the available mutant databases are not well-utilized. Toward this end, we used Caenorhabditis elegans mutant resources to delineate the functions of eight missense variants (V444I, V517D, E610K, L732F, E817K, H873P, R1105K, and G1205E) and two stop codons (W937stop and Q1434stop), including several matching variants (MatchVar) with human in ciliopathy associated IFT-140 (also called CHE-11)//IFT140 (intraflagellar transport protein 140). Moreover, MatchVars carrying C. elegans mutants, including IFT-140(G680S) and IFT-140(P702A) for the human (G704S) (dbSNP: rs150745099) and P726A (dbSNP: rs1057518064 and a conflicting variation) were created using CRISPR/Cas9. IFT140 is a key component of IFT complex A (IFT-A), which is involved in the retrograde transport of IFT along cilia and the entrance of G protein-coupled receptors into cilia. Functional analysis of all 10 variants revealed that P702A and W937stop, but not others phenocopied the ciliary phenotypes (short cilia, IFT accumulations, mislocalization of membrane proteins, and cilia entry of nonciliary proteins) of the IFT-140 null mutant, indicating that both P702A and W937stop are phenotypic in C. elegans. Our functional data offered experimental support for interpreting human variants, by using ready-to-use mutants carrying MatchVars and generating MatchVars with CRISPR/Cas9.
Collapse
Affiliation(s)
- Sebiha Cevik
- Rare Disease Laboratory, School of Life and Natural Sciences, Abdullah Gul University, Kayseri 38080, Turkey
| | - Pei Zhao
- School of Applied Science and Engineering, Fuzhou Institute of Technology, Fuzhou 350014, China
- SunyBiotech Co., Ltd., Fuzhou 35000, China
| | - Atiyye Zorluer
- Rare Disease Laboratory, School of Life and Natural Sciences, Abdullah Gul University, Kayseri 38080, Turkey
| | - Mustafa S Pir
- Rare Disease Laboratory, School of Life and Natural Sciences, Abdullah Gul University, Kayseri 38080, Turkey
| | | | - Oktay I Kaplan
- Rare Disease Laboratory, School of Life and Natural Sciences, Abdullah Gul University, Kayseri 38080, Turkey
| |
Collapse
|
13
|
Mill P, Christensen ST, Pedersen LB. Primary cilia as dynamic and diverse signalling hubs in development and disease. Nat Rev Genet 2023; 24:421-441. [PMID: 37072495 PMCID: PMC7615029 DOI: 10.1038/s41576-023-00587-9] [Citation(s) in RCA: 152] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 04/20/2023]
Abstract
Primary cilia, antenna-like sensory organelles protruding from the surface of most vertebrate cell types, are essential for regulating signalling pathways during development and adult homeostasis. Mutations in genes affecting cilia cause an overlapping spectrum of >30 human diseases and syndromes, the ciliopathies. Given the immense structural and functional diversity of the mammalian cilia repertoire, there is a growing disconnect between patient genotype and associated phenotypes, with variable severity and expressivity characteristic of the ciliopathies as a group. Recent technological developments are rapidly advancing our understanding of the complex mechanisms that control biogenesis and function of primary cilia across a range of cell types and are starting to tackle this diversity. Here, we examine the structural and functional diversity of primary cilia, their dynamic regulation in different cellular and developmental contexts and their disruption in disease.
Collapse
Affiliation(s)
- Pleasantine Mill
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | | | - Lotte B Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
14
|
Ma Y, He J, Li S, Yao D, Huang C, Wu J, Lei M. Structural insight into the intraflagellar transport complex IFT-A and its assembly in the anterograde IFT train. Nat Commun 2023; 14:1506. [PMID: 36932088 PMCID: PMC10023715 DOI: 10.1038/s41467-023-37208-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Intraflagellar transport (IFT) trains, the polymers composed of two multi-subunit complexes, IFT-A and IFT-B, carry out bidirectional intracellular transport in cilia, vital for cilia biogenesis and signaling. IFT-A plays crucial roles in the ciliary import of membrane proteins and the retrograde cargo trafficking. However, the molecular architecture of IFT-A and the assembly mechanism of the IFT-A into the IFT trains in vivo remains elusive. Here, we report the cryo-electron microscopic structures of the IFT-A complex from protozoa Tetrahymena thermophila. We find that IFT-A complexes present two distinct, elongated and folded states. Remarkably, comparison with the in situ cryo-electron tomography structure of the anterograde IFT train unveils a series of adjustments of the flexible arms in apo IFT-A when incorporated into the anterograde train. Our results provide an atomic-resolution model for the IFT-A complex and valuable insights into the assembly mechanism of anterograde IFT trains.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Institute of Precision Medicine, Shanghai, 200125, China
| | - Jun He
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Institute of Precision Medicine, Shanghai, 200125, China
| | - Shaobai Li
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Institute of Precision Medicine, Shanghai, 200125, China
| | - Deqiang Yao
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Chenhui Huang
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Institute of Precision Medicine, Shanghai, 200125, China
| | - Jian Wu
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Institute of Precision Medicine, Shanghai, 200125, China.
| | - Ming Lei
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Institute of Precision Medicine, Shanghai, 200125, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
15
|
Cellular and Molecular Mechanisms of Pathogenesis Underlying Inherited Retinal Dystrophies. Biomolecules 2023; 13:biom13020271. [PMID: 36830640 PMCID: PMC9953031 DOI: 10.3390/biom13020271] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are congenital retinal degenerative diseases that have various inheritance patterns, including dominant, recessive, X-linked, and mitochondrial. These diseases are most often the result of defects in rod and/or cone photoreceptor and retinal pigment epithelium function, development, or both. The genes associated with these diseases, when mutated, produce altered protein products that have downstream effects in pathways critical to vision, including phototransduction, the visual cycle, photoreceptor development, cellular respiration, and retinal homeostasis. The aim of this manuscript is to provide a comprehensive review of the underlying molecular mechanisms of pathogenesis of IRDs by delving into many of the genes associated with IRD development, their protein products, and the pathways interrupted by genetic mutation.
Collapse
|
16
|
Zhang X, Zhou J, Wang X, Geng J, Chen Y, Sun Y. IFT140 +/K14 + cells function as stem/progenitor cells in salivary glands. Int J Oral Sci 2022; 14:49. [PMID: 36216809 PMCID: PMC9550827 DOI: 10.1038/s41368-022-00200-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/31/2022] [Accepted: 09/05/2022] [Indexed: 11/27/2022] Open
Abstract
Stem/progenitor cells are important for salivary gland development, homeostasis maintenance, and regeneration following injury. Keratin-14+ (K14+) cells have been recognized as bona fide salivary gland stem/progenitor cells. However, K14 is also expressed in terminally differentiated myoepithelial cells; therefore, more accurate molecular markers for identifying salivary stem/progenitor cells are required. The intraflagellar transport (IFT) protein IFT140 is a core component of the IFT system that functions in signaling transduction through the primary cilia. It is reportedly expressed in mesenchymal stem cells and plays a role in bone formation. In this study, we demonstrated that IFT140 was intensively expressed in K14+ stem/progenitor cells during the developmental period and early regeneration stage following ligation-induced injuries in murine submandibular glands. In addition, we demonstrated that IFT140+/ K14+ could self-renew and differentiate into granular duct cells at the developmental stage in vivo. The conditional deletion of Ift140 from K14+ cells caused abnormal epithelial structure and function during salivary gland development and inhibited regeneration. IFT140 partly coordinated the function of K14+ stem/progenitor cells by modulating ciliary membrane trafficking. Our investigation identified a combined marker, IFT140+/K14+, for salivary gland stem/progenitor cells and elucidated the essential role of IFT140 and cilia in regulating salivary stem/progenitor cell differentiation and gland regeneration.
Collapse
Affiliation(s)
- Xueming Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No. 399, YanChang Middle Road, Shanghai, China
| | - Ji Zhou
- Department of Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No. 399, YanChang Middle Road, Shanghai, China
| | - Xinyu Wang
- Department of Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No. 399, YanChang Middle Road, Shanghai, China
| | - Jiangyu Geng
- Department of Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No. 399, YanChang Middle Road, Shanghai, China
| | - Yubei Chen
- Department of Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No. 399, YanChang Middle Road, Shanghai, China
| | - Yao Sun
- Department of Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No. 399, YanChang Middle Road, Shanghai, China.
| |
Collapse
|
17
|
Alachkar A. Aromatic patterns: Tryptophan aromaticity as a catalyst for the emergence of life and rise of consciousness. Phys Life Rev 2022; 42:93-114. [PMID: 35905538 DOI: 10.1016/j.plrev.2022.07.002] [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: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 11/28/2022]
Abstract
Sunlight held the key to the origin of life on Earth. The earliest life forms, cyanobacteria, captured the sunlight to generate energy through photosynthesis. Life on Earth evolved in accordance with the circadian rhythms tied to sensitivity to sunlight patterns. A unique feature of cyanobacterial photosynthetic proteins and circadian rhythms' molecules, and later of nearly all photon-sensing molecules throughout evolution, is that the aromatic amino acid tryptophan (Trp) resides at the center of light-harvesting active sites. In this perspective, I review the literature and integrate evidence from different scientific fields to explore the role Trp plays in photon-sensing capabilities of living organisms through its resonance delocalization of π-electrons. The observations presented here are the product of apparently unrelated phenomena throughout evolution, but nevertheless share consistent patterns of photon-sensing by Trp-containing and Trp-derived molecules. I posit the unique capacity to transfer electrons during photosynthesis in the earliest life forms is conferred to Trp due to its aromaticity. I propose this ability evolved to assume more complex functions, serving as a host for mechanisms underlying mental aptitudes - a concept which provides a theoretical basis for defining the neural correlates of consciousness. The argument made here is that Trp aromaticity may have allowed for the inception of the mechanistic building blocks used to fabricate complexity in higher forms of life. More specifically, Trp aromatic non-locality may have acted as a catalyst for the emergence of consciousness by instigating long-range synchronization and stabilizing the large-scale coherence of neural networks, which mediate functional brain activity. The concepts proposed in this perspective provide a conceptual foundation that invites further interdisciplinary dialogue aimed at examining and defining the role of aromaticity (beyond Trp) in the emergence of life and consciousness.
