1
|
Wang Z, Xia B, Qi S, Zhang X, Zhang X, Li Y, Wang H, Zhang M, Zhao Z, Kerr D, Yang L, Cai S, Yang J. Bestrophin-4 relays HES4 and interacts with TWIST1 to suppress epithelial-to-mesenchymal transition in colorectal cancer cells. eLife 2024; 12:RP88879. [PMID: 39699952 DOI: 10.7554/elife.88879] [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] [Indexed: 12/20/2024] Open
Abstract
Bestrophin isoform 4 (BEST4) is a newly identified subtype of the calcium-activated chloride channel family. Analysis of colonic epithelial cell diversity by single-cell RNA-sequencing has revealed the existence of a cluster of BEST4+ mature colonocytes in humans. However, if the role of BEST4 is involved in regulating tumour progression remains largely unknown. In this study, we demonstrate that BEST4 overexpression attenuates cell proliferation, colony formation, and mobility in colorectal cancer (CRC) in vitro, and impedes the tumour growth and the liver metastasis in vivo. BEST4 is co-expressed with hairy/enhancer of split 4 (HES4) in the nucleus of cells, and HES4 signals BEST4 by interacting with the upstream region of the BEST4 promoter. BEST4 is epistatic to HES4 and downregulates TWIST1, thereby inhibiting epithelial-to-mesenchymal transition (EMT) in CRC. Conversely, knockout of BEST4 using CRISPR/Cas9 in CRC cells revitalises tumour growth and induces EMT. Furthermore, the low level of the BEST4 mRNA is correlated with advanced and the worse prognosis, suggesting its potential role involving CRC progression.
Collapse
Affiliation(s)
- Zijing Wang
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Bihan Xia
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Shaochong Qi
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Xian Zhang
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoshuang Zhang
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Li
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Huimin Wang
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Miao Zhang
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- College of Acupuncture and Moxibustion, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Ziyi Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - David Kerr
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Li Yang
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Shijie Cai
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jilin Yang
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Georgiou M, Robson AG, Fujinami K, de Guimarães TAC, Fujinami-Yokokawa Y, Daich Varela M, Pontikos N, Kalitzeos A, Mahroo OA, Webster AR, Michaelides M. Phenotyping and genotyping inherited retinal diseases: Molecular genetics, clinical and imaging features, and therapeutics of macular dystrophies, cone and cone-rod dystrophies, rod-cone dystrophies, Leber congenital amaurosis, and cone dysfunction syndromes. Prog Retin Eye Res 2024; 100:101244. [PMID: 38278208 DOI: 10.1016/j.preteyeres.2024.101244] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
Inherited retinal diseases (IRD) are a leading cause of blindness in the working age population and in children. The scope of this review is to familiarise clinicians and scientists with the current landscape of molecular genetics, clinical phenotype, retinal imaging and therapeutic prospects/completed trials in IRD. Herein we present in a comprehensive and concise manner: (i) macular dystrophies (Stargardt disease (ABCA4), X-linked retinoschisis (RS1), Best disease (BEST1), PRPH2-associated pattern dystrophy, Sorsby fundus dystrophy (TIMP3), and autosomal dominant drusen (EFEMP1)), (ii) cone and cone-rod dystrophies (GUCA1A, PRPH2, ABCA4, KCNV2 and RPGR), (iii) predominant rod or rod-cone dystrophies (retinitis pigmentosa, enhanced S-Cone syndrome (NR2E3), Bietti crystalline corneoretinal dystrophy (CYP4V2)), (iv) Leber congenital amaurosis/early-onset severe retinal dystrophy (GUCY2D, CEP290, CRB1, RDH12, RPE65, TULP1, AIPL1 and NMNAT1), (v) cone dysfunction syndromes (achromatopsia (CNGA3, CNGB3, PDE6C, PDE6H, GNAT2, ATF6), X-linked cone dysfunction with myopia and dichromacy (Bornholm Eye disease; OPN1LW/OPN1MW array), oligocone trichromacy, and blue-cone monochromatism (OPN1LW/OPN1MW array)). Whilst we use the aforementioned classical phenotypic groupings, a key feature of IRD is that it is characterised by tremendous heterogeneity and variable expressivity, with several of the above genes associated with a range of phenotypes.
Collapse
Affiliation(s)
- Michalis Georgiou
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Anthony G Robson
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom.
| | - Kaoru Fujinami
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.
| | - Thales A C de Guimarães
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom.
| | - Yu Fujinami-Yokokawa
- UCL Institute of Ophthalmology, University College London, London, United Kingdom; Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan; Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan.
| | - Malena Daich Varela
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom.
| | - Nikolas Pontikos
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom.
| | - Angelos Kalitzeos
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom.
| | - Omar A Mahroo
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Section of Ophthalmology, King s College London, St Thomas Hospital Campus, London, United Kingdom; Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom; Department of Translational Ophthalmology, Wills Eye Hospital, Philadelphia, PA, USA.
| | - Andrew R Webster
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom.
| | - Michel Michaelides
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom.
| |
Collapse
|
3
|
Beryozkin A, Sher I, Ehrenberg M, Zur D, Newman H, Gradstein L, Simaan F, Rotenstreich Y, Goldenberg-Cohen N, Bahar I, Blumenfeld A, Rivera A, Rosin B, Deitch-Harel I, Perlman I, Mechoulam H, Chowers I, Leibu R, Ben-Yosef T, Pras E, Banin E, Sharon D, Khateb S. Best Disease: Global Mutations Review, Genotype-Phenotype Correlation, and Prevalence Analysis in the Israeli Population. Invest Ophthalmol Vis Sci 2024; 65:39. [PMID: 38411968 PMCID: PMC10910552 DOI: 10.1167/iovs.65.2.39] [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: 10/17/2023] [Accepted: 02/04/2024] [Indexed: 02/28/2024] Open
Abstract
Purpose To review all reported disease-causing mutations in BEST1, perform genotype-phenotype correlation, and estimate disease prevalence in the Israeli population. Methods Medical records of patients diagnosed with Best disease and allied diseases from nine Israeli medical centers over the past 20 years were collected, as were clinical data including ocular findings, electrophysiology results, and retina imaging. Mutation detection involved mainly whole exome sequencing and candidate gene analysis. Demographic data were obtained from the Israeli Bureau of Statistics (January 2023). A bibliometric study was also conducted to gather mutation data from online sources. Results A total of 134 patients were clinically diagnosed with Best disease and related conditions. The estimated prevalence of Best disease was calculated to be 1 in 127,000, with higher rates among Arab Muslims (1 in 76,000) than Jews (1 in 145,000). Genetic causes were identified in 76 individuals (57%), primarily showing autosomal-dominant inheritance due to BEST1 mutations (58 patients). Critical conserved domains were identified consisting of a high percentage of dominant missense mutations, primarily in transmembrane domains and the intracellular region (Ca2+ binding domain) of the BEST1 protein. Conclusions This study represents the largest cohort of patients with Best disease reported in Israel and globally. The prevalence in Israel is akin to that in Denmark but is lower than that in the United States. Critical conserved domains within the BEST1 protein are pivotal for normal functioning, and even minor missense alterations in these areas lead to a dominant disease manifestation. Genetic testing is indispensable as the gold standard for Best disease diagnosis due to the variable clinical presentation of the disease.
Collapse
Affiliation(s)
- Avigail Beryozkin
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Ifat Sher
- Goldschleger Eye Institute, Sheba Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Miriam Ehrenberg
- Ophthalmology Unit, Schneider Children's Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dinah Zur
- Ophthalmology Division, Tel Aviv Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hadas Newman
- Ophthalmology Division, Tel Aviv Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Libe Gradstein
- Department of Ophthalmology, Soroka Medical Center and Clalit Health Services, Faculty of Health Sciences, Ben-Gurion University, Be'er Sheva, Israel
| | - Francis Simaan
- Department of Ophthalmology, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Ygal Rotenstreich
- Goldschleger Eye Institute, Sheba Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nitza Goldenberg-Cohen
- Department of Ophthalmology, Bnai Zion Medical Center, Haifa, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Irit Bahar
- Ophthalmology Division, Tel Aviv Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Rabin Medical Center, Petach Tikva, Israel
| | - Anat Blumenfeld
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Antonio Rivera
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Boris Rosin
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Iris Deitch-Harel
- Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Rabin Medical Center, Petach Tikva, Israel
| | - Ido Perlman
- Ophthalmology Division, Tel Aviv Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hadas Mechoulam
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Itay Chowers
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rina Leibu
- Department of Ophthalmology, Rambam Health Care Center, Haifa, Israel
| | - Tamar Ben-Yosef
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Eran Pras
- Department of Ophthalmology, Assaf Harofeh Medical Center, Zerifin, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eyal Banin
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dror Sharon
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samer Khateb
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
4
|
Cideciyan AV, Jacobson SG, Sumaroka A, Swider M, Krishnan AK, Sheplock R, Garafalo AV, Guziewicz KE, Aguirre GD, Beltran WA, Matsui Y, Kondo M, Heon E. Photoreceptor function and structure in retinal degenerations caused by biallelic BEST1 mutations. Vision Res 2023; 203:108157. [PMID: 36450205 PMCID: PMC9825664 DOI: 10.1016/j.visres.2022.108157] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/17/2022] [Accepted: 11/07/2022] [Indexed: 11/29/2022]
Abstract
The only approved retinal gene therapy is for biallelic RPE65 mutations which cause a recessive retinopathy with a primary molecular defect located at the retinal pigment epithelium (RPE). For a distinct recessive RPE disease caused by biallelic BEST1 mutations, a pre-clinical proof-of-concept for gene therapy has been demonstrated in canine eyes. The current study was undertaken to consider potential outcome measures for a BEST1 clinical trial in patients demonstrating a classic autosomal recessive bestrophinopathy (ARB) phenotype. Spatial distribution of retinal structure showed a wide expanse of abnormalities including large intraretinal cysts, shallow serous retinal detachments, abnormalities of inner and outer segments, and an unusual prominence of the external limiting membrane. Surrounding the central macula extending from 7 to 30 deg eccentricity, outer nuclear layer was thicker than expected from a cone only retina and implied survival of many rod photoreceptors. Co-localized however, were large losses of rod sensitivity despite preserved cone sensitivities. The dissociation of rod function from rod structure observed, supports a large treatment potential in the paramacular region for biallelic bestrophinopathies.
Collapse
Affiliation(s)
- Artur V Cideciyan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Samuel G Jacobson
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander Sumaroka
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malgorzata Swider
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arun K Krishnan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca Sheplock
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra V Garafalo
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karina E Guziewicz
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gustavo D Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William A Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yoshitsugu Matsui
- Department of Ophthalmology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Mineo Kondo
- Department of Ophthalmology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Elise Heon
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 2L3, Canada
| |
Collapse
|
5
|
Cideciyan AV, Jacobson SG, Swider M, Sumaroka A, Sheplock R, Krishnan AK, Garafalo AV, Guziewicz KE, Aguirre GD, Beltran WA, Heon E. Photoreceptor Function and Structure in Autosomal Dominant Vitelliform Macular Dystrophy Caused by BEST1 Mutations. Invest Ophthalmol Vis Sci 2022; 63:12. [PMID: 36512348 DOI: 10.1167/iovs.63.13.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose The purpose of this study was to evaluate rod and cone function and outer retinal structure within macular lesions, and surrounding extralesional areas of patients with autosomal dominant Best vitelliform macular dystrophy caused by BEST1 mutations. Methods Seventeen patients from seven families were examined with dark- and light-adapted chromatic perimetry and optical coherence tomography. Subsets of patients had long-term follow-up (14-22 years, n = 6) and dark-adaptation kinetics measured (n = 5). Results Within central lesions with large serous retinal detachments, rod sensitivity was severely reduced but visual acuity and cone sensitivity were relatively retained. In surrounding extralesional areas, there was a mild but detectable widening of the subretinal space in some patients and some retinal areas. Available evidence was consistent with subretinal widening causing slower dark-adaptation kinetics. Over long-term follow-up, some eyes showed formation of de novo satellite lesions at retinal locations that years previously demonstrated subretinal widening. A subclinical abnormality consisting of a retina-wide mild thickening of the outer nuclear layer was evident in many patients and thickening increased in the subset of patients with long-term follow-up. Conclusions Outcome measures for future clinical trials should include evaluations of rod sensitivity within central lesions and quantitative measures of outer retinal structure in normal-appearing regions surrounding the lesions.
