1
|
Elbagoury NM, Tawfik CA, Abdel-Aleem AF, Fathy HM, Baddar DN, Essawi ML. A novel founder variant in BEST1 gene causing autosomal recessive bestrophinopathy. Orphanet J Rare Dis 2025; 20:248. [PMID: 40414863 PMCID: PMC12103744 DOI: 10.1186/s13023-025-03813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 05/15/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Autosomal recessive bestrophinopathy (ARB) is a rare retinal dystrophy caused by homozygous or compound heterozygous null variants in the BEST1 gene. Clinically, ARB presents with variable features including central visual impairment, global photoreceptor dysfunction (as indicated by abnormal full-field ERG), and a significantly reduced electro-oculogram (EOG) light rise, a hallmark of bestrophinopathy. Fundus examination reveals widespread retinal pigment epithelial (RPE) disturbance, vitelliform deposits in the posterior pole (more clearly visualized with fundus autofluorescence), and macular fluid accumulation. Angle-closure glaucoma, secondary to anterior chamber dysgenesis, is a potential complication. This work aims at documenting the founder effect of a novel variant in the BEST1 gene causing autosomal recessive bestrophinopathy and determining its variable clinical features. METHODS Twelve members of nine unrelated, consanguineous Egyptian families with a history of impaired central vision underwent comprehensive ophthalmological examination, fundus color photography, fundus autofluorescence (FAF), spectral-domain optical coherence tomography (SD-OCT) of the macula, and electrophysiological studies. Variant screening of coding exons of the BEST1 gene and some flanking regions was performed using the Sanger sequencing technique. The pathogenicity of the variants was tested using different in silico functional analysis tools. RESULTS The clinical examination and investigations confirmed the ARB phenotype. All twelve patients exhibited (c.365 G > C, p. Arg122Pro) a novel BEST1 gene variant in a homozygous form. On top of the classical retinal phenotype of ARB, some patients had other ocular associations: four patients were found to have angle-closure glaucoma, one patient had associated corneal dystrophy, one developed a macular hole, and one patient developed uveitis. CONCLUSION The identification of the same, novel homozygous BEST1 missense variant in twelve patients from nine unrelated, consanguineous families of Egyptian origin, suggests a founder effect. Angle-closure glaucoma was the most commonly associated ocular abnormality (30%). Our finding expands the molecular spectrum of ARB-associated variants, and identification of this founder variant can simplify genetic testing in the presence of limited resources and lead to better counseling.
Collapse
Affiliation(s)
- Nagham Maher Elbagoury
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12311, Egypt.
- Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt.
| | - Caroline Atef Tawfik
- Department of Ophthalmology, Ain Shams University, Cairo, Egypt
- Watany Eye Hospital, Cairo, Egypt
| | - Asmaa Fawzy Abdel-Aleem
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12311, Egypt
- Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - Heba Mahmoud Fathy
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12311, Egypt
- Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - Dina Nabil Baddar
- Watany Eye Hospital, Cairo, Egypt
- Research Institute of Ophthalmology, Giza, Egypt
| | - Mona Lotfi Essawi
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12311, Egypt
- Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| |
Collapse
|
2
|
Tan WLA, Neto LRP, Reverter A, McGowan M, Fortes MRS. Sequence level genome-wide associations for bull production and fertility traits in tropically adapted bulls. BMC Genomics 2023; 24:365. [PMID: 37386436 DOI: 10.1186/s12864-023-09475-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/21/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND The genetics of male fertility is complex and not fully understood. Male subfertility can adversely affect the economics of livestock production. For example, inadvertently mating bulls with poor fertility can result in reduced annual liveweight production and suboptimal husbandry management. Fertility traits, such as scrotal circumference and semen quality are commonly used to select bulls before mating and can be targeted in genomic studies. In this study, we conducted genome-wide association analyses using sequence-level data targeting seven bull production and fertility traits measured in a multi-breed population of 6,422 tropically adapted bulls. The beef bull production and fertility traits included body weight (Weight), body condition score (CS), scrotal circumference (SC), sheath score (Sheath), percentage of normal spermatozoa (PNS), percentage of spermatozoa with mid-piece abnormalities (MP) and percentage of spermatozoa with proximal droplets (PD). RESULTS After quality control, 13,398,171 polymorphisms were tested for their associations with each trait in a mixed-model approach, fitting a multi-breed genomic relationship matrix. A Bonferroni genome-wide significance threshold of 5 × 10- 8 was imposed. This effort led to identifying genetic variants and candidate genes underpinning bull fertility and production traits. Genetic variants in Bos taurus autosome (BTA) 5 were associated with SC, Sheath, PNS, PD and MP. Whereas chromosome X was significant for SC, PNS, and PD. The traits we studied are highly polygenic and had significant results across the genome (BTA 1, 2, 4, 6, 7, 8, 11, 12, 14, 16, 18, 19, 23, 28, and 29). We also highlighted potential high-impact variants and candidate genes associated with Scrotal Circumference (SC) and Sheath Score (Sheath), which warrants further investigation in future studies. CONCLUSION The work presented here is a step closer to identifying molecular mechanisms that underpin bull fertility and production. Our work also emphasises the importance of including the X chromosome in genomic analyses. Future research aims to investigate potential causative variants and genes in downstream analyses.