Collapse
Affiliation(s)
- Amal Alachkar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA; UC Irvine Center for the Neurobiology of Learning and Memory, University of California-Irvine, Irvine, CA 92697, USA; Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
18
|
Bhardwaj A, Yadav A, Yadav M, Tanwar M. Genetic dissection of non-syndromic retinitis pigmentosa. Indian J Ophthalmol 2022; 70:2355-2385. [PMID: 35791117 PMCID: PMC9426071 DOI: 10.4103/ijo.ijo_46_22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Retinitis pigmentosa (RP) belongs to a group of pigmentary retinopathies. It is the most common form of inherited retinal dystrophy, characterized by progressive degradation of photoreceptors that leads to nyctalopia, and ultimately, complete vision loss. RP is distinguished by the continuous retinal degeneration that progresses from the mid-periphery to the central and peripheral retina. RP was first described and named by Franciscus Cornelius Donders in the year 1857. It is one of the leading causes of bilateral blindness in adults, with an incidence of 1 in 3000 people worldwide. In this review, we are going to focus on the genetic heterogeneity of this disease, which is provided by various inheritance patterns, numerosity of variations and inter-/intra-familial variations based upon penetrance and expressivity. Although over 90 genes have been identified in RP patients, the genetic cause of approximately 50% of RP cases remains unknown. Heterogeneity of RP makes it an extremely complicated ocular impairment. It is so complicated that it is known as “fever of unknown origin”. For prognosis and proper management of the disease, it is necessary to understand its genetic heterogeneity so that each phenotype related to the various genetic variations could be treated.
Collapse
Affiliation(s)
- Aarti Bhardwaj
- Department of Genetics, M. D. University, Rohtak, Haryana, India
| | - Anshu Yadav
- Department of Genetics, M. D. University, Rohtak, Haryana, India
| | - Manoj Yadav
- Department of Genetics, M. D. University, Rohtak, Haryana, India
| | - Mukesh Tanwar
- Department of Genetics, M. D. University, Rohtak, Haryana, India
| |
Collapse
|
19
|
Abstract
Inherited retinal dystrophies (IRDs) are a heterogeneous group of diseases that affect more than 2 million people worldwide. Gene therapy (GT) has emerged as an exciting treatment modality with the potential to provide long-term benefit to patients. Today, gene addition is the most straightforward GT for autosomal recessive IRDs. However, there are three scenarios where this approach falls short. First, in autosomal dominant diseases caused by gain-of-function or dominant-negative mutations, the toxic mutated protein needs to be silenced. Second, a number of IRD genes exceed the limited carrying capacity of adeno-associated virus vectors. Third, there are still about 30% of patients with unknown mutations. In the first two contexts, precise editing tools, such as CRISPR-Cas9, base editors, or prime editors, are emerging as potential GT solutions for the treatment of IRDs. Here, we review gene editing tools based on CRISPR-Cas9 technology that have been used in vivo and the recent first-in-human application of CRISPR-Cas9 in an IRD.
Collapse
Affiliation(s)
- Juliette Pulman
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France.,Fondation Ophtalmologique Rothschild, Paris, France
| | - Deniz Dalkara
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
20
|
Wang L, Sun L, Wan QH, Fang SG. Comparative Genomics Provides Insights into Adaptive Evolution in Tactile-Foraging Birds. Genes (Basel) 2022; 13:genes13040678. [PMID: 35456484 PMCID: PMC9028243 DOI: 10.3390/genes13040678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 11/28/2022] Open
Abstract
Tactile-foraging birds have evolved an enlarged principal sensory nucleus (PrV) but smaller brain regions related to the visual system, which reflects the difference in sensory dependence. The “trade-off” may exist between different senses in tactile foragers, as well as between corresponding sensory-processing areas in the brain. We explored the mechanism underlying the adaptive evolution of sensory systems in three tactile foragers (kiwi, mallard, and crested ibis). The results showed that olfaction-related genes in kiwi and mallard and hearing-related genes in crested ibis were expanded, indicating they may also have sensitive olfaction or hearing, respectively. However, some genes required for visual development were positively selected or had convergent amino acid substitutions in all three tactile branches, and it seems to show the possibility of visual degradation. In addition, we may provide a new visual-degradation candidate gene PDLIM1 who suffered dense convergent amino acid substitutions within the ZM domain. At last, two genes responsible for regulating the proliferation and differentiation of neuronal progenitor cells may play roles in determining the relative sizes of sensory areas in brain. This exploration offers insight into the relationship between specialized tactile-forging behavior and the evolution of sensory abilities and brain structures.
Collapse
|
21
|
Senum SR, Li Y(SM, Benson KA, Joli G, Olinger E, Lavu S, Madsen CD, Gregory AV, Neatu R, Kline TL, Audrézet MP, Outeda P, Nau CB, Meijer E, Ali H, Steinman TI, Mrug M, Phelan PJ, Watnick TJ, Peters DJ, Ong AC, Conlon PJ, Perrone RD, Cornec-Le Gall E, Hogan MC, Torres VE, Sayer JA, Harris PC, Harris PC. Monoallelic IFT140 pathogenic variants are an important cause of the autosomal dominant polycystic kidney-spectrum phenotype. Am J Hum Genet 2022; 109:136-156. [PMID: 34890546 DOI: 10.1016/j.ajhg.2021.11.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/15/2021] [Indexed: 12/18/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), characterized by progressive cyst formation/expansion, results in enlarged kidneys and often end stage kidney disease. ADPKD is genetically heterogeneous; PKD1 and PKD2 are the common loci (∼78% and ∼15% of families) and GANAB, DNAJB11, and ALG9 are minor genes. PKD is a ciliary-associated disease, a ciliopathy, and many syndromic ciliopathies have a PKD phenotype. In a multi-cohort/-site collaboration, we screened ADPKD-diagnosed families that were naive to genetic testing (n = 834) or for whom no PKD1 and PKD2 pathogenic variants had been identified (n = 381) with a PKD targeted next-generation sequencing panel (tNGS; n = 1,186) or whole-exome sequencing (WES; n = 29). We identified monoallelic IFT140 loss-of-function (LoF) variants in 12 multiplex families and 26 singletons (1.9% of naive families). IFT140 is a core component of the intraflagellar transport-complex A, responsible for retrograde ciliary trafficking and ciliary entry of membrane proteins; bi-allelic IFT140 variants cause the syndromic ciliopathy, short-rib thoracic dysplasia (SRTD9). The distinctive monoallelic phenotype is mild PKD with large cysts, limited kidney insufficiency, and few liver cysts. Analyses of the cystic kidney disease probands of Genomics England 100K showed that 2.1% had IFT140 LoF variants. Analysis of the UK Biobank cystic kidney disease group showed probands with IFT140 LoF variants as the third most common group, after PKD1 and PKD2. The proximity of IFT140 to PKD1 (∼0.5 Mb) in 16p13.3 can cause diagnostic confusion, and PKD1 variants could modify the IFT140 phenotype. Importantly, our studies link a ciliary structural protein to the ADPKD spectrum.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
22
|
Amato A, Arrigo A, Aragona E, Manitto MP, Saladino A, Bandello F, Battaglia Parodi M. Gene Therapy in Inherited Retinal Diseases: An Update on Current State of the Art. Front Med (Lausanne) 2021; 8:750586. [PMID: 34722588 PMCID: PMC8553993 DOI: 10.3389/fmed.2021.750586] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Gene therapy cannot be yet considered a far perspective, but a tangible therapeutic option in the field of retinal diseases. Although still confined in experimental settings, the preliminary results are promising and provide an overall scenario suggesting that we are not so far from the application of gene therapy in clinical settings. The main aim of this review is to provide a complete and updated overview of the current state of the art and of the future perspectives of gene therapy applied on retinal diseases. Methods: We carefully revised the entire literature to report all the relevant findings related to the experimental procedures and the future scenarios of gene therapy applied in retinal diseases. A clinical background and a detailed description of the genetic features of each retinal disease included are also reported. Results: The current literature strongly support the hope of gene therapy options developed for retinal diseases. Although being considered in advanced stages of investigation for some retinal diseases, such as choroideremia (CHM), retinitis pigmentosa (RP), and Leber's congenital amaurosis (LCA), gene therapy is still quite far from a tangible application in clinical practice for other retinal diseases. Conclusions: Gene therapy is an extremely promising therapeutic tool for retinal diseases. The experimental data reported in this review offer a strong hope that gene therapy will be effectively available in clinical practice in the next years.