Collapse
Affiliation(s)
- Artur V Cideciyan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Samuel G Jacobson
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Malgorzata Swider
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Alexander Sumaroka
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Rebecca Sheplock
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Arun K Krishnan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Alexandra V Garafalo
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Karina E Guziewicz
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Gustavo D Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - William A Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Elise Heon
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| |
Collapse
|
6
|
Crincoli E, Zhao Z, Querques G, Sacconi R, Carlà MM, Giannuzzi F, Ferrara S, Ribarich N, L'Abbate G, Rizzo S, Souied EH, Miere A. Deep learning to distinguish Best vitelliform macular dystrophy (BVMD) from adult-onset vitelliform macular degeneration (AVMD). Sci Rep 2022; 12:12745. [PMID: 35882966 PMCID: PMC9325755 DOI: 10.1038/s41598-022-16980-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
Initial stages of Best vitelliform macular dystrophy (BVMD) and adult vitelliform macular dystrophy (AVMD) harbor similar blue autofluorescence (BAF) and optical coherence tomography (OCT) features. Nevertheless, BVMD is characterized by a worse final stage visual acuity (VA) and an earlier onset of critical VA loss. Currently, differential diagnosis requires an invasive and time-consuming process including genetic testing, electrooculography (EOG), full field electroretinogram (ERG), and visual field testing. The aim of our study was to automatically classify OCT and BAF images from stage II BVMD and AVMD eyes using a deep learning algorithm and to identify an image processing method to facilitate human-based clinical diagnosis based on non-invasive tests like BAF and OCT without the use of machine-learning technology. After the application of a customized image processing method, OCT images were characterized by a dark appearance of the vitelliform deposit in the case of BVMD and a lighter inhomogeneous appearance in the case of AVMD. By contrast, a customized method for processing of BAF images revealed that BVMD and AVMD were characterized respectively by the presence or absence of a hypo-autofluorescent region of retina encircling the central hyperautofluorescent foveal lesion. The human-based evaluation of both BAF and OCT images showed significantly higher correspondence to ground truth reference when performed on processed images. The deep learning classifiers based on BAF and OCT images showed around 90% accuracy of classification with both processed and unprocessed images, which was significantly higher than human performance on both processed and unprocessed images. The ability to differentiate between the two entities without recurring to invasive and expensive tests may offer a valuable clinical tool in the management of the two diseases.
Collapse
Affiliation(s)
- Emanuele Crincoli
- Department of Ophthalmology, Centre Hospitalier Intercommunal de Créteil, 40, avenue de Verdun, 94100, Créteil, France.,Ophthalmology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00166, Rome, Italy.,Catholic University of "Sacro Cuore", Largo Francesco Vito 1, 00166, Rome, Italy
| | - Zhanlin Zhao
- Department of Ophthalmology, Centre Hospitalier Intercommunal de Créteil, 40, avenue de Verdun, 94100, Créteil, France
| | - Giuseppe Querques
- Department of Ophthalmology University Vita-Salute IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Riccardo Sacconi
- Department of Ophthalmology University Vita-Salute IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Matteo Maria Carlà
- Ophthalmology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00166, Rome, Italy.,Catholic University of "Sacro Cuore", Largo Francesco Vito 1, 00166, Rome, Italy
| | - Federico Giannuzzi
- Ophthalmology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00166, Rome, Italy.,Catholic University of "Sacro Cuore", Largo Francesco Vito 1, 00166, Rome, Italy
| | - Silvia Ferrara
- Ophthalmology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00166, Rome, Italy.,Catholic University of "Sacro Cuore", Largo Francesco Vito 1, 00166, Rome, Italy
| | - Nicolò Ribarich
- Department of Ophthalmology University Vita-Salute IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Gaia L'Abbate
- Department of Ophthalmology University Vita-Salute IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Stanislao Rizzo
- Ophthalmology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00166, Rome, Italy.,Catholic University of "Sacro Cuore", Largo Francesco Vito 1, 00166, Rome, Italy
| | - Eric H Souied
- Department of Ophthalmology, Centre Hospitalier Intercommunal de Créteil, 40, avenue de Verdun, 94100, Créteil, France.,Ethics Committee of the Federation France Macula, 2018-27, 40 Av. de Verdun, 94010, Créteil, France
| | - Alexandra Miere
- Department of Ophthalmology, Centre Hospitalier Intercommunal de Créteil, 40, avenue de Verdun, 94100, Créteil, France.
| |
Collapse
|
7
|
Impaired Bestrophin Channel Activity in an iPSC-RPE Model of Best Vitelliform Macular Dystrophy (BVMD) from an Early Onset Patient Carrying the P77S Dominant Mutation. Int J Mol Sci 2022; 23:ijms23137432. [PMID: 35806438 PMCID: PMC9266689 DOI: 10.3390/ijms23137432] [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: 06/03/2022] [Accepted: 07/01/2022] [Indexed: 01/25/2023] Open
Abstract
Best Vitelliform Macular dystrophy (BVMD) is the most prevalent of the distinctive retinal dystrophies caused by mutations in the BEST1 gene. This gene, which encodes for a homopentameric calcium-activated ion channel, is crucial for the homeostasis and function of the retinal pigment epithelia (RPE), the cell type responsible for recycling the visual pigments generated by photoreceptor cells. In BVMD patients, mutations in this gene induce functional problems in the RPE cell layer with an accumulation of lipofucsin that evolves into cell death and loss of sight. In this work, we employ iPSC-RPE cells derived from a patient with the p.Pro77Ser dominant mutation to determine the correlation between this variant and the ocular phenotype. To this purpose, gene and protein expression and localization are evaluated in iPSC-RPE cells along with functional assays like phagocytosis and anion channel activity. Our cell model shows no differences in gene expression, protein expression/localization, or phagocytosis capacity, but presents an increased chloride entrance, indicating that the p.Pro77Ser variant might be a gain-of-function mutation. We hypothesize that this variant disturbs the neck region of the BEST1 channel, affecting channel function but maintaining cell homeostasis in the short term. This data shed new light on the different phenotypes of dominant mutations in BEST1, and emphasize the importance of understanding its molecular mechanisms. Furthermore, the data widen the knowledge of this pathology and open the door for a better diagnosis and prognosis of the disease.
Collapse
|
8
|
Nowomiejska K, Nasser F, Stingl K, Schimpf‐Linzenbold S, Biskup S, Brzozowska A, Rejdak R, Kohl S, Zrenner E. Disease expression caused by different variants in the BEST1 gene: genotype and phenotype findings in bestrophinopathies. Acta Ophthalmol 2022; 100:e847-e858. [PMID: 34327816 PMCID: PMC9328113 DOI: 10.1111/aos.14958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 06/17/2021] [Indexed: 12/30/2022]
Abstract
Purpose: To analyse the spectrum of clinical features and molecular genetic data in a series of patients carrying likely disease-associated variants in the BEST1 gene. Methods: Retrospective observational analysis of clinical data extracted from the medical records of visual function, multimodal imaging and electrophysiology of 62 eyes of 31 patients. Molecular genetic analysis was performed by means of panel-based NGS or Sanger sequencing. Results: The spectrum of variants in the BEST1 gene comprised 19 different variants and three of which are novel. Fundus photographs and OCT images allowed categorization of 52 eyes as Best vitelliform macular dystrophy (BVMD) with stages 1 to 5 and 10 eyes with autosomal recessive bestrophinopathy (ARB), with more severe phenotype. One patient was shown to be heterozygous for a variant, which has so far been described only in ARB, but this patient had the BVMD phenotype. There was no significant progression of the visual acuity during the follow-up period of 5 years both in BVMD and ARB. The most prevalent pattern of fundus autofluorescence (FAF) in BVMD was ‘patchy’. There were diverse visual field defects in static automated perimetry (SAP) depending on the stage. The Arden ratio was significantly lower in ARB patients and in eyes with stage 5 of BVMD. Conclusions: The genotype does not always predict the phenotype in patients with BVMD and ARB; however, having two mutations in the BEST1 gene causes a more severephenotype. FAFhelped to distinguish ARB from BVMD. Most of the observed eyesdidnotprogressfunctionallyduringthefollow-up.ARBandtheatrophicstageof BVMD as the disease end-stage had the worst visual functions and EOG results.
Collapse
Affiliation(s)
- Katarzyna Nowomiejska
- Chair and Department of General and Pediatric Ophthalmology Medical University of Lublin Lublin Poland
- Institute for Ophthalmic Research Center for Ophthalmology University of Tübingen Tübingen Germany
| | - Fadi Nasser
- Institute for Ophthalmic Research Center for Ophthalmology University of Tübingen Tübingen Germany
| | - Katarina Stingl
- University Eye Hospital Center for Ophthalmology University of Tübingen Tübingen Germany
- Center for Rare Eye Diseases University of Tübingen Tübingen Germany
| | | | | | - Agnieszka Brzozowska
- Department of Mathematics and Medical Biostatistics Medical University of Lublin Lublin Poland
| | - Robert Rejdak
- Chair and Department of General and Pediatric Ophthalmology Medical University of Lublin Lublin Poland
| | - Susanne Kohl
- Institute for Ophthalmic Research Center for Ophthalmology University of Tübingen Tübingen Germany
| | - Eberhart Zrenner
- Institute for Ophthalmic Research Center for Ophthalmology University of Tübingen Tübingen Germany
- Werner Reichardt Centre for Integrative Neuroscience University of Tübingen Tübingen Germany
| |
Collapse
|
9
|
Chowdhury S, Duvesh R, Kumaran M, Anjanamurthy R, Kumar J, Vanniarajan A, Devarajan B, Sundaresan P. Clinical reassessments and whole-exome sequencing uncover novel BEST1 mutation associated with bestrophinopathy phenotype. Ophthalmic Genet 2021; 43:191-200. [PMID: 34751623 DOI: 10.1080/13816810.2021.1998553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND The diagnosis of retinal dystrophies can be challenging due to the spectrum of protean phenotypic manifestations. This study employed trio-whole-exome sequencing (trio-WES) to unveil the genetic cause of an inherited retinal disorder in a south Indian family. MATERIALS AND METHODS Proband's initial ophthalmic examinations was performed in the year 2016. WES was performed on a proband-parent trio to identify causative mutation followed by Sanger validation, segregation analysis, sequence and structure-based computational analysis to assess its pathogenicity. Based on the genetic findings, detailed clinical reassessments were performed in year 2020 for the proband and available family members. RESULTS WES revealed a novel homozygous BEST1 mutation c.G310A (p.D104N) in the proband and heterozygous for the parents, indicating autosomal recessive inheritance. Segregation analysis showed heterozygous mutation in maternal grandfather and normal genotype for younger brother and maternal grandmother. Moreover, the structure-based analysis revealed the mutation p.D104N in the cytoplasmic domain, causing structural hindrance by altering hydrogen bonds and destabilizing the BEST1 protein structure. Proband's clinical assessments were consistent with autosomal recessive bestrophinopathy (ARB) phenotype. Additionally, characteristic absent light rise and decreased light peak-to-dark trough ratio (LP:DT) was observed bilaterally in EOG. CONCLUSIONS Our study demonstrates the utility of WES and clinical re-evaluations in establishing the precise diagnosis of autosomal recessive bestrophinopathy associated with a novel mutation, thus expanding the BEST1-related mutation spectrum.
Collapse
Affiliation(s)
- Susmita Chowdhury
- Department of Genetics, Aravind Medical Research Foundation, Madurai, India.,Department of Molecular Biology, Aravind Medical Research Foundation - Affiliated to Alagappa University, Karaikudi, India
| | - Roopam Duvesh
- Department of Genetics, Aravind Medical Research Foundation, Madurai, India
| | - Manojkumar Kumaran
- Department of Bioinformatics, Aravind Medical Research Foundation, Madurai, India.,School of Chemical and Biotechnology, SASTRA (Deemed to Be University), Thanjavur, India
| | - Rupa Anjanamurthy
- Department of Paediatric Ophthalmology & Adult Strabismus Services, Aravind Eye Hospital, Madurai, India
| | - Jayant Kumar
- Department of Vitreo-Retina Services, Aravind Eye Hospital, Madurai, India
| | - Ayyasamy Vanniarajan
- Department of Molecular Genetics, Aravind Medical Research Foundation, Madurai, India
| | | | - Periasamy Sundaresan
- Department of Genetics, Aravind Medical Research Foundation, Madurai, India.,Department of Molecular Biology, Aravind Medical Research Foundation - Affiliated to Alagappa University, Karaikudi, India
| |
Collapse
|
10
|
Owji AP, Kittredge A, Zhang Y, Yang T. Structure and Function of the Bestrophin family of calcium-activated chloride channels. Channels (Austin) 2021; 15:604-623. [PMID: 34612806 PMCID: PMC8496536 DOI: 10.1080/19336950.2021.1981625] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Bestrophins are a family of calcium-activated chloride channels (CaCCs) with relevance to human physiology and a myriad of eye diseases termed "bestrophinopathies". Since the identification of bestrophins as CaCCs nearly two decades ago, extensive studies from electrophysiological and structural biology perspectives have sought to define their key channel features including calcium sensing, gating, inactivation, and anion selectivity. The initial X-ray crystallography studies on the prokaryotic homolog of Best1, Klebsiella pneumoniae (KpBest), and the Best1 homolog from Gallus gallus (chicken Best1, cBest1), laid the foundational groundwork for establishing the architecture of Best1. Recent progress utilizing single-particle cryogenic electron microscopy has further elucidated the molecular mechanism of gating in cBest1 and, separately, the structure of Best2 from Bos taurus (bovine Best2, bBest2). Meanwhile, whole-cell patch clamp, planar lipid bilayer, and other electrophysiologic analyses using these models as well as the human Best1 (hBest1) have provided ample evidence describing the functional properties of the bestrophin channels. This review seeks to consolidate these structural and functional results to paint a broad picture of the underlying mechanisms comprising the bestrophin family's structure-function relationship.