Collapse
Affiliation(s)
- Wei Liang Andre Tan
- School of Chemistry and Molecular Biosciences, The University of Queensland, Chemistry Bld, 68 Cooper Rd, Brisbane City, QLD, 4072, Australia.
| | | | - Antonio Reverter
- CSIRO Agriculture and Food, 306 Carmody Road, St Lucia, QLD, 4067, Australia
| | - Michael McGowan
- School of Veterinary Science, The University of Queensland, Gatton, QLD, 4343, Australia
| | - Marina Rufino Salinas Fortes
- School of Chemistry and Molecular Biosciences, The University of Queensland, Chemistry Bld, 68 Cooper Rd, Brisbane City, QLD, 4072, Australia
| |
Collapse
|
3
|
Wang J, Tao Z, Deng H, Cui Y, Xu Z, Lyu Q, Zhao J. Therapeutic implications of nanodrug and tissue engineering for retinal pigment epithelium-related diseases. NANOSCALE 2022; 14:5657-5677. [PMID: 35352082 DOI: 10.1039/d1nr08337f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The retinal pigment epithelium (RPE), as a single layer of cells that performs multiple functions posteriorly in the eye, is a promising target site for the prevention and treatment of several clinical diseases, including proliferative diabetic retinopathy, age-related macular degeneration, chorionic neovascularization, and retinitis pigmentosa. In recent decades, several nanodrug delivery platforms and tissue-engineered RPE have been widely developed to treat RPE-related diseases. This work summarizes the recent advances in nanoplatforms and tissue engineering scaffolds developed in these fields. The diseases associated with pathological RPE and their common therapy strategies are first introduced. Then, the recent progress made with a variety of drug delivery systems is presented, with an emphasis on the modification strategies of nanomaterials for targeted delivery. Tissue engineering-mediated RPE transplantation for treating these diseases is subsequently described. Finally, the clinical translation challenges in these fields are discussed in depth. This article will offer readers a better understanding of emerging nanotechnology and tissue engineering related to the treatment of RPE-related diseases and could facilitate their widespread use in experiments in vivo and in clinical applications.
Collapse
Affiliation(s)
- Jiao Wang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen Eye Hospital affiliated to Jinan University, School of Optometry, Shenzhen University, Shenzhen 518000, China.
| | - Zhengyang Tao
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen Eye Hospital affiliated to Jinan University, School of Optometry, Shenzhen University, Shenzhen 518000, China.
| | - Hongwei Deng
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen Eye Hospital affiliated to Jinan University, School of Optometry, Shenzhen University, Shenzhen 518000, China.
| | - Yubo Cui
- Department of Ophthalmology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China.
| | - Zhirong Xu
- Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Qinghua Lyu
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen Eye Hospital affiliated to Jinan University, School of Optometry, Shenzhen University, Shenzhen 518000, China.