Collapse
Affiliation(s)
- Alessia Amato
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Alessandro Arrigo
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Emanuela Aragona
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Maria Pia Manitto
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Andrea Saladino
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Francesco Bandello
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | | |
Collapse
|
23
|
Wheway G, Thomas NS, Carroll M, Coles J, Doherty R, Goggin P, Green B, Harris A, Hunt D, Jackson CL, Lord J, Mennella V, Thompson J, Walker WT, Lucas JS. Whole genome sequencing in the diagnosis of primary ciliary dyskinesia. BMC Med Genomics 2021; 14:234. [PMID: 34556108 PMCID: PMC8461892 DOI: 10.1186/s12920-021-01084-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 09/14/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND It is estimated that 1-13% of cases of bronchiectasis in adults globally are attributable to primary ciliary dyskinesia (PCD) but many adult patients with bronchiectasis have not been investigated for PCD. PCD is a disorder caused by mutations in genes required for motile cilium structure or function, resulting in impaired mucociliary clearance. Symptoms appear in infancy but diagnosis is often late or missed, often due to the lack of a "gold standard" diagnostic tool and non-specific symptoms. Mutations in > 50 genes account for around 70% of cases, with additional genes, and non-coding, synonymous, missense changes or structural variants (SVs) in known genes presumed to account for the missing heritability. METHODS UK patients with no identified genetic confirmation for the cause of their PCD or bronchiectasis were eligible for whole genome sequencing (WGS) in the Genomics England Ltd 100,000 Genomes Project. 21 PCD probands and 52 non-cystic fibrosis (CF) bronchiectasis probands were recruited in Wessex Genome Medicine Centre (GMC). We carried out analysis of single nucleotide variants (SNVs) and SVs in all families recruited in Wessex GMC. RESULTS 16/21 probands in the PCD cohort received confirmed (n = 9), probable (n = 4) or possible (n = 3) diagnosis from WGS, although 13/16 of these could have been picked up by current standard of care gene panel testing. In the other cases, SVs were identified which were missed by panel testing. We identified variants in novel PCD candidate genes (IFT140 and PLK4) in 2 probands in the PCD cohort. 3/52 probands in the non-CF bronchiectasis cohort received a confirmed (n = 2) or possible (n = 1) diagnosis of PCD. We identified variants in novel PCD candidate genes (CFAP53 and CEP164) in 2 further probands in the non-CF bronchiectasis cohort. CONCLUSIONS Genetic testing is an important component of diagnosing PCD, especially in cases of atypical disease history. WGS is effective in cases where prior gene panel testing has found no variants or only heterozygous variants. In these cases it may detect SVs and is a powerful tool for novel gene discovery.
Collapse
Affiliation(s)
- Gabrielle Wheway
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.
- Institute for Life Sciences, University of Southampton, Southampton, UK.
| | - N Simon Thomas
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- Wessex Regional Genetics Laboratory, Salisbury NSF Foundation Trust, Salisbury District Hospital, Salisbury, UK
| | - Mary Carroll
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Janice Coles
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Regan Doherty
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Biomedical Imaging Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Patricia Goggin
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Biomedical Imaging Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ben Green
- Queen Alexandra Hospital, Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | - Amanda Harris
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - David Hunt
- Wessex Clinical Genetics Service, University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - Claire L Jackson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jenny Lord
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Vito Mennella
- Institute for Life Sciences, University of Southampton, Southampton, UK
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - James Thompson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Woolf T Walker
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jane S Lucas
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| |
Collapse
|
24
|
Clinical exome sequencing facilitates the understanding of genetic heterogeneity in Leber congenital amaurosis patients with variable phenotype in southern India. EYE AND VISION 2021; 8:20. [PMID: 33957996 PMCID: PMC8101128 DOI: 10.1186/s40662-021-00243-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/14/2021] [Indexed: 11/30/2022]
Abstract
Background Leber congenital amaurosis (LCA), primarily characterized by retinal degeneration is the most severe form of inherited retinal dystrophy (IRD) responsible for congenital blindness. The presence of phenotypic heterogeneity makes the diagnosis of LCA challenging, especially in the absence of pronounced disease pathognomonic, yet it can be well comprehended by employing molecular diagnosis. Therefore, the present study aimed to reveal the causative mutations in ten LCA patients with variable phenotypes using clinical exome sequencing (CES). Methods CES was performed in ten unrelated LCA patients. Ophthalmic information and family history of all patients were obtained to make a meaningful interpretation. The clinical exome data was analyzed and prioritized using a bioinformatics pipeline to identify mutations, which was further validated by Sanger sequencing. Segregation analysis was also performed on available family members. Results CES led to the identification of causative mutations in nine LCA patients. Seven patients harbored a mutation in six LCA candidate genes, including RPE65, LCA5 (n = 2), CRX, PRPH2, CEP290, and ALMS1, while two patients possess a mutation in IFT80 and RP1, known to cause other diseases. Three novel mutations in LCA5 (c.1823del), CRX (c.848del) and CEP290 (c.2483G > T) were identified. The current study reports for the first time, a mutation in PRPH2, CEP290, and ALMS1 from the Indian population. Additionally, we observed a novel association of LCA phenotype with IFT80 known to cause Jeune syndrome. Based on the genetic finding, the patient AS09, who harbored a mutation in the RP1 gene, was re-diagnosed with early-onset retinitis pigmentosa. Conclusion In conclusion, the results underline the importance of CES in clinically diagnosed LCA patients with variable phenotypes. The correlation between mutations in candidate genes and clinical phenotypes, helps to refine the clinical diagnosis. However, molecular evaluation with a larger cohort of LCA patients is needed for better understanding of the mutational spectrum in southern India. Supplementary Information The online version contains supplementary material available at 10.1186/s40662-021-00243-5.
Collapse
|
25
|
Sánchez-Bellver L, Toulis V, Marfany G. On the Wrong Track: Alterations of Ciliary Transport in Inherited Retinal Dystrophies. Front Cell Dev Biol 2021; 9:623734. [PMID: 33748110 PMCID: PMC7973215 DOI: 10.3389/fcell.2021.623734] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/09/2021] [Indexed: 01/14/2023] Open
Abstract
Ciliopathies are a group of heterogeneous inherited disorders associated with dysfunction of the cilium, a ubiquitous microtubule-based organelle involved in a broad range of cellular functions. Most ciliopathies are syndromic, since several organs whose cells produce a cilium, such as the retina, cochlea or kidney, are affected by mutations in ciliary-related genes. In the retina, photoreceptor cells present a highly specialized neurosensory cilium, the outer segment, stacked with membranous disks where photoreception and phototransduction occurs. The daily renewal of the more distal disks is a unique characteristic of photoreceptor outer segments, resulting in an elevated protein demand. All components necessary for outer segment formation, maintenance and function have to be transported from the photoreceptor inner segment, where synthesis occurs, to the cilium. Therefore, efficient transport of selected proteins is critical for photoreceptor ciliogenesis and function, and any alteration in either cargo delivery to the cilium or intraciliary trafficking compromises photoreceptor survival and leads to retinal degeneration. To date, mutations in more than 100 ciliary genes have been associated with retinal dystrophies, accounting for almost 25% of these inherited rare diseases. Interestingly, not all mutations in ciliary genes that cause retinal degeneration are also involved in pleiotropic pathologies in other ciliated organs. Depending on the mutation, the same gene can cause syndromic or non-syndromic retinopathies, thus emphasizing the highly refined specialization of the photoreceptor neurosensory cilia, and raising the possibility of photoreceptor-specific molecular mechanisms underlying common ciliary functions such as ciliary transport. In this review, we will focus on ciliary transport in photoreceptor cells and discuss the molecular complexity underpinning retinal ciliopathies, with a special emphasis on ciliary genes that, when mutated, cause either syndromic or non-syndromic retinal ciliopathies.
Collapse
Affiliation(s)
- Laura Sánchez-Bellver
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine (IBUB-IRSJD), Universitat de Barcelona, Barcelona, Spain
| | - Vasileios Toulis
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona, Spain
- CIBERER, ISCIII, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Marfany
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine (IBUB-IRSJD), Universitat de Barcelona, Barcelona, Spain
- CIBERER, ISCIII, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Diñeiro M, Capín R, Cifuentes GÁ, Fernández‐Vega B, Villota E, Otero A, Santiago A, Pruneda PC, Castillo D, Viejo‐Díaz M, Hernando I, Durán NS, Álvarez R, Lago CG, Ordóñez GR, Fernández‐Vega Á, Cabanillas R, Cadiñanos J. Comprehensive genomic diagnosis of inherited retinal and optical nerve disorders reveals hidden syndromes and personalized therapeutic options. Acta Ophthalmol 2020; 98:e1034-e1048. [PMID: 32483926 PMCID: PMC7754416 DOI: 10.1111/aos.14479] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/29/2020] [Indexed: 01/14/2023]
Abstract
PURPOSE In the era of precision medicine, genomic characterization of blind patients is critical. Here, we evaluate the effects of comprehensive genetic analysis on the etiologic diagnosis of potentially hereditary vision loss and its impact on clinical management. METHODS We studied 100 non-syndromic and syndromic Spanish patients with a clinical diagnosis of blindness caused by alterations on the retina, choroid, vitreous and/or optic nerve. We used a next-generation sequencing (NGS) panel (OFTALMOgenics™), developed and validated within this study, including up to 362 genes previously associated with these conditions. RESULTS We identified the genetic cause of blindness in 45% of patients (45/100). A total of 28.9% of genetically diagnosed cases (13/45) were syndromic and, of those, in 30.8% (4/13) extraophthalmic features had been overlooked and/or not related to visual impairment before genetic testing, including cases with Mainzer-Saldino, Bardet-Biedl, mucolipidosis and MLCRD syndromes. In two additional cases-syndromic blindness had been proposed before, but not specifically diagnosed, and one patient with Heimler syndrome had been misdiagnosed as an Usher case before testing. 33.3% of the genetically diagnosed patients (15/45) had causative variants in genes targeted by clinical trials exploring the curative potential of gene therapy approaches. CONCLUSION Comprehensive genomic testing provided clinically relevant insights in a large proportion of blind patients, identifying potential therapeutic opportunities or previously undiagnosed syndromes in 42.2% of the genetically diagnosed cases (19/45).