Collapse
Affiliation(s)
- Aaron P Owji
- Department of Pharmacology, Columbia University, NY, USA
| | - Alec Kittredge
- Department of Pharmacology, Columbia University, NY, USA
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, NY, USA
| | - Tingting Yang
- Department of Ophthalmology, Columbia University, NY, USA
| |
Collapse
|
11
|
Ravichandran G, Raju SV, Sarkar P, N. T. S, Al Olayan EM, Aloufi AS, Elokaby MA, Arshad A, Mala K, Arockiaraj J. Bestrophin‐derived peptide, WP17, elicits cell wall disruption‐mediated bactericidal activity against Micrococcus luteus and anti‐neoplastic effect against murine melanoma cells. Pept Sci (Hoboken) 2021; 113. [DOI: 10.1002/pep2.24220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/06/2021] [Indexed: 01/10/2023]
Abstract
AbstractThe cDNa sequence of Bestrophin‐1 (BEST‐1) was identified from a previously constructed transcriptome data set of freshwater prawn Macrobrachium rosenbergii (Mr). Basal and temporal gene expression analysis of MrBEST‐1 showed its antimicrobial immune effectiveness during viral and bacterial infections. The protein sequence encoded by cDNA of MrBEST‐1 was examined and a short antimicrobial molecule, named WP17 was identified using a bioinformatics tool. Further, the antibacterial ability of the identified WP17 peptide was evaluated against a number of bacterial strains, in which the peptide showed potential bactericidal activity against Micrococcus luteus (MTCC 6164), Staphylococcus aureus (ATCC 9144), Escherichia coli (ATCC 9637), Klebsiella pneumonia (CI 7376) and Bacillus subtilis (ATCC 6051). Based on the results, further assays focused on M. luteus MTCC 6164. The mode of action of MrWP17 on M. luteus MTCC 6164 was analyzed using FACS and FESEM. Toxicity analysis suggested that WP17 impaired the viability of cells in murine melanoma cells (B16F10); however, no cytotoxicity was observed against kidney embryonic cells (HEK293), even at higher concentrations. Similarly, the gene expression analysis of WP17 peptide treated murine cells elicited an extrinsic apoptotic pathway. In the present study, we have demonstrated the involvement of MrBEST‐1 in immune mechanisms through its short peptide molecule that has potential antimicrobial activity.
Collapse
Affiliation(s)
- Gayathri Ravichandran
- SRM Research Institute, SRM Institute of Science and Technology Chennai Tamil Nadu India
| | - Stefi V. Raju
- SRM Research Institute, SRM Institute of Science and Technology Chennai Tamil Nadu India
| | - Purabi Sarkar
- SRM Research Institute, SRM Institute of Science and Technology Chennai Tamil Nadu India
| | - Saraswathi N. T.
- Molecular Biophysics Lab School of Chemical and Biotechnology, SASTRA Deemed to be University Thanjavur Tamil Nadu India
| | - Ebtesam M. Al Olayan
- Department of Zoology College of Science, King Saud University Riyadh Saudi Arabia
| | - Abeer S. Aloufi
- Department of Zoology College of Science, King Saud University Riyadh Saudi Arabia
| | - Mohamed A. Elokaby
- Aquaculture Division National Institute of Oceanography and Fisheries (NIOF) Alexandria Egypt
| | - Aziz Arshad
- International Institute of Aquaculture and Aquatic Sciences (I‐AQUAS) Universiti Putra Malaysia Port Dickson Negeri Sembilan Malaysia
- Department of Aquaculture, Faculty of Agriculture University Putra Malaysia Serdang Selangor Malaysia
| | - Kanchana Mala
- Department of Medical Research Medical College Hospital & Research Centre, SRM Institute of Science & Technology Chennai Tamil Nadu India
| | - Jesu Arockiaraj
- SRM Research Institute, SRM Institute of Science and Technology Chennai Tamil Nadu India
| |
Collapse
|
12
|
Singh Grewal S, Smith JJ, Carr AJF. Bestrophinopathies: perspectives on clinical disease, Bestrophin-1 function and developing therapies. Ther Adv Ophthalmol 2021; 13:2515841421997191. [PMID: 33738427 PMCID: PMC7934022 DOI: 10.1177/2515841421997191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 12/17/2022] Open
Abstract
Bestrophinopathies are a group of clinically distinct inherited retinal dystrophies that typically affect the macular region, an area synonymous with central high acuity vision. This spectrum of disorders is caused by mutations in bestrophin1 (BEST1), a protein thought to act as a Ca2+-activated Cl- channel in the retinal pigment epithelium (RPE) of the eye. Although bestrophinopathies are rare, over 250 individual pathological mutations have been identified in the BEST1 gene, with many reported to have various clinical expressivity and incomplete penetrance. With no current clinical treatments available for patients with bestrophinopathies, understanding the role of BEST1 in cells and the pathological pathways underlying disease has become a priority. Induced pluripotent stem cell (iPSC) technology is helping to uncover disease mechanisms and develop treatments for RPE diseases, like bestrophinopathies. Here, we provide a comprehensive review of the pathophysiology of bestrophinopathies and highlight how patient-derived iPSC-RPE are being used to test new genomic therapies in vitro.
Collapse
Affiliation(s)
| | - Joseph J Smith
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Amanda-Jayne F Carr
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
13
|
Videv P, Mladenov N, Andreeva T, Mladenova K, Moskova-Doumanova V, Nikolaev G, Petrova SD, Doumanov JA. Condensing Effect of Cholesterol on hBest1/POPC and hBest1/SM Langmuir Monolayers. MEMBRANES 2021; 11:membranes11010052. [PMID: 33451008 PMCID: PMC7828479 DOI: 10.3390/membranes11010052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 11/22/2022]
Abstract
Human bestrophin-1 protein (hBest1) is a transmembrane channel associated with the calcium-dependent transport of chloride ions in the retinal pigment epithelium as well as with the transport of glutamate and GABA in nerve cells. Interactions between hBest1, sphingomyelins, phosphatidylcholines and cholesterol are crucial for hBest1 association with cell membrane domains and its biological functions. As cholesterol plays a key role in the formation of lipid rafts, motional ordering of lipids and modeling/remodeling of the lateral membrane structure, we examined the effect of different cholesterol concentrations on the surface tension of hBest1/POPC (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine) and hBest1/SM Langmuir monolayers in the presence/absence of Ca2+ ions using surface pressure measurements and Brewster angle microscopy studies. Here, we report that cholesterol: (1) has negligible condensing effect on pure hBest1 monolayers detected mainly in the presence of Ca2+ ions, and; (2) induces a condensing effect on composite hBest1/POPC and hBest1/SM monolayers. These results offer evidence for the significance of intermolecular protein–lipid interactions for the conformational dynamics of hBest1 and its biological functions as multimeric ion channel.
Collapse
Affiliation(s)
- Pavel Videv
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (N.M.); (K.M.); (V.M.-D.); (G.N.); (S.D.P.)
| | - Nikola Mladenov
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (N.M.); (K.M.); (V.M.-D.); (G.N.); (S.D.P.)
- Faculty of Medicine, Medical University-Sofia, 1 Sv. Georgi Sofiiski Str., 1431 Sofia, Bulgaria
| | - Tonya Andreeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 21, 1113 Sofia, Bulgaria;
- Faculty of Applied Chemistry, Reutlingen University, Alteburgstraße 150, 72762 Reutlingen, Germany
| | - Kirilka Mladenova
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (N.M.); (K.M.); (V.M.-D.); (G.N.); (S.D.P.)
| | - Veselina Moskova-Doumanova
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (N.M.); (K.M.); (V.M.-D.); (G.N.); (S.D.P.)
| | - Georgi Nikolaev
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (N.M.); (K.M.); (V.M.-D.); (G.N.); (S.D.P.)
| | - Svetla D. Petrova
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (N.M.); (K.M.); (V.M.-D.); (G.N.); (S.D.P.)
| | - Jordan A. Doumanov
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (N.M.); (K.M.); (V.M.-D.); (G.N.); (S.D.P.)
- Correspondence: ; Tel.: +359-2-8167262
| |
Collapse
|
14
|
Skarphedinsdottir SB, Eysteinsson T, Árnason SS. Mechanisms of Ion Transport Across the Mouse Retinal Pigment Epithelium Measured In Vitro. Invest Ophthalmol Vis Sci 2021; 61:31. [PMID: 32539134 PMCID: PMC7416899 DOI: 10.1167/iovs.61.6.31] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Purpose To examine ion transport across the mouse retinal pigment epithelium (RPE), measured by the short-circuit current (ISC) and transepithelial resistance (TER). Methods Sheets of RPE from mice (C57BL6/J) with retina, choroid, and sclera attached were mounted in Ussing chambers (0.031-cm2 aperture) and Krebs solution. The ISC and TER were recorded with voltage clamps. Receptors implicated in ion transport were blocked or stimulated by ligands applied to both sides. Results The mean initial ISC was −12.0 ± 3.9 µA/cm2 (basolateral negative), and mean TER was 67.1 ± 8.0 ohm·cm2. RPE preparations remained stable for 3 hours, with ISC decreasing by 0.078 ± 0,033 µA/cm2/hr. Adenosine triphosphate (100 µM) increased ISC by 2.22 ± 0.41 µA/cm2 (P = 0.003). Epinephrine (100 µM) increased ISC by 1.14 ± 0.19 µA/cm2 (P = 0.011). Bumetanide (100 µM) reduced ISC by 1.72 ± 0.73 µA/cm2 (P = 0.027). Ouabain (1 mM) induced a biphasic response: an ISC increase from −7.9 ± 2.4 to −15.49 ± 2.12 µA/cm2 and then a decrease to −3.7 ± 2.2 µA/cm2. Ouabain increased TER by 15.3 ± 4.8 ohm·cm2. These compounds were added sequentially. Apical [K+]o at zero mM transiently increased ISC by 3.36 ± 1.06 µA/cm2. Ba++ decreased ISC from −10.4 ± 3.1 to −6.6 ± 1.8 µA/cm2 (P = 0.01). Ba++ reversed the K+-free response, with Isc decreasing further from −5.65 ± 1.24 to −3.37 ± 0.79 µA/cm2 (P = 0.029). Conclusions The ISC and TER can be recorded from the mouse RPE for 3 hours. Adrenergic and purinergic receptors affect murine RPE ion transport. Sodium–potassium adenosine triphosphatase plays a role in net ion transport across mouse RPE, and Na-K-2Cl cotransporter activity partly accounts for transepithelial ion transport. Mimicking light-induced changes, low subretinal [K+]o increases ion transport transiently, dependent on K+ channels.
Collapse
|
15
|
Kittredge A, Zhang Y, Yang T. Evaluating BEST1 mutations in pluripotent stem cell-derived retinal pigment epithelial cells. Methods Enzymol 2021; 654:365-382. [PMID: 34120722 PMCID: PMC9801436 DOI: 10.1016/bs.mie.2021.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Bestrophin-1 (BEST1) is a calcium-activated chloride channel (CaCC) predominantly expressed at the basolateral membrane of the retinal pigment epithelium (RPE). Over 250 mutations in the BEST1 gene have been documented to cause at least five retinal degenerative disorders, commonly termed bestrophinopathies, to which no treatment is currently available. Therefore, understanding the influences of BEST1 disease-causing mutations on the physiological function of BEST1 in RPE is critical for deciphering the pathology of bestrophinopathies and developing therapeutic strategies for patients. However, this task has been impeded by the rarity of BEST1 mutations and limited accessibility to native human RPE cells. Here, we describe a pluripotent stem cell (PSC)-based pipeline for reproducibly generating RPE cells expressing endogenous or exogenous mutant BEST1, which provides us with a powerful "disease-in-a-dish" approach for studying BEST1 mutations in physiological environments.
Collapse
Affiliation(s)
- Alec Kittredge
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, United States
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, New York, NY, United States,Corresponding authors: ; ;
| | - Tingting Yang
- Department of Ophthalmology, Columbia University, New York, NY, United States,Corresponding authors: ; ;
| |
Collapse
|
16
|
Ormel L, Lauritzen KH, Schreiber R, Kunzelmann K, Gundersen V. GABA, but Not Bestrophin-1, Is Localized in Astroglial Processes in the Mouse Hippocampus and the Cerebellum. Front Mol Neurosci 2020; 13:135. [PMID: 32848599 PMCID: PMC7399226 DOI: 10.3389/fnmol.2020.00135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 07/07/2020] [Indexed: 11/13/2022] Open
Abstract
GABA is proposed to act as a gliotransmitter in the brain. Differences in GABA release from astroglia are thought to underlie differences in tonic inhibition between the cerebellum and the CA1 hippocampus. Here we used quantitative immunogold cytochemistry to localize and compare the levels of GABA in astroglia in these brain regions. We found that the density of GABA immunogold particles was similar in delicate processes of Bergman glia in the cerebellum and astrocytes in the CA1 hippocampus. The astrocytic GABA release is proposed to be mediated by, among others, the Ca2+ activated Cl- channel bestrophin-1. The bestrophin-1 antibodies did not show any significant bestrophin-1 signal in the brain of wt mice, nor in bestrophin-1 knockout mice. The bestrophin-1 signal was low both on Western blots and immunofluorescence laser scanning microscopic images. These results suggest that GABA is localized in astroglia, but in similar concentrations in the cerebellum and CA1 hippocampus, and thus cannot account for differences in tonic inhibition between these brain regions. Furthermore, our data seem to suggest that the GABA release from astroglia previously observed in the hippocampus and cerebellum occurs via mechanisms other than bestrophin-1.