- Institute of Microscale Optoelectronics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jun Zhao
- Department of Ophthalmology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China.
| |
Collapse
|
4
|
He XS, Ye WL, Zhang YJ, Yang XQ, Liu F, Wang JR, Ding XL, Yang Y, Zhang RN, Zhao YY, Bi HX, Guo LC, Gan WJ, Wu H. Oncogenic potential of BEST4 in colorectal cancer via activation of PI3K/Akt signaling. Oncogene 2022; 41:1166-1177. [PMID: 35058597 DOI: 10.1038/s41388-021-02160-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/02/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022]
Abstract
BEST4 is a member of the bestrophin protein family that plays a critical role in human intestinal epithelial cells. However, its role and mechanism in colorectal cancer (CRC) remain largely elusive. Here, we investigated the role and clinical significance of BEST4 in CRC. Our results demonstrate that BEST4 expression is upregulated in clinical CRC samples and its high-level expression correlates with advanced TNM (tumor, lymph nodes, distant metastasis) stage, LNM (lymph node metastasis), and poor survival. Functional studies revealed that ectopic expression of BEST4 promoted CRC cell proliferation and metastasis, whereas the depletion of BEST4 had the opposite effect both in vitro and in vivo. Mechanistically, BEST4 binds to the p85α regulatory subunit of phosphatidylinositol-3-kinase (PI3K) and promotes p110 kinase activity; this leads to activation of Akt signaling and expression of MYC and CCND1, which are critical regulators of cell proliferation and metastasis. In clinical samples, the expression of BEST4 is positively associated with the expression of phosphorylated Akt, MYC and CCND1. Pharmacological inhibition of Akt activity markedly repressed BEST4-mediated Akt signaling and proliferation and metastasis of CRC cells. Importantly, the interaction between BEST4 and p85α was also enhanced by epidermal growth factor (EGF) in CRC cells. Therapeutically, BEST4 suppression effectively sensitized CRC cells to gefitinib treatment in vivo. Taken together, our findings indicate the oncogenic potential of BEST4 in colorectal carcinogenesis and metastasis by modulating BEST4/PI3K/Akt signaling, highlighting a potential strategy for CRC therapy.
Collapse
Affiliation(s)
- Xiao-Shun He
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Wen-Long Ye
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yu-Juan Zhang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Xiao-Qin Yang
- Department of Bioinformatics, Medical College of Soochow University, Soochow University, Suzhou, 215123, China
| | - Feng Liu
- Department of General Surgery, Canglang Hospital of Suzhou, Suzhou, 215009, China
| | - Jing-Ru Wang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Xiao-Lu Ding
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yun Yang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Ruo-Nan Zhang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yuan-Yuan Zhao
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Hai-Xia Bi
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Ling-Chuan Guo
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China.
| | - Wen-Juan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated to Soochow University, Soochow University, Suzhou, 215124, China.
- Department of Pathology, Medical Center of Soochow University, Soochow University, Suzhou, 215124, China.
| | - Hua Wu
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China.
- Department of Pathology, Dushu Lake Hospital Affiliated to Soochow University, Soochow University, Suzhou, 215124, China.