Collapse
Affiliation(s)
- Marta Diñeiro
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.OviedoSpain
| | - Raquel Capín
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.OviedoSpain
| | | | | | - Eva Villota
- Instituto Oftalmológico Fernández‐Vega (IOFV)OviedoSpain
| | - Andrea Otero
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.OviedoSpain
| | - Adrián Santiago
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.OviedoSpain
| | | | - David Castillo
- Disease Research And Medicine (DREAMgenics) S. L.OviedoSpain
| | | | - Inés Hernando
- Hospital Universitario Central de AsturiasOviedoSpain
| | - Noelia S. Durán
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.OviedoSpain
| | - Rebeca Álvarez
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.OviedoSpain
| | - Claudia G. Lago
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.OviedoSpain
| | | | | | - Rubén Cabanillas
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.OviedoSpain
| | - Juan Cadiñanos
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.OviedoSpain
| |
Collapse
|
27
|
Vig A, Poulter JA, Ottaviani D, Tavares E, Toropova K, Tracewska AM, Mollica A, Kang J, Kehelwathugoda O, Paton T, Maynes JT, Wheway G, Arno G, Khan KN, McKibbin M, Toomes C, Ali M, Di Scipio M, Li S, Ellingford J, Black G, Webster A, Rydzanicz M, Stawiński P, Płoski R, Vincent A, Cheetham ME, Inglehearn CF, Roberts A, Heon E. DYNC2H1 hypomorphic or retina-predominant variants cause nonsyndromic retinal degeneration. Genet Med 2020; 22:2041-2051. [PMID: 32753734 PMCID: PMC7708302 DOI: 10.1038/s41436-020-0915-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Determining the role of DYNC2H1 variants in nonsyndromic inherited retinal disease (IRD). METHODS Genome and exome sequencing were performed for five unrelated cases of IRD with no identified variant. In vitro assays were developed to validate the variants identified (fibroblast assay, induced pluripotent stem cell [iPSC] derived retinal organoids, and a dynein motility assay). RESULTS Four novel DYNC2H1 variants (V1, g.103327020_103327021dup; V2, g.103055779A>T; V3, g.103112272C>G; V4, g.103070104A>C) and one previously reported variant (V5, g.103339363T>G) were identified. In proband 1 (V1/V2), V1 was predicted to introduce a premature termination codon (PTC), whereas V2 disrupted the exon 41 splice donor site causing incomplete skipping of exon 41. V1 and V2 impaired dynein-2 motility in vitro and perturbed IFT88 distribution within cilia. V3, homozygous in probands 2-4, is predicted to cause a PTC in a retina-predominant transcript. Analysis of retinal organoids showed that this new transcript expression increased with organoid differentiation. V4, a novel missense variant, was in trans with V5, previously associated with Jeune asphyxiating thoracic dystrophy (JATD). CONCLUSION The DYNC2H1 variants discussed herein were either hypomorphic or affecting a retina-predominant transcript and caused nonsyndromic IRD. Dynein variants, specifically DYNC2H1 variants are reported as a cause of non syndromic IRD.
Collapse
Affiliation(s)
- Anjali Vig
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, The University of Toronto, Toronto, Canada
| | - James A Poulter
- Department of Ophthalmology, St James' University Hospital, Leeds, UK
| | | | - Erika Tavares
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Katerina Toropova
- Department of Biological Sciences, Birbeck, University of London, London, UK
| | - Anna Maria Tracewska
- DNA Analysis Unit, ŁUKASIEWICZ Research Network-PORT Polish Center for Technology Development, Wrocław, Poland
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Antonio Mollica
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Jasmine Kang
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | | | - Tara Paton
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Jason T Maynes
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, Canada
| | | | - Gavin Arno
- UCL Institute of Ophthalmology, London, UK
- Moorfields Eye Hospital, London, UK
| | - Kamron N Khan
- Department of Ophthalmology, St James' University Hospital, Leeds, UK
| | - Martin McKibbin
- Department of Ophthalmology, St James' University Hospital, Leeds, UK
| | - Carmel Toomes
- Department of Ophthalmology, St James' University Hospital, Leeds, UK
| | - Manir Ali
- Department of Ophthalmology, St James' University Hospital, Leeds, UK
| | - Matteo Di Scipio
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Shuning Li
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Jamie Ellingford
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicines and health, The University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre (MAHSC), University of Manchester, Manchester, UK
| | - Graeme Black
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicines and health, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Andrew Webster
- UCL Institute of Ophthalmology, London, UK
- Moorfields Eye Hospital, London, UK
| | | | - Piotr Stawiński
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Ajoy Vincent
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, The University of Toronto, Toronto, Canada
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, Canada
| | | | | | - Anthony Roberts
- Department of Biological Sciences, Birbeck, University of London, London, UK.
| | - Elise Heon
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.
- Institute of Medical Science, The University of Toronto, Toronto, Canada.
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, Canada.
| |
Collapse
|
28
|
Review: Intraflagellar transport proteins in the retina. Mol Vis 2020; 26:652-660. [PMID: 33088169 PMCID: PMC7553723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 10/02/2020] [Indexed: 10/28/2022] Open
Abstract
Intraflagellar transport (IFT) is an essential process in all organisms that serves to move proteins along flagella or cilia in either direction. IFT is performed by IFT particles, which are multiprotein complexes organized into two subcomplexes, A and B. The IFT proteins form interactions with each other, with cargo proteins, and with membranes during the transport process. Several IFT proteins are expressed in many parts of the retina, such as the outer plexiform and outer nuclear layers, and function in the transport of photoreceptor proteins between the inner and outer segments. Mutants of IFT protein genes have been characterized in model organisms such as Chlamydomonas, C. elegans, zebrafish, and the mouse. These mutants have defective ciliogenesis or abnormalities in retinal photoreceptors. Mutations in IFT genes are associated with syndromic and non-syndromic forms of retinal disease in humans, frequently with early onset of disease.
Collapse
|
29
|
Tatour Y, Ben-Yosef T. Syndromic Inherited Retinal Diseases: Genetic, Clinical and Diagnostic Aspects. Diagnostics (Basel) 2020; 10:diagnostics10100779. [PMID: 33023209 PMCID: PMC7600643 DOI: 10.3390/diagnostics10100779] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
Inherited retinal diseases (IRDs), which are among the most common genetic diseases in humans, define a clinically and genetically heterogeneous group of disorders. Over 80 forms of syndromic IRDs have been described. Approximately 200 genes are associated with these syndromes. The majority of syndromic IRDs are recessively inherited and rare. Many, although not all, syndromic IRDs can be classified into one of two major disease groups: inborn errors of metabolism and ciliopathies. Besides the retina, the systems and organs most commonly involved in syndromic IRDs are the central nervous system, ophthalmic extra-retinal tissues, ear, skeleton, kidney and the cardiovascular system. Due to the high degree of phenotypic variability and phenotypic overlap found in syndromic IRDs, correct diagnosis based on phenotypic features alone may be challenging and sometimes misleading. Therefore, genetic testing has become the benchmark for the diagnosis and management of patients with these conditions, as it complements the clinical findings and facilitates an accurate clinical diagnosis and treatment.
Collapse
|
30
|
Yang XR, Benson MD, MacDonald IM, Innes AM. A diagnostic approach to syndromic retinal dystrophies with intellectual disability. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:538-570. [PMID: 32918368 DOI: 10.1002/ajmg.c.31834] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
Inherited retinal dystrophies are a group of monogenic disorders that, as a whole, contribute significantly to the burden of ocular disease in both pediatric and adult patients. In their syndromic forms, retinal dystrophies can be observed in association with intellectual disability, frequently alongside other systemic manifestations. There are now over 80 genes implicated in syndromic retinal dystrophies with intellectual disability. Identifying and accurately characterizing these disorders allows the clinician to narrow the differential diagnosis, evaluate for relevant associated features, arrive at a timely and accurate diagnosis, and address both sight-threatening ocular manifestations and morbidity-causing systemic manifestations. The co-occurrence of retinal dystrophy and intellectual disability in an individual can be challenging to investigate, diagnose, and counsel given the considerable phenotypic and genotypic heterogeneity that exists within this broad group of disorders. We performed a review of the current literature and propose an algorithm to facilitate the evaluation, and clinical and mechanistic classification, of these individuals.
Collapse
Affiliation(s)
- Xiao-Ru Yang
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Matthew D Benson
- Department of Ophthalmology, University of Alberta, Edmonton, Alberta, Canada
| | - Ian M MacDonald
- Department of Ophthalmology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - A Micheil Innes
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
31
|
Weisschuh N, Obermaier CD, Battke F, Bernd A, Kuehlewein L, Nasser F, Zobor D, Zrenner E, Weber E, Wissinger B, Biskup S, Stingl K, Kohl S. Genetic architecture of inherited retinal degeneration in Germany: A large cohort study from a single diagnostic center over a 9-year period. Hum Mutat 2020; 41:1514-1527. [PMID: 32531858 DOI: 10.1002/humu.24064] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/27/2020] [Accepted: 05/04/2020] [Indexed: 12/29/2022]
Abstract
We aimed to unravel the molecular genetic basis of inherited retinal degeneration (IRD) in a comprehensive cohort of patients diagnosed in the largest center for IRD in Germany. A cohort of 2,158 affected patients from 1,785 families diagnosed with IRD was analyzed by targeted next-generation sequencing (NGS). Patients with single-gene disorders (i.e., choroideremia and retinoschisis) were analyzed by Sanger sequencing and multiplex ligation-dependent probe amplification. Our study cohort accounts for ∼7% of the estimated 30,000 patients with IRD in Germany, thereby providing representative data for both the prevalence of IRDs and the mutation spectrum of IRD genes for the population in Germany. We achieved a molecular diagnostic rate of 35-95%, depending on the clinical entities, with a high detection rate for achromatopsia, retinoschisis, and choroideremia, and a low detection rate for central areolar choroidal dystrophy and macular dystrophy. A total of 1,161 distinct variants were identified, including 501 novel variants, reaffirming the known vast genetic heterogeneity of IRD in a mainly outbred European population. This study demonstrates the clinical utility of panel-based NGS in a large and highly heterogeneous cohort from an outbred population and for the first time gives a comprehensive representation of the genetic landscape of IRDs in Germany. The data are valuable and crucial for the scientific community and healthcare providers, but also for the pharmaceutical industry in the progressing field of personalized medicine and gene therapy.