Collapse
Affiliation(s)
- Lasse Ormel
- Section of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Neurology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Knut H Lauritzen
- Section of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Rainer Schreiber
- Department of Physiology, University of Regensburg, Regensburg, Germany
| | - Karl Kunzelmann
- Department of Physiology, University of Regensburg, Regensburg, Germany
| | - Vidar Gundersen
- Section of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Section for Movement Disorders, Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
17
|
Miscibility of hBest1 and sphingomyelin in surface films - A prerequisite for interaction with membrane domains. Colloids Surf B Biointerfaces 2020; 189:110893. [PMID: 32113084 DOI: 10.1016/j.colsurfb.2020.110893] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/08/2020] [Accepted: 02/20/2020] [Indexed: 01/07/2023]
Abstract
Human bestrophin-1 (hBest1) is a transmembrane Ca2+- dependent anion channel, associated with the transport of Cl-, HCO3- ions, γ-aminobutiric acid (GABA), glutamate (Glu), and regulation of retinal homeostasis. Its mutant forms cause retinal degenerative diseases, defined as Bestrophinopathies. Using both physicochemical - surface pressure/mean molecular area (π/A) isotherms, hysteresis, compressibility moduli of hBest1/sphingomyelin (SM) monolayers, Brewster angle microscopy (BAM) studies, and biological approaches - detergent membrane fractionation, Laurdan (6-dodecanoyl-N,N-dimethyl-2-naphthylamine) and immunofluorescence staining of stably transfected MDCK-hBest1 and MDCK II cells, we report: 1) Ca2+, Glu and GABA interact with binary hBest1/SM monolayers at 35 °C, resulting in changes in hBest1 surface conformation, structure, self-organization and surface dynamics. The process of mixing in hBest1/SM monolayers is spontaneous and the effect of protein on binary films was defined as "fluidizing", hindering the phase-transition of monolayer from liquid-expanded to intermediate (LE-M) state; 2) in stably transfected MDCK-hBest1 cells, bestrophin-1 was distributed between detergent resistant (DRM) and detergent-soluble membranes (DSM) - up to 30 % and 70 %, respectively; in alive cells, hBest1 was visualized in both liquid-ordered (Lo) and liquid-disordered (Ld) fractions, quantifying protein association up to 35 % and 65 % with Lo and Ld. Our results indicate that the spontaneous miscibility of hBest1 and SM is a prerequisite to diverse protein interactions with membrane domains, different structural conformations and biological functions.
Collapse
|
18
|
Abstract
Bestrophinopathies are a group of clinically distinct inherited retinal dystrophies that lead to the gradual loss of vision in and around the macular area. There are no treatments for patients suffering from bestrophinopathies, and no measures can be taken to prevent visual deterioration in those who have inherited disease-causing mutations. Bestrophinopathies are caused by mutations in the Bestrophin1 gene (BEST1), a protein found exclusively in the retinal pigment epithelial (RPE) cells of the eye. Mutations in BEST1 affect the function of the RPE leading to the death of overlying retinal cells and subsequent vision loss. The pathogenic mechanisms arising from BEST1 mutations are still not fully understood, and it is not clear how mutations in BEST1 lead to diseases with distinct clinical features. This chapter discusses BEST1, the use of model systems to investigate the effects of mutations and the potential to investigate individual bestrophinopathies using induced pluripotent stem cells.
Collapse
|
19
|
Ji C, Li Y, Kittredge A, Hopiavuori A, Ward N, Yao P, Fukuda Y, Zhang Y, Tsang SH, Yang T. Investigation and Restoration of BEST1 Activity in Patient-derived RPEs with Dominant Mutations. Sci Rep 2019; 9:19026. [PMID: 31836750 PMCID: PMC6910965 DOI: 10.1038/s41598-019-54892-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/19/2019] [Indexed: 12/24/2022] Open
Abstract
BEST1 is a Ca2+-activated Cl- channel predominantly expressed in retinal pigment epithelium (RPE), and over 250 genetic mutations in the BEST1 gene have been identified to cause retinal degenerative disorders generally known as bestrophinopathies. As most BEST1 mutations are autosomal dominant, it is of great biomedical interest to determine their disease-causing mechanisms and the therapeutic potential of gene therapy. Here, we characterized six Best vitelliform macular dystrophy (BVMD)-associated BEST1 dominant mutations by documenting the patients' phenotypes, examining the subcellular localization of endogenous BEST1 and surface Ca2+-dependent Cl- currents in patient-derived RPEs, and analyzing the functional influences of these mutations on BEST1 in HEK293 cells. We found that all six mutations are loss-of-function with different levels and types of deficiencies, and further demonstrated the restoration of Ca2+-dependent Cl- currents in patient-derived RPE cells by WT BEST1 gene supplementation. Importantly, BEST1 dominant and recessive mutations are both rescuable at a similar efficacy by gene augmentation via adeno-associated virus (AAV), providing a proof-of-concept for curing the vast majority of bestrophinopathies.
Collapse
Affiliation(s)
- Changyi Ji
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Yao Li
- Department of Ophthalmology, Columbia University, New York, NY, 10032, USA
| | - Alec Kittredge
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Austin Hopiavuori
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Nancy Ward
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Peng Yao
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, School of Medicine & Dentistry, Rochester, NY, 14586, USA
| | - Yohta Fukuda
- Division of Advance Pharmaco-Science, Graduate School of Pharmaceutical Science, Osaka University, Yamadaoka 1-6, Suita, Osaka, 565-0871, Japan
| | - Yu Zhang
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA.
- Department of Ophthalmology, Columbia University, New York, NY, 10032, USA.
| | - Stephen H Tsang
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology and Pathology & Cell Biology, Edward S. Harkness Eye Institute, Columbia Stem Cell Initiative, New York Presbyterian Hospital/Columbia University, New York, NY, 10032, USA.
| | - Tingting Yang
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA.
- Department of Ophthalmology, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
20
|
Outer Retinal Alterations Associated With Visual Outcomes in Best Vitelliform Macular Dystrophy. Am J Ophthalmol 2019; 208:429-437. [PMID: 31465755 DOI: 10.1016/j.ajo.2019.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 01/07/2023]
Abstract
PURPOSE To describe outer retinal structure in patients with Best vitelliform macular dystrophy (BVMD) using spectral-domain optical coherence tomography (OCT) and correlate these results with best-corrected visual acuity (BCVA) and patient age. DESIGN Retrospective cross-sectional study. METHODS Patients with molecularly confirmed BVMD were compared with normal control subjects (NCs). A complete clinical evaluation was performed, including BCVA, fundus photography, spectral-domain OCT, and fundus autofluorescence. Spectral-domain OCT images were analyzed to determine the stage of the lesion, the central macular thickness (CMT), the foveal outer nuclear layer (ONL) thickness, and tomographic structural changes. RESULTS Forty-two patients with BVMD (42 eyes) with a molecular diagnosis and 42 NCs (42 eyes) were included. Clinical stages (Gass clinical classification) were distributed as follows: 4.8% for stage 1, 23.8% for stage 2, 16.6% for stage 3, 45.2% for stage 4, and 9.5% for stage 5. The presence of subretinal fluid and vitelliform material was noted in 76% and 79% of the BVMD eyes examined, respectively, and was not associated with BCVA modification (P = .758 and P = .968, respectively). The median ONL thickness was significantly lower compared with the NCs (P < .001). BCVA was significantly correlated with stage (R = 0.710; P < .01), age (R = 0.448; P < .01), CMT (R = -0.411; P < .01), and ONL thickness (R = -0.620; P < .01). The disruption of the external limiting membrane and the ellipsoid zone was associated with a decreased BCVA (P < .001 for both). Among the 32 eyes with subretinal detachment, photoreceptor outer segment length was significantly correlated with BCVA (R = -0.467; P < .01) and ONL thickness (R = 0.444; P = < .01). CONCLUSION This study shows the correlation between BCVA, age, and spectral-domain OCT features in patients with BVMD. ONL thickness as well as photoreceptor outer segment length are relevant functional correlates and outcome measures to follow photoreceptor impairments and disease progression.
Collapse
|
21
|
Rahman N, Georgiou M, Khan KN, Michaelides M. Macular dystrophies: clinical and imaging features, molecular genetics and therapeutic options. Br J Ophthalmol 2019; 104:451-460. [PMID: 31704701 PMCID: PMC7147237 DOI: 10.1136/bjophthalmol-2019-315086] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/24/2019] [Accepted: 10/21/2019] [Indexed: 11/03/2022]
Abstract
Macular dystrophies (MDs) consist of a heterogeneous group of disorders that are characterised by bilateral symmetrical central visual loss. Advances in genetic testing over the last decade have led to improved knowledge of the underlying molecular basis. The developments in high-resolution multimodal retinal imaging have also transformed our ability to make accurate and more timely diagnoses and more sensitive quantitative assessment of disease progression, and allowed the design of optimised clinical trial endpoints for novel therapeutic interventions. The aim of this review was to provide an update on MDs, including Stargardt disease, Best disease, X-linked r etinoschisis, pattern dystrophy, Sorsby fundus dystrophy and autosomal dominant drusen. It highlights the range of innovations in retinal imaging, genotype-phenotype and structure-function associations, animal models of disease and the multiple treatment strategies that are currently in clinical trial or planned in the near future, which are anticipated to lead to significant changes in the management of patients with MDs.
Collapse
Affiliation(s)
| | - Michalis Georgiou
- Moorfields Eye Hospital, London, UK.,Institute of Ophthalmology, UCL, London, UK
| | - Kamron N Khan
- Ophthalmology Department, St James's University Hospital, Leeds, UK
| | - Michel Michaelides
- Moorfields Eye Hospital, London, UK .,Institute of Ophthalmology, UCL, London, UK
| |
Collapse
|
22
|
Lin TC, Lin YY, Hsu CC, Yang YP, Yang CH, Hwang DK, Wang CY, Liu YY, Lo WL, Chiou SH, Peng CH, Chen SJ, Chang YL. Nanomedicine-based Curcumin Approach Improved ROS Damage in Best Dystrophy-specific Induced Pluripotent Stem Cells. Cell Transplant 2019; 28:1345-1357. [PMID: 31313605 PMCID: PMC6802151 DOI: 10.1177/0963689719860130] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Best dystrophy (BD), also termed best vitelliform macular dystrophy (BVMD), is a
juvenile-onset form of macular degeneration and can cause central visual loss.
Unfortunately, there is no clear definite therapy for BD or improving the visual function
on this progressive disease. The human induced pluripotent stem cell (iPSC) system has
been recently applied as an effective tool for genetic consultation and chemical drug
screening. In this study, we developed patient-specific induced pluripotent stem cells
(BD-iPSCs) from BD patient-derived dental pulp stromal cells and then differentiated
BD-iPSCs into retinal pigment epithelial cells (BD-RPEs). BD-RPEs were used as an
expandable platform for in vitro candidate drug screening. Compared with unaffected
sibling-derived iPSC-derived RPE cells (Ctrl-RPEs), BD-RPEs exhibited typical RPE-specific
markers with a lower expression of the tight junction protein ZO-1 and Bestrophin-1
(BEST1), as well as reduced phagocytic capabilities. Notably, among all candidate drugs,
curcumin was the most effective for upregulating both the BEST1 and ZO-1 genes in BD-RPEs.
Using the iPSC-based drug-screening platform, we further found that curcumin can
significantly improve the mRNA expression levels of Best gene in BD-iPSC-derived RPEs.
Importantly, we demonstrated that curcumin-loaded PLGA nanoparticles (Cur-NPs) were
efficiently internalized by BD-RPEs. The Cur-NPs-based controlled release formulation
further increased the expression of ZO-1 and Bestrophin-1, and promoted the function of
phagocytosis and voltage-dependent calcium channels in BD-iPSC-derived RPEs. We further
demonstrated that Cur-NPs enhanced the expression of antioxidant enzymes with a decrease
in intracellular ROS production and hydrogen peroxide-induced oxidative stress.
Collectively, these data supported that Cur-NPs provide a potential cytoprotective effect
by regulating the anti-oxidative abilities of degenerated RPEs. In addition, the
application of patient-specific iPSCs provides an effective platform for drug screening
and personalized medicine in incurable diseases.