| |
Collapse
|
5
|
Wang L, Zhu H, Sun W, Liang L, Li H, Han C, Huang W, Zhao B, Peng P, Qin M, Shi L, Mo Y, Huang J. Low expression of bestrophin-2 is associated with poor prognosis in colon cancer. Gene 2021; 813:146117. [PMID: 34902511 DOI: 10.1016/j.gene.2021.146117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/10/2021] [Accepted: 12/06/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES The purpose of this research was to confirm the prognostic value of bestrophin-2 (BEST2), one of the hub genes in colon cancer, via bioinformatics analysis and validation in public databases and immunohistochemistry detection. METHODS The GEO2R online tool and Venn diagram software were utilized to identify differentially expressed genes (DEGs) from expression profiles, including GSE20916, GSE44861 and GSE74602, from the Gene Expression Omnibus (GEO). The overall survival (OS) and disease-free survival (DFS) of colon cancer patients from The Cancer Genome Atlas (TCGA) were analyzed through Kaplan-Meier survival curves. Verification of the significance of BEST2 in colon cancer was based on TCGA, Genotype Tissue Expression (GTEx) and 10 datasets from GEO. BEST2 expression was detected with immunohistochemistry (IHC) in 330 colon tissue samples on microarrays including 165 colon cancerand 165 adjacent normal tissues. For further validation, comprehensive analysis from tissue microarrays and multiple datasets was performed by the summarizing of receiver operating characteristic (SROC) curves and the standard mean differences (SMDs). BEST2 expression in various kinds of colon cancer tissues and cell lines in the context of pancancer was obtained from the Expression Atlas database. The CBioPortal database was queried to identify BEST2 gene alterations and mutation status in colon cancer. Correlated genes (CEGs) with BEST2 and DEGs from public database data were assembled for functional and pathway enrichment analysis. RESULTS We identified 85 DEGs from the three datasets and screened out BEST2 as a prognostic predictor via the TCGA database. Colon cancer patients with high expression of BEST2 had better survival than patients with low BEST2 (HR = 0.5, P = 0.006) as shown in Kaplan-Meier survival curves in GEPIA. In all, 1463 colon cancer tissues and 1023 colon normal tissues were gathered via public databases as well as in-house tissue microarrays. The comprehensiveexpression analysis suggested low-expression of BEST2 in colon cancer (SMD = -2.48, 95% CI [-3.15- -1.80]) and the notable efficacy of BEST2 expression in differentiating colon cancer from noncancer samples (AUC = 0.97). Gene alteration status of BEST2 occurred in 5% of colon cancer cases, mostly missense mutations and deep deletions. Genes positively correlated with BEST2 and DEGs primarily aggregated in pathways such as anion absorption, digestive juice secretion, cAMP signaling and so on (P < 0.05). CONCLUSION Ampleevidencesupportsthe role of BEST2 in distinguishing colon cancer from normal tissues in this research. Low expression of BEST2 is correlated with a shorter OS, which implies that BEST2 can be employed as a potential biomarker and therapeutictarget in colon cancer.
Collapse
Affiliation(s)
- Li Wang
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Huawei Zhu
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Weiliang Sun
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Li Liang
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Hui Li
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Chenglong Han
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Wenfeng Huang
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Bi Zhao
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Peng Peng
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Mengbin Qin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Ling Shi
- Department of Pathology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Yueqing Mo
- Department of Pathology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Jiean Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Cao X, Soleimani M, Hughes BA. SLC26A7 constitutes the thiocyanate-selective anion conductance of the basolateral membrane of the retinal pigment epithelium. Am J Physiol Cell Physiol 2020; 319:C641-C656. [PMID: 32726161 DOI: 10.1152/ajpcell.00027.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anion channels in the retinal pigment epithelium (RPE) play an essential role in the transport of Cl- between the outer retina and the choroidal blood to regulate the ionic composition and volume of the subretinal fluid that surrounds the photoreceptor outer segments. Recently, we reported that the anion conductance of the mouse RPE basolateral membrane is highly selective for the biologically active anion thiocyanate (SCN-), a property that does not correspond with any of the Cl- channels that have been found to be expressed in the RPE to date. The purpose of this study was to determine the extent to which SLC26A7, a SCN- permeable-anion exchanger/channel that was reported to be expressed in human RPE, contributes to the RPE basolateral anion conductance. We show by quantitative RT-PCR that Slc26a7 is highly expressed in mouse RPE compared with other members of the Slc26 gene family and Cl- channel genes known to be expressed in the RPE. By applying immunofluorescence microscopy to mouse retinal sections and isolated cells, we localized SLC26A7 to the RPE basolateral membrane. Finally, we performed whole cell and excised patch recordings from RPE cells acutely isolated from Slc26a7 knockout mice to show that the SCN- conductance and permeability of its basolateral membrane are dramatically smaller relative to wild-type mouse RPE cells. These findings establish SLC26A7 as the SCN--selective conductance of the RPE basolateral membrane and provide new insight into the physiology of an anion channel that may participate in anion transport and pH regulation by the RPE.