Collapse
Affiliation(s)
- Nicole Weisschuh
- Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Carolin D Obermaier
- Praxis für Humangenetik, Tübingen, Germany.,Center for Genomics and Transcriptomics, CeGaT GmbH, Tübingen, Germany
| | - Florian Battke
- Center for Genomics and Transcriptomics, CeGaT GmbH, Tübingen, Germany
| | - Antje Bernd
- Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.,Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Laura Kuehlewein
- Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.,Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Fadi Nasser
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Ditta Zobor
- Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Eberhart Zrenner
- Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience (CIN), University of Tübingen, Tübingen, Germany
| | - Eva Weber
- Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Bernd Wissinger
- Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Saskia Biskup
- Praxis für Humangenetik, Tübingen, Germany.,Center for Genomics and Transcriptomics, CeGaT GmbH, Tübingen, Germany
| | - Katarina Stingl
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Susanne Kohl
- Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
32
|
Chen X, Wang X, Jiang C, Xu M, Liu Y, Qi R, Qi X, Sun X, Xie P, Liu Q, Yan B, Sheng X, Zhao C. IFT52 as a Novel Candidate for Ciliopathies Involving Retinal Degeneration. Invest Ophthalmol Vis Sci 2019; 59:4581-4589. [PMID: 30242358 DOI: 10.1167/iovs.17-23351] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Mutations in the intraflagellar transport protein 52 homolog (IFT52) gene are reported to interrupt ciliary function and cause short-rib thoracic dysplasia (SRTD), a specific form of skeletal ciliopathy. However, the roles of these mutations in retinal ciliopathy are inexplicit. We herein aim to study the impact of IFT52 mutations in retinopathies. Methods A patient with syndromic ciliopathy, presenting mild SRTD (skeletal ciliopathy) and Liber congenital amaurosis (LCA; retinal ciliopathy), and nine unaffected family members were recruited. Comprehensive systemic evaluations, including ophthalmic tests, were received by the patient. Whole genome sequencing (WGS) was applied for genetic annotation. An in vitro cell system was employed to study the pathogenicity of the variant. Results WGS identified a homozygous missense variation in IFT52, c.556A>G (p.T186A), carried by the patient but absent in both unaffected siblings. In silico analysis supported the pathogenic nature of this highly conserved variant. Structural analysis suggested that this substitution could generate a novel hydrogen bond between the mutated residue 186 and proline at residue 192, thus potentially interrupting the tertiary structure and the stability of the IFT52 protein. In vitro cellular study indicated that this mutation might disturb the stability of encoded IFT52 protein and dramatically disrupt cilia elongation in hTERT-RPE1 cells in a loss-of-function manner. Conclusions This report expands ocular phenotypes of IFT52 mutation-caused ciliopathy to include retinal ciliopathy and demonstrates its deleterious nature in interrupting primary ciliary function. Our study hence highlights the need for screening for IFT52 mutations in LCA patients and ophthalmic reviews of patients carrying IFT52 mutations.
Collapse
Affiliation(s)
- Xue Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, State Key Laboratory of Reproductive Medicine, Nanjing, China.,Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia of State Health Ministry (Fudan University) and Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiaoguang Wang
- Department of Ophthalmology, Ningxia Eye Hospital, People's Hospital of Ningxia Hui Autonomous Region (First Affiliated Hospital of Northwest University for Nationalities), Yinchuan, China
| | - Chao Jiang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, State Key Laboratory of Reproductive Medicine, Nanjing, China
| | - Min Xu
- Department of Ophthalmology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Yang Liu
- Department of Ophthalmology, Ningxia Eye Hospital, People's Hospital of Ningxia Hui Autonomous Region (First Affiliated Hospital of Northwest University for Nationalities), Yinchuan, China
| | - Rui Qi
- Medical College of Northwest Minzu University, Lanzhou, China
| | - Xiaolong Qi
- Department of Ophthalmology, Ningxia Eye Hospital, People's Hospital of Ningxia Hui Autonomous Region (First Affiliated Hospital of Northwest University for Nationalities), Yinchuan, China
| | - Xiantao Sun
- Department of Ophthalmology, Children's Hospital of Zhengzhou, Zhengzhou, China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, State Key Laboratory of Reproductive Medicine, Nanjing, China
| | - Qinghuai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, State Key Laboratory of Reproductive Medicine, Nanjing, China
| | - Biao Yan
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia of State Health Ministry (Fudan University) and Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xunlun Sheng
- Department of Ophthalmology, Ningxia Eye Hospital, People's Hospital of Ningxia Hui Autonomous Region (First Affiliated Hospital of Northwest University for Nationalities), Yinchuan, China
| | - Chen Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, State Key Laboratory of Reproductive Medicine, Nanjing, China.,Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia of State Health Ministry (Fudan University) and Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,Department of Ophthalmology, Children's Hospital of Zhengzhou, Zhengzhou, China
| |
Collapse
|
33
|
Photoreceptor cell development requires prostaglandin signaling in the zebrafish retina. Biochem Biophys Res Commun 2019; 510:230-235. [DOI: 10.1016/j.bbrc.2019.01.073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 01/16/2019] [Indexed: 01/02/2023]
|
34
|
Picariello T, Brown JM, Hou Y, Swank G, Cochran DA, King OD, Lechtreck K, Pazour GJ, Witman GB. A global analysis of IFT-A function reveals specialization for transport of membrane-associated proteins into cilia. J Cell Sci 2019; 132:jcs220749. [PMID: 30659111 PMCID: PMC6382014 DOI: 10.1242/jcs.220749] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 01/02/2019] [Indexed: 12/28/2022] Open
Abstract
Intraflagellar transport (IFT), which is essential for the formation and function of cilia in most organisms, is the trafficking of IFT trains (i.e. assemblies of IFT particles) that carry cargo within the cilium. Defects in IFT cause several human diseases. IFT trains contain the complexes IFT-A and IFT-B. To dissect the functions of these complexes, we studied a Chlamydomonas mutant that is null for the IFT-A protein IFT140. The mutation had no effect on IFT-B but destabilized IFT-A, preventing flagella assembly. Therefore, IFT-A assembly requires IFT140. Truncated IFT140, which lacks the N-terminal WD repeats of the protein, partially rescued IFT and supported formation of half-length flagella that contained normal levels of IFT-B but greatly reduced amounts of IFT-A. The axonemes of these flagella had normal ultrastructure and, as investigated by SDS-PAGE, normal composition. However, composition of the flagellar 'membrane+matrix' was abnormal. Analysis of the latter fraction by mass spectrometry revealed decreases in small GTPases, lipid-anchored proteins and cell signaling proteins. Thus, IFT-A is specialized for the import of membrane-associated proteins. Abnormal levels of the latter are likely to account for the multiple phenotypes of patients with defects in IFT140.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Tyler Picariello
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jason M Brown
- Department of Biology, Salem State University, Salem, MA 01970, USA
| | - Yuqing Hou
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Gregory Swank
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Deborah A Cochran
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Oliver D King
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Karl Lechtreck
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - George B Witman
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
35
|
Chekuri A, Guru AA, Biswas P, Branham K, Borooah S, Soto-Hermida A, Hicks M, Khan NW, Matsui H, Alapati A, Raghavendra PB, Roosing S, Sarangapani S, Mathavan S, Telenti A, Heckenlively JR, Riazuddin SA, Frazer KA, Sieving PA, Ayyagari R. IFT88 mutations identified in individuals with non-syndromic recessive retinal degeneration result in abnormal ciliogenesis. Hum Genet 2018; 137:447-458. [PMID: 29978320 DOI: 10.1007/s00439-018-1897-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/21/2018] [Indexed: 12/26/2022]
Abstract
Whole genome sequencing (WGS) was performed to identify the variants responsible for inherited retinal degeneration (IRD) in a Caucasian family. Segregation analysis of selected rare variants with pathogenic potential identified a set of compound heterozygous changes p.Arg266*:c.796C>T and p.Ala568Thr:c.1702G>A in the intraflagellar transport protein-88 (IFT88) gene segregating with IRD. Expression of IFT88 with the p.Arg266* and p.Ala568Thr mutations in mIMDC3 cells by transient transfection and in HeLa cells by introducing the mutations using CRISPR-cas9 system suggested that both mutations result in the formation of abnormal ciliary structures. The introduction of the IFT88 p.Arg266* variant in the homozygous state in HeLa cells by CRISPR-Cas9 genome-editing revealed that the mutant transcript undergoes nonsense-mediated decay leading to a significant depletion of IFT88 transcript. Additionally, abnormal ciliogenesis was observed in these cells. These observations suggest that the rare and unique combination of IFT88 alleles observed in this study provide insight into the physiological role of IFT88 in humans and the likely mechanism underlying retinal pathology in the pedigree with IRD.