Collapse
Affiliation(s)
- Tai-Chi Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei
| | - Yi-Ying Lin
- Institute of Pharmacology, National Yang-Ming University, Taipei
| | - Chih-Chen Hsu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei
| | - Yi-Ping Yang
- Institute of Pharmacology, National Yang-Ming University, Taipei.,School of Medicine, National Yang-Ming University, Taipei.,Department of Medical Research, Taipei Veterans General Hospital, Taipei
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei
| | - De-Kuang Hwang
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei.,School of Medicine, National Yang-Ming University, Taipei
| | - Chien-Ying Wang
- School of Medicine, National Yang-Ming University, Taipei.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei
| | - Yung-Yang Liu
- School of Medicine, National Yang-Ming University, Taipei.,Department of Chest, Taipei Veterans General Hospital, Taipei
| | - Wen-Liang Lo
- Department of Stomatology, Taipei Veterans General Hospital & Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei
| | - Shih-Hwa Chiou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei.,Institute of Pharmacology, National Yang-Ming University, Taipei.,Department of Medical Research, Taipei Veterans General Hospital, Taipei
| | - Chi-Hsien Peng
- Department of Ophthalmology, Shin Kong Wu Ho-Su Memorial Hospital & Fu-Jen Catholic University, Taipei
| | - Shih-Jen Chen
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei.,School of Medicine, National Yang-Ming University, Taipei
| | - Yuh-Lih Chang
- Institute of Pharmacology, National Yang-Ming University, Taipei.,School of Medicine, National Yang-Ming University, Taipei.,Department of Pharmacology, Taipei Veterans General Hospital, Taipei.,School of Pharmaceutical Sciences, National Yang-Ming University, Taipei
| |
Collapse
|
23
|
Milenkovic A, Schmied D, Tanimoto N, Seeliger MW, Sparrow JR, Weber BHF. The Y227N mutation affects bestrophin-1 protein stability and impairs sperm function in a mouse model of Best vitelliform macular dystrophy. Biol Open 2019; 8:bio.041335. [PMID: 31201163 PMCID: PMC6679414 DOI: 10.1242/bio.041335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Human bestrophin-1 (BEST1) is an integral membrane protein known to function as a Ca2+-activated and volume-regulated chloride channel. The majority of disease-associated mutations in BEST1 constitute missense mutations and were shown in vitro to lead to a reduction in mutant protein half-life causing Best disease (BD), a rare autosomal dominant macular dystrophy. To further delineate BEST1-associated pathology in vivo and to provide an animal model useful to explore experimental treatment efficacies, we have generated a knock-in mouse line (Best1Y227N). Heterozygous and homozygous mutants revealed no significant ocular abnormalities up to 2 years of age. In contrast, knock-in animals demonstrated a severe phenotype in the male reproductive tract. In heterozygous Best1Y227N males, Best1 protein was significantly reduced in testis and almost absent in homozygous mutant mice, although mRNA transcription of wild-type and knock-in allele is present and similar in quantity. Degradation of mutant Best1 protein in testis was associated with adverse effects on sperm motility and the capability to fertilize eggs. Based on these results, we conclude that mice carrying the Best1 Y227N mutation reveal a reproducible pathologic phenotype and thus provide a valuable in vivo tool to evaluate efficacy of drug therapies aimed at restoring Best1 protein stability and function.
Collapse
Affiliation(s)
- Andrea Milenkovic
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Denise Schmied
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Naoyuki Tanimoto
- Division of Ocular Neurodegeneration, Centre for Ophthalmology, Institute for Ophthalmic Research, 72076 Tübingen, Germany.,Department of Ophthalmology, University of Kiel, 24105 Kiel, Germany
| | - Mathias W Seeliger
- Division of Ocular Neurodegeneration, Centre for Ophthalmology, Institute for Ophthalmic Research, 72076 Tübingen, Germany
| | - Janet R Sparrow
- Department of Ophthalmology, Harkness Eye Institute, Columbia University Medical Center, 10032 New York, USA
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
24
|
Garnai SJ, Brinkmeier ML, Emery B, Aleman TS, Pyle LC, Veleva-Rotse B, Sisk RA, Rozsa FW, Ozel AB, Li JZ, Moroi SE, Archer SM, Lin CM, Sheskey S, Wiinikka-Buesser L, Eadie J, Urquhart JE, Black GC, Othman MI, Boehnke M, Sullivan SA, Skuta GL, Pawar HS, Katz AE, Huryn LA, Hufnagel RB, Camper SA, Richards JE, Prasov L. Variants in myelin regulatory factor (MYRF) cause autosomal dominant and syndromic nanophthalmos in humans and retinal degeneration in mice. PLoS Genet 2019; 15:e1008130. [PMID: 31048900 PMCID: PMC6527243 DOI: 10.1371/journal.pgen.1008130] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/20/2019] [Accepted: 04/09/2019] [Indexed: 01/11/2023] Open
Abstract
Nanophthalmos is a rare, potentially devastating eye condition characterized by small eyes with relatively normal anatomy, a high hyperopic refractive error, and frequent association with angle closure glaucoma and vision loss. The condition constitutes the extreme of hyperopia or farsightedness, a common refractive error that is associated with strabismus and amblyopia in children. NNO1 was the first mapped nanophthalmos locus. We used combined pooled exome sequencing and strong linkage data in the large family used to map this locus to identify a canonical splice site alteration upstream of the last exon of the gene encoding myelin regulatory factor (MYRF c.3376-1G>A), a membrane bound transcription factor that undergoes autoproteolytic cleavage for nuclear localization. This variant produced a stable RNA transcript, leading to a frameshift mutation p.Gly1126Valfs*31 in the C-terminus of the protein. In addition, we identified an early truncating MYRF frameshift mutation, c.769dupC (p.S264QfsX74), in a patient with extreme axial hyperopia and syndromic features. Myrf conditional knockout mice (CKO) developed depigmentation of the retinal pigment epithelium (RPE) and retinal degeneration supporting a role of this gene in retinal and RPE development. Furthermore, we demonstrated the reduced expression of Tmem98, another known nanophthalmos gene, in Myrf CKO mice, and the physical interaction of MYRF with TMEM98. Our study establishes MYRF as a nanophthalmos gene and uncovers a new pathway for eye growth and development.
Collapse
Affiliation(s)
- Sarah J. Garnai
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Michelle L. Brinkmeier
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Tomas S. Aleman
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- Scheie Eye Institute, Department of Ophthalmology, Philadelphia, PA, United States of America
| | - Louise C. Pyle
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Biliana Veleva-Rotse
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Robert A. Sisk
- Cincinnati Eye Institute, Cincinnati, Ohio, United States of America
| | - Frank W. Rozsa
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Molecular and Behavior Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Ayse Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Jun Z. Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Sayoko E. Moroi
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Steven M. Archer
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Cheng-mao Lin
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Sarah Sheskey
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Laurel Wiinikka-Buesser
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - James Eadie
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Jill E. Urquhart
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, St Mary’s Hospital, Manchester, United Kingdom
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Graeme C.M. Black
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, St Mary’s Hospital, Manchester, United Kingdom
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Mohammad I. Othman
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Scot A. Sullivan
- Dean McGee Eye Institute, Department of Ophthalmology, University of Oklahoma, Oklahoma City, OK
| | - Gregory L. Skuta
- Dean McGee Eye Institute, Department of Ophthalmology, University of Oklahoma, Oklahoma City, OK
| | - Hemant S. Pawar
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Alexander E. Katz
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Laryssa A. Huryn
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Robert B. Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States of America
| | | | - Sally A. Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States of America
| | - Julia E. Richards
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Lev Prasov
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States of America
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
25
|
Jaffal L, Joumaa WH, Assi A, Helou C, Condroyer C, El Dor M, Cherfan G, Zeitz C, Audo I, Zibara K, El Shamieh S. Novel Missense Mutations in BEST1 Are Associated with Bestrophinopathies in Lebanese Patients. Genes (Basel) 2019; 10:genes10020151. [PMID: 30781664 PMCID: PMC6409913 DOI: 10.3390/genes10020151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/07/2019] [Accepted: 02/07/2019] [Indexed: 12/25/2022] Open
Abstract
To identify Bestrophin 1 (BEST1) causative mutations in six Lebanese patients from three families, of whom four had a presumed clinical diagnosis of autosomal recessive bestrophinopathy (ARB) and two showed a phenotype with a single vitelliform lesion, patients were subjected to standard ophthalmic examinations. In addition, BEST1 exons and their flanking regions were amplified and sequenced by Sanger sequencing. Co-segregation and detailed bio-informatic analyses were performed. Clinical examination results were consistent with ARB diagnosis for all index patients showing multifocal vitelliform lesions and a markedly reduced light peak in the electrooculogram, including the two patients with a single vitelliform lesion. In all cases, most likely disease-causing BEST1 mutations co-segregated with the phenotype. The ARB cases showed homozygous missense variants (M1, c.209A>G, p.(Asp70Gly) in exon 3, M2, c.1403C>T; p.(Pro468Leu) in exon 10 and M3, c.830C>T, p.(Thr277Met) in exon 7), while the two patients with a single vitelliform lesion were compound heterozygous for M1 and M2. To our knowledge, this is the first study describing mutations in Lebanese patients with bestrophinopathy, where novel biallelic BEST1 mutations associated with two phenotypes were identified. Homozygous mutations were associated with multifocal lesions, subretinal fluid, and intraretinal cysts, whereas compound heterozygous ones were responsible for a single macular vitelliform lesion.
Collapse
Affiliation(s)
- Lama Jaffal
- Department of Biological and Environmental Sciences, Faculty of Science, Beirut Arab University, Debbieh 1107 2809, Lebanon.
| | - Wissam H Joumaa
- Rammal Hassan Rammal Research Laboratory, PhyToxE research group, Department of Life and Earth Sciences, Faculty of Sciences, Lebanese University, Nabatieh 1700, Lebanon.
| | - Alexandre Assi
- Retinal Service, Beirut Eye & ENT Specialist Hospital, Beirut 1106, Lebanon.
| | - Charles Helou
- Retinal Service, Beirut Eye & ENT Specialist Hospital, Beirut 1106, Lebanon.
| | - Christel Condroyer
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012 Paris, France.
| | - Maya El Dor
- Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.
| | - Georges Cherfan
- Retinal Service, Beirut Eye & ENT Specialist Hospital, Beirut 1106, Lebanon.
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012 Paris, France.
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012 Paris, France.
- CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DGOS CIC1423, F-75012 Paris, France.
- University College London Institute of Ophthalmology, London EC1V 9EL, UK.
| | - Kazem Zibara
- Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.
- ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon.
| | - Said El Shamieh
- Rammal Hassan Rammal Research Laboratory, PhyToxE research group, Department of Life and Earth Sciences, Faculty of Sciences, Lebanese University, Nabatieh 1700, Lebanon.
- Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut 1107 2809, Lebanon.
| |
Collapse
|
26
|
Milenkovic A, Milenkovic VM, Wetzel CH, Weber BHF. BEST1 protein stability and degradation pathways differ between autosomal dominant Best disease and autosomal recessive bestrophinopathy accounting for the distinct retinal phenotypes. Hum Mol Genet 2019; 27:1630-1641. [PMID: 29668979 PMCID: PMC5905664 DOI: 10.1093/hmg/ddy070] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 02/19/2018] [Indexed: 12/21/2022] Open
Abstract
Mutations in bestrophin-1 (BEST1) are associated with distinct retinopathies, notably three forms with autosomal dominant inheritance and one condition with an autosomal recessive mode of transmission. The molecular mechanisms underlying their distinct retinal phenotypes are mostly unknown. Although heterozygous missense mutations in BEST1 reveal dominant-negative effects in patients with autosomal dominant Best disease (BD), heterozygous mutations associated with autosomal recessive bestrophinopathy (ARB) display no disease phenotype. Here we show that the recessive mutations trigger a strong and fast protein degradation process in the endoplasmic reticulum (ER), thereby favoring a decreased stoichiometry of mutant versus normal BEST1 subunits in the assembly of the homo-pentameric BEST1 chloride channel. In contrast, dominant mutations escape ER-associated degradation and are subjected to a slightly delayed post-ER degradation via the endo-lysosomal degradation pathway. As a result, increased formation of a non-functional BEST1 channel occurs due to a roughly equimolar incorporation of normal and mutant BEST1 subunits into the channel complex. Taken together, our data provide insight into the molecular pathways of dominantly and recessively acting BEST1 missense mutations suggesting that the site of subcellular protein quality control as well as the rate and degree of mutant protein degradation are ultimately responsible for the distinct retinal disease phenotypes in BD and ARB.
Collapse
Affiliation(s)
- Andrea Milenkovic
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, 93053 Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, 93053 Regensburg, Germany
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
- To whom correspondence should be addressed at: Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauß-Allee 11, D-93053 Regensburg, Germany. Tel: +49 9419445400; Fax: +49 9419445402;
| |
Collapse
|
27
|
Hanif AM, Yan J, Jain N. Pattern Dystrophy: An Imprecise Diagnosis in the Age of Precision Medicine. Int Ophthalmol Clin 2019; 59:173-194. [PMID: 30585925 DOI: 10.1097/iio.0000000000000262] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
|
28
|
Kittredge A, Ji C, Zhang Y, Yang T. Differentiation, Maintenance, and Analysis of Human Retinal Pigment Epithelium Cells: A Disease-in-a-dish Model for BEST1 Mutations. J Vis Exp 2018. [PMID: 30199040 DOI: 10.3791/57791] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Although over 200 genetic mutations in the human BEST1 gene have been identified and linked to retinal degenerative diseases, the pathological mechanisms remain elusive mainly due to the lack of a good in vivo model for studying BEST1 and its mutations under physiological conditions. BEST1 encodes an ion channel, namely BESTROPHIN1 (BEST1), which functions in retinal pigment epithelium (RPE); however, the extremely limited accessibility to native human RPE cells represents a major challenge for scientific research. This protocol describes how to generate human RPEs bearing BEST1 disease-causing mutations by induced differentiation from human pluripotent stem cells (hPSCs). As hPSCs are self-renewable, this approach allows researchers to have a steady source of hPSC-RPEs for various experimental analyses, such as immunoblotting, immunofluorescence, and patch clamp, and thus provides a very powerful disease-in-a-dish model for BEST1-associated retinal conditions. Notably, this strategy can be applied to study RPE (patho)physiology and other genes of interest natively expressed in RPE.
Collapse
Affiliation(s)
- Alec Kittredge
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry
| | - Changyi Ji
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry
| | - Yu Zhang
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry;
| | - Tingting Yang
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry;
| |
Collapse
|
29
|
Next generation sequencing identifies novel disease-associated BEST1 mutations in Bestrophinopathy patients. Sci Rep 2018; 8:10176. [PMID: 29976937 PMCID: PMC6033935 DOI: 10.1038/s41598-018-27951-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/13/2018] [Indexed: 12/30/2022] Open
Abstract
Bestinopathies are a spectrum of retinal disorders associated with mutations in BEST1 including autosomal recessive bestrophinopathy (ARB) and autosomal dominant Best vitelliform macular dystrophy (BVMD). We applied whole-exome sequencing on four unrelated Indian families comprising eight affected and twelve unaffected individuals. We identified five mutations in BEST1, including p.Tyr131Cys in family A, p.Arg150Pro in family B, p.Arg47His and p.Val216Ile in family C and p.Thr91Ile in family D. Among these, p.Tyr131Cys, p.Arg150Pro and p.Val216Ile have not been previously reported. Further, the inheritance pattern of BEST1 mutations in the families confirmed the diagnosis of ARB in probands in families A, B and C, while the inheritance of heterozygous BEST1 mutation in family D (p.Thr91Ile) was suggestive of BVMD. Interestingly, the ARB families A and B carry homozygous mutations while family C was a compound heterozygote with a mutation in an alternate BEST1 transcript isoform, highlighting a role for alternate BEST1 transcripts in bestrophinopathy. In the BVMD family D, the heterozygous BEST1 mutation found in the proband was also found in the asymptomatic parent, suggesting an incomplete penetrance and/or the presence of additional genetic modifiers. Our report expands the list of pathogenic BEST1 genotypes and the associated clinical diagnosis.
Collapse
|
30
|
Wang XN, You QS, Li Q, Li Y, Mao Y, Hu F, Zhao HY, Tsai FF, Peng XY. Findings of Optical Coherence Tomography Angiography in Best Vitelliform Macular Dystrophy. Ophthalmic Res 2018; 60:214-220. [PMID: 29656284 DOI: 10.1159/000487488] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/07/2018] [Indexed: 01/15/2023]
Abstract
PURPOSE To evaluate the vascular anatomy of eyes with Best vitelliform macular dystrophy (BVMD) using optical coherence tomography angiography (OCTA). METHODS This retrospective case-control study enrolled 11 consecutive BVMD patients and 13 age-matched healthy participants. Both eyes of each participant were imaged using a macular OCTA scan (3 × 3 mm) by 70-kHz 840-nm spectral-domain OCT. The flow signal was calculated using the split-spectrum amplitude-decorrelation angiography (SSADA) algorithm. RESULTS Qualitative analysis showed uneven hypo- and hyperintense signal changes at the choriocapillary level in OCTA images of BVMD patients. Quantitative analysis showed significant reductions in the flow density of the superficial vascular layer (whole: 49.2% vs. 53.9%, p < 0.001) and the choriocapillary flow area (5.1 vs. 5.5 mm2, p = 0.02) in BVMD patients compared to controls. The choriocapillary flow area in the postvitelliform group was reduced compared to that of the vitelliform group. There was a statistically significant association between choriocapillary flow area and superficial vascular flow density (p = 0.045), choriocapillary flow area and foveal avascular zone area (p = 0.03). CONCLUSIONS Vascular changes in BVMD were apparent in the choriocapillary layer. The changes became more striking in late stages of the disease. OCTA provides useful quantitative measurements for staging and monitoring the progression of BVMD.
Collapse
Affiliation(s)
- Xiao Na Wang
- Beijing Institute of Ophthalmology, Beijing Tong ren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tong ren Hospital, Capital Medical University, Beijing, China
| | - Qi Sheng You
- Beijing Institute of Ophthalmology, Beijing Tong ren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tong ren Hospital, Capital Medical University, Beijing, China
| | - Qian Li
- Beijing Institute of Ophthalmology, Beijing Tong ren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tong ren Hospital, Capital Medical University, Beijing, China
| | - Yang Li
- Beijing Institute of Ophthalmology, Beijing Tong ren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tong ren Hospital, Capital Medical University, Beijing, China
| | - Yu Mao
- Beijing Institute of Ophthalmology, Beijing Tong ren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tong ren Hospital, Capital Medical University, Beijing, China
| | - Feng Hu
- Beijing Institute of Ophthalmology, Beijing Tong ren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tong ren Hospital, Capital Medical University, Beijing, China
| | - Hui Ying Zhao
- Beijing Institute of Ophthalmology, Beijing Tong ren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tong ren Hospital, Capital Medical University, Beijing, China
| | - Frank F Tsai
- Sharp Rees Stealy Medical Group, San Diego, California, USA
| | - Xiao Yan Peng
- Beijing Institute of Ophthalmology, Beijing Tong ren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tong ren Hospital, Capital Medical University, Beijing,
| |
Collapse
|
31
|
|
32
|
Marmorstein AD, Johnson AA, Bachman LA, Andrews-Pfannkoch C, Knudsen T, Gilles BJ, Hill M, Gandhi JK, Marmorstein LY, Pulido JS. Mutant Best1 Expression and Impaired Phagocytosis in an iPSC Model of Autosomal Recessive Bestrophinopathy. Sci Rep 2018. [PMID: 29540715 PMCID: PMC5852082 DOI: 10.1038/s41598-018-21651-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autosomal recessive bestrophinopathy (ARB) is caused by mutations in the gene BEST1 which encodes bestrophin 1 (Best1), an anion channel expressed in retinal pigment epithelial (RPE) cells. It has been hypothesized that ARB represents the human null phenotype for BEST1 and that this occurs due to nonsense mediated decay (NMD). To test this hypothesis, we generated induced pluripotent stem cells (iPSCs) from a patient with ARB and her parents. After differentiation to retinal pigment epithelial (iPSC-RPE) cells, both BEST1 mRNA and Best1 protein expression were compared to controls. BEST1 mRNA expression levels, determined by quantitative PCR, were similar in ARB iPSC-RPE, parental cells, and genetically unrelated controls. Western blotting revealed that CRALBP and RPE65 were expressed within the range delineated by unrelated controls in iPSC-RPE from the ARB donor and her parents. Best1 protein was detected in different clones of ARB iPSC-RPE, but at reduced levels compared to all controls. When tested for the ability to phagocytose photoreceptor outer segments, ARB iPSC-RPE exhibited impaired internalization. These data suggest that impaired phagocytosis is a trait common to the bestrophinopathies. Furthermore, ARB is not universally the result of NMD and ARB, in this patient, is not due to the absence of Best1.
Collapse
Affiliation(s)
- Alan D Marmorstein
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Adiv A Johnson
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lori A Bachman
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Travis Knudsen
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Benjamin J Gilles
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Matthew Hill
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jarel K Gandhi
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lihua Y Marmorstein
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jose S Pulido
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
33
|
Li Y, Zhang Y, Xu Y, Kittredge A, Ward N, Chen S, Tsang SH, Yang T. Patient-specific mutations impair BESTROPHIN1's essential role in mediating Ca 2+-dependent Cl - currents in human RPE. eLife 2017; 6. [PMID: 29063836 PMCID: PMC5655127 DOI: 10.7554/elife.29914] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/13/2017] [Indexed: 01/06/2023] Open
Abstract
Mutations in the human BEST1 gene lead to retinal degenerative diseases displaying progressive vision loss and even blindness. BESTROPHIN1, encoded by BEST1, is predominantly expressed in retinal pigment epithelium (RPE), but its physiological role has been a mystery for the last two decades. Using a patient-specific iPSC-based disease model and interdisciplinary approaches, we comprehensively analyzed two distinct BEST1 patient mutations, and discovered mechanistic correlations between patient clinical phenotypes, electrophysiology in their RPEs, and the structure and function of BESTROPHIN1 mutant channels. Our results revealed that the disease-causing mechanism of BEST1 mutations is centered on the indispensable role of BESTROPHIN1 in mediating the long speculated Ca2+-dependent Cl- current in RPE, and demonstrate that the pathological potential of BEST1 mutations can be evaluated and predicted with our iPSC-based 'disease-in-a-dish' approach. Moreover, we demonstrated that patient RPE is rescuable with viral gene supplementation, providing a proof-of-concept for curing BEST1-associated diseases.
Collapse
Affiliation(s)
- Yao Li
- Jonas Children's Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology and Pathology & Cell Biology, Edward S. Harkness Eye Institute, New York Presbyterian Hospital/Columbia University, New York, United States
| | - Yu Zhang
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, United States
| | - Yu Xu
- Jonas Children's Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology and Pathology & Cell Biology, Edward S. Harkness Eye Institute, New York Presbyterian Hospital/Columbia University, New York, United States.,Department of Ophthalmology, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Alec Kittredge
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, United States
| | - Nancy Ward
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, United States
| | - Shoudeng Chen
- Molecular Imaging Center, Department of Experimental Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Stephen H Tsang
- Jonas Children's Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology and Pathology & Cell Biology, Edward S. Harkness Eye Institute, New York Presbyterian Hospital/Columbia University, New York, United States
| | - Tingting Yang
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, United States
| |
Collapse
|
34
|
Hardin JS, Schaefer GB, Sallam AB, Williams MK, Uwaydat S. A unique case series of autosomal recessive bestrophinopathy exhibiting multigenerational inheritance. Ophthalmic Genet 2017; 38:570-574. [PMID: 28481155 DOI: 10.1080/13816810.2017.1318926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Autosomal recessive bestrophinopathy (ARB) is a retinal disease caused by biallelic mutations of the BEST1 gene. It has a variable phenotype with white flecks in the retina, multifocal yellow subretinal deposits, macular edema, choroidal neovascularization, hyperopia, and electrophysiological abnormalities. We describe a family with ARB and multigenerational inheritance. METHODS Three generations of a Middle Eastern family (a woman, one son, and two grandchildren) were evaluated by our ocular genetics team. Eye examinations, fundus photography, and optical coherence tomography (OCT) were performed. Genetic testing was obtained on examined patients and available relatives. RESULTS The proband demonstrated counting fingers vision and white flecks in the retinal periphery, with macular subretinal fluid (SRF), loss of outer photoreceptor segments, and epiretinal membrane (ERM) on OCT. Two grandchildren demonstrated decreased vision, multifocal yellow subretinal deposits, and SRF on OCT. Two grandchildren examined elsewhere were reported to be similarly affected. A son's examination was normal except for extra-macular scars (from prior toxoplasmosis) and ERM. Genetic history revealed consanguinity and testing showed homozygosity for BEST1 mutations in the proband and two grandchildren c.473G>A/c.473G>A (R218H /R218H) and heterozygosity in two unaffected sons and two unaffected daughters-in-law c.473G>A/WT (p.R218H/WT). DISCUSSION We present a consanguineous family of five affected individuals with ARB and four confirmed carriers. Their pedigree was consistent with dominant inheritance and incomplete penetrance. Genetic testing clarified the diagnosis and mode of inheritance. We describe the genetic findings, phenotypic variability, and recessive inheritance of an often dominantly inherited mutation as notable elements in their case.