Collapse
Affiliation(s)
- Anil Chekuri
- Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, JRC 206, La Jolla, CA, 92093, USA
| | - Aditya A Guru
- Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, JRC 206, La Jolla, CA, 92093, USA
| | - Pooja Biswas
- Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, JRC 206, La Jolla, CA, 92093, USA.,School of Biotechnology, REVA University, Bengaluru, Karnataka, India
| | - Kari Branham
- Ophthalmology and Visual Science, University of Michigan Kellogg Eye Center, Ann Arbor, MI, USA
| | - Shyamanga Borooah
- Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, JRC 206, La Jolla, CA, 92093, USA
| | - Angel Soto-Hermida
- Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, JRC 206, La Jolla, CA, 92093, USA
| | | | - Naheed W Khan
- Ophthalmology and Visual Science, University of Michigan Kellogg Eye Center, Ann Arbor, MI, USA
| | - Hiroko Matsui
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Akhila Alapati
- Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, JRC 206, La Jolla, CA, 92093, USA
| | - Pongali B Raghavendra
- School of Biotechnology, REVA University, Bengaluru, Karnataka, India.,School of Regenerative Medicine, Manipal University-MAHE, Bangalore, India
| | - Susanne Roosing
- Department of Human Genetics, Radboud University Nijmegen Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | | - John R Heckenlively
- Ophthalmology and Visual Science, University of Michigan Kellogg Eye Center, Ann Arbor, MI, USA
| | - S Amer Riazuddin
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kelly A Frazer
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.,Division of Genome Information Sciences, Department of Pediatrics, Rady Children's Hospital, San Diego, CA, USA
| | - Paul A Sieving
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Radha Ayyagari
- Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, JRC 206, La Jolla, CA, 92093, USA.
| |
Collapse
|
36
|
Liu H, Li W, Zhang Y, Zhang Z, Shang X, Zhang L, Zhang S, Li Y, Somoza AV, Delpi B, Gerton GL, Foster JA, Hess RA, Pazour GJ, Zhang Z. IFT25, an intraflagellar transporter protein dispensable for ciliogenesis in somatic cells, is essential for sperm flagella formation. Biol Reprod 2018; 96:993-1006. [PMID: 28430876 DOI: 10.1093/biolre/iox029] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 04/13/2017] [Indexed: 12/25/2022] Open
Abstract
Intraflagellar transport (IFT) is a conserved mechanism essential for the assembly and maintenance of most eukaryotic cilia and flagella. However, IFT25, a component of the IFT complex, is not required for the formation of cilia in somatic tissues. In mice, the gene is highly expressed in the testis, and its expression is upregulated during the final phase when sperm flagella are formed. To investigate the role of IFT25 in sperm flagella formation, the gene was specifically disrupted in male germ cells. All homozygous knockout mice survived to adulthood and did not show any gross abnormalities. However, all homozygous knockout males were completely infertile. Sperm numbers were reduced and these sperm were completely immotile. Multiple morphological abnormalities were observed in sperm, including round heads, short and bent tails, with some tails showing branched flagella and others with frequent abnormal thicknesses, as well as swollen tips of the tail. Transmission electron microscopy revealed that flagellar accessory structures, including the fibrous sheath and outer dense fibers, were disorganized, and most sperm had also lost the "9+2" microtubule structure. In the testis, IFT25 forms a complex with other IFT proteins. In Ift25 knockout testes, IFT27, an IFT25 binding partner, was missing, and IFT20 and IFT81 levels were also reduced. Our findings suggest that IFT25, although not necessary for the formation of cilia in somatic cells, is indispensable for sperm flagellum formation and male fertility in mice.
Collapse
Affiliation(s)
- Hong Liu
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Wei Li
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yong Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Dermatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengang Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuejun Shang
- Department of Andrology, Jinling Hospital, Nanjing University, School of Medicine, Nanjing, China
| | - Ling Zhang
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Shiyang Zhang
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yanwei Li
- Department of Computer Science, Wellesley College, Wellesley, Massachusetts, USA
| | - Andres V Somoza
- Department of Humanities and Sciences, Honor College, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Brandon Delpi
- Department of Biology, Randolph-Macon College, Ashland, Virginia, USA
| | - George L Gerton
- Center for Research on Reproduction and Women's Health Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James A Foster
- Department of Biology, Randolph-Macon College, Ashland, Virginia, USA
| | - Rex A Hess
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, Illinois, USA
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Zhibing Zhang
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
37
|
Molecular Diagnosis of 34 Japanese Families with Leber Congenital Amaurosis Using Targeted Next Generation Sequencing. Sci Rep 2018; 8:8279. [PMID: 29844330 PMCID: PMC5974356 DOI: 10.1038/s41598-018-26524-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Leber congenital amaurosis (LCA) is a genetically and clinically heterogeneous disease, and represents the most severe form of inherited retinal dystrophy (IRD). The present study reports the mutation spectra and frequency of known LCA and IRD-associated genes in 34 Japanese families with LCA (including three families that were previously reported). A total of 74 LCA- and IRD-associated genes were analysed via targeted-next generation sequencing (TS), while recently discovered LCA-associated genes, as well as known variants not able to be screened using this approach, were evaluated via additional Sanger sequencing, long-range polymerase chain reaction, and/or copy number variation analyses. The results of these analyses revealed 30 potential pathogenic variants in 12 (nine LCA-associated and three other IRD-associated) genes among 19 of the 34 analysed families. The most frequently mutated genes were CRB1, NMNAT1, and RPGRIP1. The results also showed the mutation spectra and frequencies identified in the analysed Japanese population to be distinctly different from those previously identified for other ethnic backgrounds. Finally, the present study, which is the first to conduct a NGS-based molecular diagnosis of a large Japanese LCA cohort, achieved a detection rate of approximately 56%, indicating that TS is a valuable method for molecular diagnosis of LCA cases in the Japanese population.
Collapse
|
38
|
Zhang Y, Liu H, Li W, Zhang Z, Zhang S, Teves ME, Stevens C, Foster JA, Campbell GE, Windle JJ, Hess RA, Pazour GJ, Zhang Z. Intraflagellar transporter protein 140 (IFT140), a component of IFT-A complex, is essential for male fertility and spermiogenesis in mice. Cytoskeleton (Hoboken) 2018; 75:70-84. [PMID: 29236364 DOI: 10.1002/cm.21427] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 11/11/2022]
Abstract
Intraflagellar transport (IFT) is a conserved mechanism essential for the assembly and maintenance of most eukaryotic cilia and flagella. However, little is known about its role in sperm flagella formation and male fertility. IFT140 is a component of IFT-A complex. In mouse, it is highly expressed in the testis. Ift140 gene was inactivated specifically in mouse spermatocytes/spermatids. The mutant mice did not show any gross abnormalities, but all were infertile and associated with significantly reduced sperm number and motility. Multiple sperm morphological abnormalities were discovered, including amorphous heads, short/bent flagella and swollen tail tips, as well as vesicles along the flagella due to spermiogenesis defects. The epididymides contained round bodies of cytoplasm derived from the sloughing of the cytoplasmic lobes and residual bodies. Knockout of Ift140 did not significantly affect testicular expression levels of selective IFT components but localization of IFT27 and IFT88, two components of IFT-B complex, was changed. Our findings demonstrate that IFT140 is a key regulator for male fertility and normal spermiogenesis in mice. It not only plays a role in sperm flagella assembling, but is also involved in critical assembly of proteins that interface between the germ cell plasma and the Sertoli cell.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Hong Liu
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298.,School of Public Health, Wuhan University of Science and Technology, Wuhan 430060, Hubei
| | - Wei Li
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Zhengang Zhang
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298.,Department of Gastroenterology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei
| | - Shiyang Zhang
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298.,School of Public Health, Wuhan University of Science and Technology, Wuhan 430060, Hubei
| | - Maria E Teves
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Courtney Stevens
- Department of Biology, Randolph-Macon College, Ashland, Virginia 23005
| | - James A Foster
- Department of Biology, Randolph-Macon College, Ashland, Virginia 23005
| | - Gregory E Campbell
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Jolene J Windle
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, 2001 S. Lincoln, Urbana, Illinois 61802-6199
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Zhibing Zhang
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298.,School of Public Health, Wuhan University of Science and Technology, Wuhan 430060, Hubei
| |
Collapse
|
39
|
DuPont M, Jones EM, Xu M, Chen R. Investigating the disease association of USH2A p.C759F variant by leveraging large retinitis pigmentosa cohort data. Ophthalmic Genet 2017; 39:291-292. [PMID: 29283788 DOI: 10.1080/13816810.2017.1418388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Mariana DuPont
- a College of Arts and Sciences , Dillard University , New Orleans , LA , USA
| | - Evan M Jones
- b Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,c Human Genome Sequencing Center , Baylor College of Medicine , Houston , TX , USA
| | - Mingchu Xu
- b Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,c Human Genome Sequencing Center , Baylor College of Medicine , Houston , TX , USA
| | - Rui Chen
- b Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,c Human Genome Sequencing Center , Baylor College of Medicine , Houston , TX , USA
| |
Collapse
|
40
|
Low T, Kostakis A, Balasubramanian M. Compound heterozygous variants in IFT140 as a cause of nonsyndromic retinitis pigmentosa. Ophthalmic Genet 2017; 39:286-287. [DOI: 10.1080/13816810.2017.1393827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Tisiana Low
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | | - Meena Balasubramanian
- Sheffield Clinical Genetics Service, Sheffield Children’s NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
41
|
Bujakowska KM, Liu Q, Pierce EA. Photoreceptor Cilia and Retinal Ciliopathies. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028274. [PMID: 28289063 DOI: 10.1101/cshperspect.a028274] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Photoreceptors are sensory neurons designed to convert light stimuli into neurological responses. This process, called phototransduction, takes place in the outer segments (OS) of rod and cone photoreceptors. OS are specialized sensory cilia, with analogous structures to those present in other nonmotile cilia. Deficient morphogenesis and/or dysfunction of photoreceptor sensory cilia (PSC) caused by mutations in a variety of photoreceptor-specific and common cilia genes can lead to inherited retinal degenerations (IRDs). IRDs can manifest as isolated retinal diseases or syndromic diseases. In this review, we describe the structure and composition of PSC and different forms of ciliopathies with retinal involvement. We review the genetics of the IRDs, which are monogenic disorders but genetically diverse with regard to causality.