Collapse
Affiliation(s)
- Joshua S Hardin
- a Jones Eye Institute, University of Arkansas for Medical Sciences , Little Rock , Arkansas , USA
| | - G Bradley Schaefer
- b Section of Genetics and Metabolism , University of Arkansas for Medical Sciences , Little Rock , Arkansas , USA
| | - Ahmed B Sallam
- a Jones Eye Institute, University of Arkansas for Medical Sciences , Little Rock , Arkansas , USA
| | - M Kathryn Williams
- b Section of Genetics and Metabolism , University of Arkansas for Medical Sciences , Little Rock , Arkansas , USA
| | - Sami Uwaydat
- a Jones Eye Institute, University of Arkansas for Medical Sciences , Little Rock , Arkansas , USA
| |
Collapse
|
35
|
Qian CX, Charran D, Strong CR, Steffens TJ, Jayasundera T, Heckenlively JR. Optical Coherence Tomography Examination of the Retinal Pigment Epithelium in Best Vitelliform Macular Dystrophy. Ophthalmology 2017; 124:456-463. [DOI: 10.1016/j.ophtha.2016.11.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/15/2016] [Accepted: 11/15/2016] [Indexed: 10/20/2022] Open
|
36
|
A NOVEL P.ASP304GLY MUTATION IN BEST1 GENE ASSOCIATED WITH ATYPICAL BEST VITELLIFORM MACULAR DYSTROPHY PHENOTYPE AND HIGH INTRAFAMILIAL VARIABILITY. Retina 2017; 36:1733-40. [PMID: 26807628 DOI: 10.1097/iae.0000000000000966] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE To report the atypical phenotypic characteristics of patients with a novel p.Asp304Gly mutation in BEST1. METHODS Affected individuals underwent a complete ophthalmic examination, including best-corrected visual acuity, fundus autofluorescence, spectral domain optical coherence tomography, and electrophysiologic testing. All individuals were screened for mutations in the BEST1 gene. RESULTS Five patients of the same Italian family were clinically examined. All patients complained of decreased vision as the initial symptom. Best-corrected visual acuity ranged from 20/800 to 20/32. On fundus examination, all patients showed atypical Best vitelliform macular dystrophy phenotype with multifocal macular and extramacular involvement. The spectral domain optical coherence tomography characteristics of central macular and extramacular lesions varied in each patient and included "giant" choroidal excavation, extensive flat macular elevation with hyporeflective subretinal material accumulation surrounded by hyperautofluorescent spots/annulus, and extensive hypoautofluorescent extramacular atrophic areas. Electrooculogram was always abnormal with Arden ratio lower than 1.55, whereas electroretinogram was normal in the two younger patients and abnormal (low amplitude) in the three older patients. Genetic analysis revealed a novel missense mutation in BEST1, substituting aspartate for glycine at amino acid 304. CONCLUSION We describe the atypical phenotype and high intrafamilial variability associated with a new mutation in the BEST1 gene in an Italian family affected with Best vitelliform macular dystrophy. Clinicians should consider screening the BEST1 gene even in the absence of the typical phenotype and in case of high intrafamilial variability.
Collapse
|
37
|
Caceres PS, Benedicto I, Lehmann GL, Rodriguez-Boulan EJ. Directional Fluid Transport across Organ-Blood Barriers: Physiology and Cell Biology. Cold Spring Harb Perspect Biol 2017; 9:a027847. [PMID: 28003183 PMCID: PMC5334253 DOI: 10.1101/cshperspect.a027847] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Directional fluid flow is an essential process for embryo development as well as for organ and organism homeostasis. Here, we review the diverse structure of various organ-blood barriers, the driving forces, transporters, and polarity mechanisms that regulate fluid transport across them, focusing on kidney-, eye-, and brain-blood barriers. We end by discussing how cross talk between barrier epithelial and endothelial cells, perivascular cells, and basement membrane signaling contribute to generate and maintain organ-blood barriers.
Collapse
Affiliation(s)
- Paulo S Caceres
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Ignacio Benedicto
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Guillermo L Lehmann
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Enrique J Rodriguez-Boulan
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
38
|
Johnson AA, Guziewicz KE, Lee CJ, Kalathur RC, Pulido JS, Marmorstein LY, Marmorstein AD. Bestrophin 1 and retinal disease. Prog Retin Eye Res 2017; 58:45-69. [PMID: 28153808 DOI: 10.1016/j.preteyeres.2017.01.006] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 12/18/2022]
Abstract
Mutations in the gene BEST1 are causally associated with as many as five clinically distinct retinal degenerative diseases, which are collectively referred to as the "bestrophinopathies". These five associated diseases are: Best vitelliform macular dystrophy, autosomal recessive bestrophinopathy, adult-onset vitelliform macular dystrophy, autosomal dominant vitreoretinochoroidopathy, and retinitis pigmentosa. The most common of these is Best vitelliform macular dystrophy. Bestrophin 1 (Best1), the protein encoded by the gene BEST1, has been the subject of a great deal of research since it was first identified nearly two decades ago. Today we know that Best1 functions as both a pentameric anion channel and a regulator of intracellular Ca2+ signaling. Best1 is an integral membrane protein which, within the eye, is uniquely expressed in the retinal pigment epithelium where it predominantly localizes to the basolateral plasma membrane. Within the brain, Best1 expression has been documented in both glial cells and astrocytes where it functions in both tonic GABA release and glutamate transport. The crystal structure of Best1 has revealed critical information about how Best1 functions as an ion channel and how Ca2+ regulates that function. Studies using animal models have led to critical insights into the physiological roles of Best1 and advances in stem cell technology have allowed for the development of patient-derived, "disease in a dish" models. In this article we review our knowledge of Best1 and discuss prospects for near-term clinical trials to test therapies for the bestrophinopathies, a currently incurable and untreatable set of diseases.
Collapse
Affiliation(s)
- Adiv A Johnson
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA; Nikon Instruments, Melville, NY, USA
| | - Karina E Guziewicz
- Department of Clinical Studies-Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C Justin Lee
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Ravi C Kalathur
- New York Structural Biology Center, New York Consortium on Membrane Protein Structure, New York, NY, USA
| | - Jose S Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
39
|
Flat Anterior Chamber after Trabeculectomy in Secondary Angle-Closure Glaucoma with BEST1 Gene Mutation: Case Series. PLoS One 2017; 12:e0169395. [PMID: 28056057 PMCID: PMC5215797 DOI: 10.1371/journal.pone.0169395] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/12/2016] [Indexed: 12/25/2022] Open
Abstract
Purpose Trabeculectomy has been regarded as a mainstay of initial treatment in eyes of angle closure glaucoma (ACG) with peripheral anterior synechia > 180° in the Chinese population while its efficacy in secondary ACG with BEST1 gene mutation remains unclear. We set out to investigate the treatment outcome of trabeculectomy for secondary ACG in a group of patients with autosomal recessive bestrophinopathy (ARB). Methods In this retrospective case series study, 8 secondary ACG patients with ARB and their 4 recruited family members underwent a thorough ophthalmic examination including best-corrected visual acuity, Goldmann applanation tonometry, gonioscopy, and fundus examinations. Ultrasound biomicroscopy, optical coherence tomography (OCT), ultrasound A-scan, B-scan, electro-oculography (EOG), Humphrey perimetry, fundus photography, fundus fluorescein angiography (FFA) and indocyanine green angiography (ICGA) were also performed. Blood samples were obtained in the patients and their available family members to analyze the variants of the BEST1 gene. Trabeculectomy was performed in the 8 patients (15 eyes). Results The age of onset varied from 13 to 38 years. The average axial length (AL) of the affected eyes was 21.82 ± 0.92 mm and the average anterior chamber depth (ACD) was 2.19 ± 0.29 mm. There was marked axial shallowing of the anterior chamber in all 15 eyes after trabeculectomy, and was not improved with potent mydriatics. The IOP was elevated in 3 eyes. Variable degree of yellowish subretinal deposits was observed in the posterior retina. The FFA showed punctuate or patched hyperfluorescence suggesting retinal pigment epithelium impairment. The ICGA demonstrated dilatation of choroidal vessels. The OCT revealed diffused neuroretinal detachment in the posterior and midperipheral retina, with intraretinal fluid collections, and hyperreflective subretinal accumulations. The average subfoveal choroidal thickness of the patients was 382.36 ± 80.09 μm. All the patients and enrolled family members carried mutation in BEST1 gene. Conclusions ARB is a rare condition with fundus manifestations mimicking various diseases. Careful discrimination should be taken to exclude any secondary causes for ACG before treatment. Concerning the high incidence of postoperative shallow anterior chamber, selection of filtering surgery should be very careful in these patients.
Collapse
|
40
|
Effects of Ca 2+ ions on bestrophin-1 surface films. Colloids Surf B Biointerfaces 2016; 149:226-232. [PMID: 27768912 DOI: 10.1016/j.colsurfb.2016.10.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/07/2016] [Accepted: 10/12/2016] [Indexed: 01/01/2023]
Abstract
Human bestrophin-1 (hBest1) is a transmembrane calcium-activated chloride channel protein - member of the bestrophin family of anion channels, predominantly expressed in the membrane of retinal pigment epithelium (RPE) cells. Mutations in the protein cause ocular diseases, named Bestrophinopathies. Here, we present the first Fourier transform infrared (FTIR) study of the secondary structure elements of hBest1, π/A isotherms and hysteresis, Brewster angle microscopy (BAM) and atomic force microscopy (AFM) visualization of the aggregation state of protein molecules dispersed as Langmuir and Langmuir-Blodgett films. The secondary structure of hBest1 consists predominantly of 310-helices (27.2%), α-helixes (16.3%), β-turns and loops (32.2%). AFM images of hBest1 suggest approximate lateral dimensions of 100×160Å and 75Å height. Binding of calcium ions (Ca2+) induces conformational changes in the protein secondary structure leading to assembly of protein molecules and changes in molecular and macro-organization of hBest1 in monolayers. These data provide basic information needed in pursuit of molecular mechanisms underlying retinal and other pathologies linked to this protein.
Collapse
|
41
|
Moshfegh Y, Velez G, Li Y, Bassuk AG, Mahajan VB, Tsang SH. BESTROPHIN1 mutations cause defective chloride conductance in patient stem cell-derived RPE. Hum Mol Genet 2016; 25:2672-2680. [PMID: 27193166 DOI: 10.1093/hmg/ddw126] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/13/2016] [Accepted: 04/18/2016] [Indexed: 12/20/2022] Open
Abstract
Bestrophin1 (BEST1) is expressed in human retinal pigment epithelium (RPE) and mutations in the BEST1 gene commonly cause retinal dysfunction and macular degeneration. BEST1 is presumed to assemble into a calcium-activated chloride channel and be involved in chloride transport but there is no direct evidence in live human RPE cells to support this idea. To test whether BEST1 functions as a chloride channel in living tissue, BEST1-mutant RPE (R218H, L234P, A243T) were generated from patient-derived induced pluripotent stem cells and compared with wild-type RPE in a retinal environment, using a biosensor that visualizes calcium-induced chloride ion flux in the cell. Calcium stimulation elicited chloride ion export in normal RPE but not in RPE derived from three patients with BEST1 mutations. These data, along with three-dimensional modeling, provide evidence that BEST1 assembles into a key calcium-sensing chloride channel in human RPE.
Collapse
Affiliation(s)
- Yasmin Moshfegh
- Barbara & Donald Jonas Laboratory of Regenerative Medicine, and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Pathology & Cell Biology, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Gabriel Velez
- Omics Lab.,Department of Ophthalmology and Visual Sciences.,Medical Scientist Training Program
| | - Yao Li
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | | | - Vinit B Mahajan
- Department of Ophthalmology and Visual Sciences.,Medical Scientist Training Program
| | - Stephen H Tsang
- Barbara & Donald Jonas Laboratory of Regenerative Medicine, and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Pathology & Cell Biology, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA .,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| |
Collapse
|
42
|
Dalvin LA, Pulido JS, Marmorstein AD. Vitelliform dystrophies: Prevalence in Olmsted County, Minnesota, United States. Ophthalmic Genet 2016; 38:143-147. [PMID: 27120116 DOI: 10.1080/13816810.2016.1175645] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Vitelliform dystrophies are a group of macular degenerative diseases characterized by round yellow lesions in the macula. While often idiopathic, vitelliform dystrophies include inherited maculopathies such as Best disease and some cases of pattern dystrophy. The prevalence of vitelliform dystrophies in the United States has not been reported. This study examined the prevalence of vitelliform dystrophies in Olmsted County, Minnesota. MATERIALS AND METHODS The Rochester Epidemiology Project database was used to identify all cases of vitelliform or pattern dystrophy in Olmsted County from 1 January 2000-31 December 2014. RESULTS Overall, 27 patients had true vitelliform lesions, indicating a prevalence of 1 in 5500. Of these, two had genetically confirmed Best disease, and an additional five to seven carried a diagnosis of Best disease, which chart reviews confirmed as probable cases; 18-20 patients had adult-onset vitelliform macular dystrophy. The prevalence of Best disease was 1 in 16,500 to 1 in 21,000. Adult-onset vitelliform macular dystrophy was found in 1 in 7400 to 1 in 8200. CONCLUSIONS Vitelliform dystrophies affect 1 in 5500 individuals in Olmsted County. While the values in this study provide good estimates for the prevalence of Best disease versus adult-onset vitelliform macular dystrophy, the results are limited by dependence on diagnoses made by other ophthalmologists and underutilization of genetic testing. Thus, these diseases should be thought of as at least as prevalent as reported here. As therapies for Best disease and other macular degenerative diseases are quickly becoming a reality, genetic testing should be employed as the gold standard for diagnosis of these diseases.
Collapse
Affiliation(s)
- Lauren A Dalvin
- a Department of Ophthalmology , Mayo Clinic , Rochester , Minnesota , USA
| | - Jose S Pulido
- a Department of Ophthalmology , Mayo Clinic , Rochester , Minnesota , USA.,b Department of Molecular Medicine , Mayo Clinic , Rochester , Minnesota , USA
| | - Alan D Marmorstein
- a Department of Ophthalmology , Mayo Clinic , Rochester , Minnesota , USA
| |
Collapse
|
43
|
|
44
|
Dalvin LA, Abou Chehade JE, Chiang J, Fuchs J, Iezzi R, Marmorstein AD. Retinitis pigmentosa associated with a mutation in BEST1. Am J Ophthalmol Case Rep 2016; 2:11-17. [PMID: 29503890 PMCID: PMC5757359 DOI: 10.1016/j.ajoc.2016.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/18/2016] [Accepted: 03/26/2016] [Indexed: 11/24/2022] Open
Abstract
Purpose There is only one prior report associating mutations in BEST1 with a diagnosis of retinitis pigmentosa (RP). The imaging studies presented in that report were more atypical of RP and shared features of autosomal recessive bestrophinopathy and autosomal dominant vitreoretinochoroidopathy. Here, we present a patient with a clinical phenotype consistent with classic features of RP. Observations The patient in this report was diagnosed with simplex RP based on clinically-evident bone spicules with characteristic ERG and EOG findings. The patient had associated massive cystoid macular edema which resolved following a short course of oral acetazolamide. Genetic testing revealed that the patient carries a novel heterozygous deletion mutation in BEST1 which is not carried by either parent. While this suggests BEST1 is causative, the patient also inherited heterozygous copies of several mutations in other genes known to cause recessive retinal degenerative disease. Conclusions and Importance How some mutations in BEST1 associate with peripheral retinal degeneration phenotypes, while others manifest as macular degeneration phenotypes is currently unknown. We speculate that RP due to BEST1 mutation requires mutations in other modifier genes.
Collapse
Affiliation(s)
- Lauren A Dalvin
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| | - Jackson E Abou Chehade
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| | - John Chiang
- CEI Diagnostic Laboratory, Casey Eye Institute, 3375 SW Terwilliger Blvd, Portland, OR, 97239, United States
| | - Josefine Fuchs
- Department of Ophthalmology, Rigshospitalet, Blegdamsvej 9, 2100, København Ø, Denmark
| | - Raymond Iezzi
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| | - Alan D Marmorstein
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| |
Collapse
|
45
|
Sparrow JR, Duncker T, Woods R, Delori FC. Quantitative Fundus Autofluorescence in Best Vitelliform Macular Dystrophy: RPE Lipofuscin is not Increased in Non-Lesion Areas of Retina. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 854:285-90. [PMID: 26427423 PMCID: PMC5779626 DOI: 10.1007/978-3-319-17121-0_38] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the lipofuscin of retinal pigment epithelial (RPE) cells has been implicated in the pathogenesis of Best vitelliform macular dystrophy, we quantified fundus autofluorescence (quantitative fundus autofluorescence, qAF) as an indirect measure of RPE lipofuscin levels. Mean non-lesion qAF was found to be within normal limits for age. By spectral domain optical coherence tomography (SD-OCT) vitelliform lesions presented as fluid-filled subretinal detachments containing reflective material. We discuss photoreceptor outer segment debris as the source of the intense fluorescence of these lesions and loss of anion channel functioning as an explanation for the bullous photoreceptor-RPE detachment. Unexplained is the propensity of the disease for central retina.
Collapse
Affiliation(s)
- Janet R Sparrow
- Department of Ophthalmology, Harkness Eye Institute, Columbia University Medical Center, 635 W. 165th Street, 10032, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, 635 W. 165th Street, 10032, New York, NY, USA.
| | - Tobias Duncker
- Department of Ophthalmology, Harkness Eye Institute, Columbia University Medical Center, 635 W. 165th Street, 10032, New York, NY, USA.
| | - Russell Woods
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute, 02114, Boston, MA, USA.
| | - François C Delori
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute, 02114, Boston, MA, USA.
| |
Collapse
|
46
|
Giblin JP, Comes N, Strauss O, Gasull X. Ion Channels in the Eye: Involvement in Ocular Pathologies. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 104:157-231. [PMID: 27038375 DOI: 10.1016/bs.apcsb.2015.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The eye is the sensory organ of vision. There, the retina transforms photons into electrical signals that are sent to higher brain areas to produce visual sensations. In the light path to the retina, different types of cells and tissues are involved in maintaining the transparency of avascular structures like the cornea or lens, while others, like the retinal pigment epithelium, have a critical role in the maintenance of photoreceptor function by regenerating the visual pigment. Here, we have reviewed the roles of different ion channels expressed in ocular tissues (cornea, conjunctiva and neurons innervating the ocular surface, lens, retina, retinal pigment epithelium, and the inflow and outflow systems of the aqueous humor) that are involved in ocular disease pathophysiologies and those whose deletion or pharmacological modulation leads to specific diseases of the eye. These include pathologies such as retinitis pigmentosa, macular degeneration, achromatopsia, glaucoma, cataracts, dry eye, or keratoconjunctivitis among others. Several disease-associated ion channels are potential targets for pharmacological intervention or other therapeutic approaches, thus highlighting the importance of these channels in ocular physiology and pathophysiology.
Collapse
Affiliation(s)
- Jonathan P Giblin
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Nuria Comes
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Xavier Gasull
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
47
|
Johnson AA, Bachman LA, Gilles BJ, Cross SD, Stelzig KE, Resch ZT, Marmorstein LY, Pulido JS, Marmorstein AD. Autosomal Recessive Bestrophinopathy Is Not Associated With the Loss of Bestrophin-1 Anion Channel Function in a Patient With a Novel BEST1 Mutation. Invest Ophthalmol Vis Sci 2015. [PMID: 26200502 DOI: 10.1167/iovs.15-16910] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
PURPOSE Mutations in BEST1, encoding bestrophin-1 (Best1), cause autosomal recessive bestrophinopathy (ARB). Encoding bestrophin-1 is a pentameric anion channel localized to the basolateral plasma membrane of the RPE. Here, we characterize the effects of the mutations R141H (CGC > CAC) and I366fsX18 (c.1098_1100+7del), identified in a patient in our practice, on Best1 trafficking, oligomerization, and channel activity. METHODS Currents of Cl- were assessed in transfected HEK293 cells using whole-cell patch clamp. Best1 localization was assessed by confocal microscopy in differentiated, human-induced pluripotent stem cell-derived RPE (iPSC-RPE) cells following expression of mutants via adenovirus-mediated gene transfer. Oligomerization was evaluated by coimmunoprecipitation in iPSC-RPE and MDCK cells. RESULTS Compared to Best1, Best1 I366fsX18 currents were increased while Best1 R141H Cl- currents were diminished. Coexpression of Best1 R141H with Best1 or Best1 I366fsX18 resulted in rescued channel activity. Overexpressed Best1, Best1 R141H, and Best1 I366fsX18 were all properly localized in iPSC-RPE cells; Best1 R141H and Best1 I366fsX18 coimmunoprecipitated with endogenous Best1 in iPSC-RPE cells and with each other in MDCK cells. CONCLUSIONS The first 366 amino acids of Best1 are sufficient to mediate channel activity and homo-oligomerization. The combination of Best1 and Best1 R141H does not cause disease, while Best1 R141H together with Best1 I366fsX18 causes ARB. Since both combinations generate comparable Cl- currents, this indicates that ARB in this patient is not caused by a loss of channel activity. Moreover, Best1 I366fsX18 differs from Best1 in that it lacks most of the cytosolic C-terminal domain, suggesting that the loss of this region contributes significantly to the pathogenesis of ARB in this patient.
Collapse
Affiliation(s)
- Adiv A Johnson
- Department of Ophthalmology Mayo Clinic, Rochester, Minnesota, United States
| | - Lori A Bachman
- Department of Ophthalmology Mayo Clinic, Rochester, Minnesota, United States
| | - Benjamin J Gilles
- Department of Ophthalmology Mayo Clinic, Rochester, Minnesota, United States
| | - Samuel D Cross
- Department of Ophthalmology Mayo Clinic, Rochester, Minnesota, United States
| | - Kimberly E Stelzig
- Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Zachary T Resch
- Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Lihua Y Marmorstein
- Department of Ophthalmology Mayo Clinic, Rochester, Minnesota, United States
| | - Jose S Pulido
- Department of Ophthalmology Mayo Clinic, Rochester, Minnesota, United States 3Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Alan D Marmorstein
- Department of Ophthalmology Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
48
|
BEST1: the Best Target for Gene and Cell Therapies. Mol Ther 2015; 23:1805-9. [PMID: 26388462 DOI: 10.1038/mt.2015.177] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/14/2015] [Indexed: 12/14/2022] Open
Abstract
A retinal pigmented epithelial (RPE) disorder, bestrophinopathy has recently been proven to be amenable to gene and cell-based therapies in preclinical models. RPE disorders and allied retinal degenerations exhibit significant genetic heterogeneity, and diverse mutations can result in similar disease phenotypes. Several RPE disorders have recently become targets for gene therapies in humans. The year 2011 brought a new advance in cell-based therapies, with the Food and Drug Administration approving clinical trials using embryonic stem cells for an RPE disorder known as age-related macular degeneration. Recent studies on induced pluripotent stem (iPS)-RPE generation indicate strong potential for developing patient-specific disease models in vitro, which could eventually enable personalized treatment. This mini-review will briefly highlight the suitability of the retina for gene and cell therapies, the pathophysiology of bestrophinopathy, and the research and treatment opportunities afforded by stem cell and genetic therapies.
Collapse
|
49
|
Marmorstein AD, Kinnick TR, Stanton JB, Johnson AA, Lynch RM, Marmorstein LY. Bestrophin-1 influences transepithelial electrical properties and Ca2+ signaling in human retinal pigment epithelium. Mol Vis 2015; 21:347-59. [PMID: 25878489 PMCID: PMC4390793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/30/2015] [Indexed: 11/16/2022] Open
Abstract
PURPOSE Mutations in BEST1, encoding Bestrophin-1 (Best1), cause Best vitelliform macular dystrophy (BVMD) and other inherited retinal degenerative diseases. Best1 is an integral membrane protein localized to the basolateral plasma membrane of the retinal pigment epithelium (RPE). Data from numerous in vitro and in vivo models have demonstrated that Best1 regulates intracellular Ca2+ levels. Although it is known from in vitro and crystal structure data that Best1 is also a calcium-activated anion channel, evidence for Best1 functioning as a channel in human RPE is lacking. To assess Best1-associated channel activity in the RPE, we examined the transepithelial electrical properties of fetal human RPE (fhRPE) cells, which express endogenous Best1. METHODS Using adenovirus-mediated gene transfer, we overexpressed Best1 and the BVMD mutant Best1W93C in fhRPE cells and assessed resting transepithelial potential (TEP), transepithelial resistance, short circuit current (Isc), and intracellular Ca2+ levels. Cl- currents were directly measured in transfected HEK293 cells using whole-cell patch clamp. RESULTS Best1W93C showed ablated Cl- currents and, when co-expressed, suppressed the channel activity of Best1 in HEK293 cells. In fhRPE, overexpression of Best1 increased TEP and Isc, while Best1W93C diminished TEP and Isc. Substitution of Cl- in the bath media resulted in a significant reduction of Isc in monolayers overexpressing Best1, but no significant Isc change in monolayers expressing Best1W93C. We removed Ca2+ as a limit on transepithelial electrical properties by treating cells with ionomycin, and found that changes in Isc and TEP for monolayers expressing Best1 were absent in monolayers expressing Best1W93C. Similarly, inhibition of calcium-activated anion channels with niflumic acid reduced both Isc and TEP of control and Best1 monolayers, but did not notably affect Best1W93C monolayers. Stimulation with extracellular ATP induced an increase in TEP in control monolayers that was greater than that observed in those expressing Best1(W93C). Examination of [Ca2+]i following ATP stimulation demonstrated that the expression of Best1W93C impaired intracellular Ca2+ signaling. CONCLUSIONS These data indicate that Best1 activity strongly influences electrophysiology and Ca2+ signaling in RPE cells, and that a common BVMD mutation disrupts both of these parameters. Our findings support the hypothesis that Best1 functions as an anion channel in human RPE.
Collapse
|
50
|
Best vitelliform macular dystrophy: literature review. Open Med (Wars) 2014. [DOI: 10.2478/s11536-013-0333-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AbstractBest vitelliform macular dystrophy (BVD) is a slowly progressive form of macular dystrophy. In most cases this disease begins in childhood although sometimes it can develop in later age. The diagnosis of BVD is based on family history, clinical and electrophysiological findings. Clinical signs are variable, yet the majority of patients have a typical yellow yolk-like macular lesion in the eye fundus. Lesions are usually bilateral, but in rare cases can be unilateral. Atrophy of the macula may develop after many years. The mutations responsible for Best vitelliform macular dystrophy are found in a gene called VMD2, which encodes a transmembrane protein named bestrophin-1 (hBest1) that is a Ca2+-sensitive chloride channel. Most reported cases causing the disease are in exons 2, 4, 6 and 8 in patients with BVD. In this article we discuss the etiology of Best’s vitelliform macular dystrophy, clinical presentation, diagnostics, genetic and current treatment possibilities.
Collapse
|