Collapse
Affiliation(s)
- Kinga M Bujakowska
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | - Qin Liu
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | - Eric A Pierce
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
42
|
Ku CA, Hull S, Arno G, Vincent A, Carss K, Kayton R, Weeks D, Anderson GW, Geraets R, Parker C, Pearce DA, Michaelides M, MacLaren RE, Robson AG, Holder GE, Heon E, Raymond FL, Moore AT, Webster AR, Pennesi ME. Detailed Clinical Phenotype and Molecular Genetic Findings in CLN3-Associated Isolated Retinal Degeneration. JAMA Ophthalmol 2017; 135:749-760. [PMID: 28542676 DOI: 10.1001/jamaophthalmol.2017.1401] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Mutations in genes traditionally associated with syndromic retinal disease are increasingly found to cause nonsyndromic inherited retinal degenerations. Mutations in CLN3 are classically associated with juvenile neuronal ceroid lipofuscinosis, a rare neurodegenerative disease with early retinal degeneration and progressive neurologic deterioration, but have recently also been identified in patients with nonsyndromic inherited retinal degenerations. To our knowledge, detailed clinical characterization of such cases has yet to be reported. Objective To provide detailed clinical, electrophysiologic, structural, and molecular genetic findings in nonsyndromic inherited retinal degenerations associated with CLN3 mutations. Design, Setting, and Participants A multi-institutional case series of 10 patients who presented with isolated nonsyndromic retinal disease and mutations in CLN3. Patient ages ranged from 16 to 70 years; duration of follow-up ranged from 3 to 29 years. Main Outcomes and Measures Longitudinal clinical evaluation, including full ophthalmic examination, multimodal retinal imaging, perimetry, and electrophysiology. Molecular analyses were performed using whole-genome sequencing or whole-exome sequencing. Electron microscopy studies of peripheral lymphocytes and CLN3 transcript analysis with polymerase chain reaction amplification were performed in a subset of patients. Results There were 7 females and 3 males in this case series, with a mean (range) age at last review of 37.1 (16-70) years. Of the 10 patients, 4 had a progressive late-onset rod-cone dystrophy, with a mean (range) age at onset of 29.7 (20-40) years, and 6 had an earlier onset rod-cone dystrophy, with a mean (range) age at onset of 12.1 (7-17) years. Ophthalmoscopic examination features included macular edema, mild intraretinal pigment migration, and widespread atrophy in advanced disease. Optical coherence tomography imaging demonstrated significant photoreceptor loss except in patients with late-onset disease who had a focal preservation of the ellipsoid zone and outer nuclear layer in the fovea. Electroretinography revealed a rod-cone pattern of dysfunction in 6 patients and were completely undetectable in 2 patients. Six novel CLN3 variants were identified in molecular analyses. Conclusions and Relevance This report describes detailed clinical, imaging, and genetic features of CLN3-associated nonsyndromic retinal degeneration. The age at onset and natural progression of retinal disease differs greatly between syndromic and nonsyndromic CLN3 disease, which may be associated with genotypic differences.
Collapse
Affiliation(s)
- Cristy A Ku
- Casey Eye Institute, Oregon Health & Science University, Portland
| | - Sarah Hull
- University College London Institute of Ophthalmology, London, England3Moorfields Eye Hospital, London, England
| | - Gavin Arno
- University College London Institute of Ophthalmology, London, England3Moorfields Eye Hospital, London, England
| | - Ajoy Vincent
- Department of Ophthalmology and Vision Sciences, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Keren Carss
- National Health Service Blood and Transplant Centre, Department of Haematology, University of Cambridge, Cambridge, England6National Institute for Health Research BioResource: Rare Diseases, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, England
| | - Robert Kayton
- Pathology Department, Oregon Health & Science University, Portland
| | - Douglas Weeks
- Pathology Department, Oregon Health & Science University, Portland
| | - Glenn W Anderson
- Histopathology Department, Great Ormond Street Hospital for Children, London, England
| | - Ryan Geraets
- Sanford Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota
| | - Camille Parker
- Sanford Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota
| | - David A Pearce
- Sanford Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota10Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls
| | - Michel Michaelides
- University College London Institute of Ophthalmology, London, England3Moorfields Eye Hospital, London, England
| | - Robert E MacLaren
- Moorfields Eye Hospital, London, England11Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, England12Oxford University Hospitals National Health Service Foundation Trust, Oxford, England
| | - Anthony G Robson
- University College London Institute of Ophthalmology, London, England3Moorfields Eye Hospital, London, England
| | - Graham E Holder
- University College London Institute of Ophthalmology, London, England3Moorfields Eye Hospital, London, England
| | - Elise Heon
- Department of Ophthalmology and Vision Sciences, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - F Lucy Raymond
- National Health Service Blood and Transplant Centre, Department of Haematology, University of Cambridge, Cambridge, England6National Institute for Health Research BioResource: Rare Diseases, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, England13Cambridge Institute for Medical Research, Department of Medical Genetics, University of Cambridge, Cambridge, England
| | - Anthony T Moore
- University College London Institute of Ophthalmology, London, England3Moorfields Eye Hospital, London, England14Department of Ophthalmology, University of California, San Francisco Medical School, San Francisco
| | - Andrew R Webster
- University College London Institute of Ophthalmology, London, England3Moorfields Eye Hospital, London, England
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health & Science University, Portland
| |
Collapse
|
43
|
Genetic characterization and disease mechanism of retinitis pigmentosa; current scenario. 3 Biotech 2017; 7:251. [PMID: 28721681 DOI: 10.1007/s13205-017-0878-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 07/10/2017] [Indexed: 12/21/2022] Open
Abstract
Retinitis pigmentosa is a group of genetically transmitted disorders affecting 1 in 3000-8000 individual people worldwide ultimately affecting the quality of life. Retinitis pigmentosa is characterized as a heterogeneous genetic disorder which leads by progressive devolution of the retina leading to a progressive visual loss. It can occur in syndromic (with Usher syndrome and Bardet-Biedl syndrome) as well as non-syndromic nature. The mode of inheritance can be X-linked, autosomal dominant or autosomal recessive manner. To date 58 genes have been reported to associate with retinitis pigmentosa most of them are either expressed in photoreceptors or the retinal pigment epithelium. This review focuses on the disease mechanisms and genetics of retinitis pigmentosa. As retinitis pigmentosa is tremendously heterogeneous disorder expressing a multiplicity of mutations; different variations in the same gene might induce different disorders. In recent years, latest technologies including whole-exome sequencing contributing effectively to uncover the hidden genesis of retinitis pigmentosa by reporting new genetic mutations. In future, these advancements will help in better understanding the genotype-phenotype correlations of disease and likely to develop new therapies.
Collapse
|
44
|
The phenotypic variability of HK1-associated retinal dystrophy. Sci Rep 2017; 7:7051. [PMID: 28765615 PMCID: PMC5539152 DOI: 10.1038/s41598-017-07629-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/28/2017] [Indexed: 12/05/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a clinically and genetically heterogeneous group of Mendelian disorders primarily affecting photoreceptor cells. The same IRD-causing variant may lead to different retinal symptoms, demonstrating pleiotropic phenotype traits influenced by both underlying genetic and environmental factors. In the present study, we identified four unrelated IRD families with the HK1 p.E851K variant, which was previously reported to cause autosomal dominant retinitis pigmentosa (RP), and described their detailed clinical phenotypes. Interestingly, we found that in addition to RP, this particular variant can also cause dominant macular dystrophy and cone-rod dystrophy, which primarily affect cone photoreceptors instead of rods. Our results identified pleiotropic effects for an IRD-causing variant and provide more insights into the involvement of a hexokinase in retinal pathogenesis.
Collapse
|
45
|
Dharmat R, Liu W, Ge Z, Sun Z, Yang L, Li Y, Wang K, Thomas K, Sui R, Chen R. IFT81 as a Candidate Gene for Nonsyndromic Retinal Degeneration. Invest Ophthalmol Vis Sci 2017; 58:2483-2490. [PMID: 28460050 PMCID: PMC5413215 DOI: 10.1167/iovs.16-19133] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Purpose IFT81, a core component of the IFT-B complex, involved in the bidirectional transport of ciliary proteins, has been recently implicated in syndromic ciliopathies. However, none of the IFT-B core complex proteins have been associated with nonsyndromic retinal dystrophies. Given the importance of ciliary transport in photoreceptor function and structural maintenance, we sought to investigate the impact of IFT (intraflagellar transport) mutations in nonsyndromic retinopathies. Methods Whole exome sequencing was performed on 50 cone-rod dystrophy (CRD) patients that were previously screened for mutations in known retinal disease genes. The impact of candidate mutation was studied using in vitro cell system and in vivo zebrafish assay to determine the pathogenicity of the variant. Results Compound heterozygous mutations in IFT81, including one nonsense (c.1213C>T, p.R405*) and one missense variant (c.1841T>C, p.L614P), were identified in a nonsyndromic CRD proband. Extensive functional analyses of the missense variant in cell culture and zebrafish strongly suggests its pathogenic nature. Loss of IFT81 impairs ciliogenesis and, interestingly, the missense variant displayed significantly reduced rescue of ciliogenesis in the IFT81 knockdown in vitro system. Consistently, dramatic reduction of rescue efficiency of the ift81 mutant zebrafish embryo by mRNA with the missense variant was observed, further supporting its pathogenicity. Conclusions Consistent with the function of the IFT-B complex in the maintenance of photoreceptor cilium, we report a case of mutations in a core IFT-B protein, IFT81. This represents the first report of mutations in IFT81 as a candidate gene for nonsyndromic retinal dystrophy, hence expanding the phenotype spectrum of IFT-B components.
Collapse
Affiliation(s)
- Rachayata Dharmat
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States 2Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Wei Liu
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Zhongqi Ge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States 2Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Zixi Sun
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lizhu Yang
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States 2Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Keqing Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States 2Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Kandace Thomas
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Ruifang Sui
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States 2Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States 4Department of Structural and Computational Biology & Molecular Biophysics, Baylor College of Medicine, Houston, Texas, United States 5Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States 6Program of Developmental Biology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
46
|
Helm BM, Willer JR, Sadeghpour A, Golzio C, Crouch E, Vergano SS, Katsanis N, Davis EE. Partial uniparental isodisomy of chromosome 16 unmasks a deleterious biallelic mutation in IFT140 that causes Mainzer-Saldino syndrome. Hum Genomics 2017; 11:16. [PMID: 28724397 PMCID: PMC5517791 DOI: 10.1186/s40246-017-0111-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/29/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The ciliopathies represent an umbrella group of >50 clinical entities that share both clinical features and molecular etiology underscored by structural and functional defects of the primary cilium. Despite the advances in gene discovery, this group of entities continues to pose a diagnostic challenge, in part due to significant genetic and phenotypic heterogeneity and variability. We consulted a pediatric case from asymptomatic, non-consanguineous parents who presented as a suspected ciliopathy due to a constellation of retinal, renal, and skeletal findings. RESULTS Although clinical panel sequencing of genes implicated in nephrotic syndromes yielded no likely causal mutation, an oligo-SNP microarray identified a ~20-Mb region of homozygosity, with no altered gene dosage, on chromosome 16p13. Intersection of the proband's phenotypes with known disease genes within the homozygous region yielded a single candidate, IFT140, encoding a retrograde intraflagellar transport protein implicated previously in several ciliopathies, including the phenotypically overlapping Mainzer-Saldino syndrome (MZSDS). Sanger sequencing yielded a maternally inherited homozygous c.634G>A; p.Gly212Arg mutation altering the exon 6 splice donor site. Functional studies in cells from the proband showed that the locus produced two transcripts: a majority message containing a mis-splicing event that caused a premature termination codon and a minority message homozygous for the p.Gly212Arg allele. Zebrafish in vivo complementation studies of the latter transcript demonstrated a loss of function effect. Finally, we conducted post-hoc trio-based whole exome sequencing studies to (a) test the possibility of other causal loci in the proband and (b) explain the Mendelian error of segregation for the IFT140 mutation. We show that the proband harbors a chromosome 16 maternal heterodisomy, with segmental isodisomy at 16p13, likely due to a meiosis I error in the maternal gamete. CONCLUSIONS Using clinical phenotyping combined with research-based genetic and functional studies, we have characterized a recurrent IFT140 mutation in the proband; together, these data are consistent with MZSDS. Additionally, we report a rare instance of a uniparental isodisomy unmasking a deleterious mutation to cause a ciliary disorder.
Collapse
Affiliation(s)
- Benjamin M Helm
- Division of Medical Genetics and Metabolism, Children's Hospital of The King's Daughters, Norfolk, VA, 23507, USA.,Department of Medical & Molecular Genetics, Riley Hospital for Children at IU Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jason R Willer
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, 27701, USA
| | - Azita Sadeghpour
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, 27701, USA
| | - Christelle Golzio
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, 27701, USA.,Institute of Genetics and Molecular and Cellular Biology, 67404, Illkirch, France
| | - Eric Crouch
- Department of Ophthalmology, Children's Hospital of the King's Daughters, Norfolk, VA, 23507, USA
| | - Samantha Schrier Vergano
- Division of Medical Genetics and Metabolism, Children's Hospital of The King's Daughters, Norfolk, VA, 23507, USA.,Department of Pediatrics, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, 27701, USA.
| | - Erica E Davis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, 27701, USA.
| |
Collapse
|
47
|
Lessieur EM, Fogerty J, Gaivin RJ, Song P, Perkins BD. The Ciliopathy Gene ahi1 Is Required for Zebrafish Cone Photoreceptor Outer Segment Morphogenesis and Survival. Invest Ophthalmol Vis Sci 2017; 58:448-460. [PMID: 28118669 PMCID: PMC5270624 DOI: 10.1167/iovs.16-20326] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Purpose Joubert syndrome (JBTS) is an autosomal recessive ciliopathy with considerable phenotypic variability. In addition to central nervous system abnormalities, a subset of JBTS patients exhibit retinal dystrophy and/or kidney disease. Mutations in the AHI1 gene are causative for approximately 10% of all JBTS cases. The purpose of this study was to generate ahi1 mutant alleles in zebrafish and to characterize the retinal phenotypes. Methods Zebrafish ahi1 mutants were generated using transcription activator-like effector nucleases (TALENs). Expression analysis was performed by whole-mount in situ hybridization. Anatomic and molecular characterization of photoreceptors was investigated by histology, electron microscopy, and immunohistochemistry. The optokinetic response (OKR) behavior assay was used to assess visual function. Kidney cilia were evaluated by whole-mount immunostaining. Results The ahi1lri46 mutation in zebrafish resulted in shorter cone outer segments but did not affect visual behavior at 5 days after fertilization (dpf). No defects in rod morphology or rhodopsin localization were observed at 5 dpf. By 5 months of age, cone degeneration and rhodopsin mislocalization in rod photoreceptors was observed. The connecting cilium formed normally and Cc2d2a and Cep290 localized properly. Distal pronephric duct cilia were absent in mutant fish; however, only 9% of ahi1 mutants had kidney cysts by 5 dpf, suggesting that the pronephros remained largely functional. Conclusions The results indicate that Ahi1 is required for photoreceptor disc morphogenesis and outer segment maintenance in zebrafish.
Collapse
Affiliation(s)
- Emma M Lessieur
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States 2Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, United States
| | - Joseph Fogerty
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Robert J Gaivin
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Ping Song
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Brian D Perkins
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States 2Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
48
|
Broadgate S, Yu J, Downes SM, Halford S. Unravelling the genetics of inherited retinal dystrophies: Past, present and future. Prog Retin Eye Res 2017; 59:53-96. [PMID: 28363849 DOI: 10.1016/j.preteyeres.2017.03.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 02/07/2023]
Abstract
The identification of the genes underlying monogenic diseases has been of interest to clinicians and scientists for many years. Using inherited retinal dystrophies as an example of monogenic disease we describe the history of molecular genetic techniques that have been pivotal in the discovery of disease causing genes. The methods that were developed in the 1970's and 80's are still in use today but have been refined and improved. These techniques enabled the concept of the Human Genome Project to be envisaged and ultimately realised. When the successful conclusion of the project was announced in 2003 many new tools and, as importantly, many collaborations had been developed that facilitated a rapid identification of disease genes. In the post-human genome project era advances in computing power and the clever use of the properties of DNA replication has allowed the development of next-generation sequencing technologies. These methods have revolutionised the identification of disease genes because for the first time there is no need to define the position of the gene in the genome. The use of next generation sequencing in a diagnostic setting has allowed many more patients with an inherited retinal dystrophy to obtain a molecular diagnosis for their disease. The identification of novel genes that have a role in the development or maintenance of retinal function is opening up avenues of research which will lead to the development of new pharmacological and gene therapy approaches. Neither of which can be used unless the defective gene and protein is known. The continued development of sequencing technologies also holds great promise for the advent of truly personalised medicine.
Collapse
Affiliation(s)
- Suzanne Broadgate
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Levels 5 and 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| | - Jing Yu
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Levels 5 and 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| | - Susan M Downes
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, OX3 9DU, UK
| | - Stephanie Halford
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Levels 5 and 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK.
| |
Collapse
|
49
|
Coussa RG, Lopez Solache I, Koenekoop RK. Leber congenital amaurosis, from darkness to light: An ode to Irene Maumenee. Ophthalmic Genet 2017; 38:7-15. [PMID: 28095138 DOI: 10.1080/13816810.2016.1275021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This article is dedicated to Irene Hussels Maumenee, Professor of Human Genetics and Ophthalmology, Johns Hopkins' Wilmer Eye Institute, Ocular Genetics Fellowship director in 1994-1995. Leber congenital amaurosis (LCA) has almost come full circle, from a profound and molecularly uncharacterized form of congenital retinal blindness to one in which a large number of causative genes and disease pathways are known, and the world's first human retinal disease to be treated by gene therapy. Dr. Maumenee's insights, efforts, and leadership have contributed significantly to this remarkable scientific journey. In this manuscript, we present a short summary of the known LCA genes, LCA disease subtypes, and emerging treatment options. Our manuscript consolidates previous knowledge with current findings in an attempt to provide a more comprehensive understanding of LCA.
Collapse
Affiliation(s)
- Razek Georges Coussa
- a Department of Paediatric Surgery, Montreal Children's Hospital , McGill University Health Centre , Montreal , Quebec , Canada.,b The McGill Ocular Genetics Laboratory, Paediatric Ophthalmology Division , Montreal Children's Hospital, McGill University Health Centre , Montreal , Quebec , Canada
| | - Irma Lopez Solache
- b The McGill Ocular Genetics Laboratory, Paediatric Ophthalmology Division , Montreal Children's Hospital, McGill University Health Centre , Montreal , Quebec , Canada
| | - Robert K Koenekoop
- a Department of Paediatric Surgery, Montreal Children's Hospital , McGill University Health Centre , Montreal , Quebec , Canada.,b The McGill Ocular Genetics Laboratory, Paediatric Ophthalmology Division , Montreal Children's Hospital, McGill University Health Centre , Montreal , Quebec , Canada
| |
Collapse
|
50
|
Abstract
Leber congenital amaurosis (LCA) is a group of severe inherited retinal dystrophies that lead to early childhood blindness. In the last decade, interest in LCA has increased as advances in genetics have been applied to better identify, classify, and treat LCA. To date, 23 LCA genes have been identified. Gene replacement in the RPE65 form of LCA represents a major advance in treatment, although limitations have been recognized. In this article, we review the clinical and genetic features of LCA and evaluate the evidence available for gene therapy in RPE65 disease.
Collapse
Affiliation(s)
- Maan Alkharashi
- a Boston Children's Hospital, Harvard Medical School , Boston , MA , USA.,b Department of Ophthalmology , King Saud University , Riyadh , Saudi Arabia
| | - Anne B Fulton
- b Department of Ophthalmology , King Saud University , Riyadh , Saudi Arabia
| |
Collapse
|