Collapse
Affiliation(s)
- Xu Cao
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan
| | | | - Bret A Hughes
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
9
|
Kajii TS, Oka A, Saito F, Mitsui J, Iida J. Whole-exome sequencing in a Japanese pedigree implicates a rare non-synonymous single-nucleotide variant in BEST3 as a candidate for mandibular prognathism. Bone 2019; 122:193-198. [PMID: 30849546 DOI: 10.1016/j.bone.2019.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/12/2019] [Accepted: 03/04/2019] [Indexed: 12/26/2022]
Abstract
Mandibular prognathism is a phenotype of facial deformity seen in populations around the world, but with higher incidence among East Asian populations. Five genome-wide nonparametric linkage analyses and a genome-wide association study to identify susceptibility loci of the phenotype have shown inconsistent results. To explore variants related to mandibular prognathism, we undertook whole-exome sequencing in a Japanese pedigree. The pedigree was ascertained as mandibular prognathism. The pedigree comprised 15 individuals from 4 generations. Four affected individuals across 2 generations and 5 unaffected individuals were chosen for whole-exome sequencing. Five non-synonymous single-nucleotide variants (SNVs) of UBASH3B, OR6M1, OR8D4, OR8B4, and BEST3 genes were detected in all 4 affected individuals, but in none of the 5 unaffected individuals. A non-synonymous SNV of the BEST3 gene, Chr12(GRCh37):g.70048878G>T, NM_032735.2:c.1816C>A, p.(L606I), was identified as rare missense variant. BEST3 is located on chromosome 12q15 and encodes bestrophin 3 from the bestrophin family of anion channels. The 4 other non-synonymous SNVs of UBASH3B, OR6M1, OR8D4, and OR8B4 were not considered plausible candidates for mandibular prognathism. Our whole-exome sequencing implicates a rare non-synonymous SNV of BEST3 as a candidate for mandibular prognathism in the Japanese pedigree.
Collapse
Affiliation(s)
- Takashi S Kajii
- Section of Orthodontics, Department of Oral Growth and Development, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan.
| | - Akira Oka
- Institute of Medical Sciences, Tokai University, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Fumio Saito
- Department of Orthodontics, Division of Oral Functional Science, Graduate School of Dental Medicine, Hokkaido University, Kita 13 Nishi 7, Kita-ku, Sapporo 060-8586, Japan
| | - Jun Mitsui
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Junichiro Iida
- Department of Orthodontics, Division of Oral Functional Science, Graduate School of Dental Medicine, Hokkaido University, Kita 13 Nishi 7, Kita-ku, Sapporo 060-8586, Japan
| |
Collapse
|
10
|
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
|
11
|
Golubinskaya V, Vontell R, Supramaniam V, Wyatt-Ashmead J, Gustafsson H, Mallard C, Nilsson H. Bestrophin-3 Expression in a Subpopulation of Astrocytes in the Neonatal Brain After Hypoxic-Ischemic Injury. Front Physiol 2019; 10:23. [PMID: 30761013 PMCID: PMC6362097 DOI: 10.3389/fphys.2019.00023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/10/2019] [Indexed: 11/23/2022] Open
Abstract
Bestrophin-3, a potential candidate for a calcium-activated chloride channel, recently was suggested to have cell-protective functions. We studied the expression and alternative splicing of bestrophin-3 in neonatal mouse brain and after hypoxic-ischemic (HI) injury and in human neonatal brain samples. HI brain injury was induced in 9-day old mice by unilateral permanent common carotid artery occlusion in combination with exposure to 10% oxygen for 50 min. Endoplasmic reticulum stress was induced by thapsigargin treatment in primary culture of mouse brain astrocytes. We also investigated expression of bestrophin-3 protein in a sample of human neonatal brain tissue. Bestrophin-3 protein expression was detected with immunohistochemical methods and western blot; mRNA expression and splicing were analyzed by RT-PCR. HI induced a brain tissue infarct and a pronounced increase in the endoplasmic reticulum-associated marker CHOP. Three days after HI a population of astrocytes co-expressed bestrophin-3 and nestin in a penumbra-like area of the injured hemisphere. However, total levels of Bestrophin-3 protein in mouse cortex were reduced after injury. Mouse astrocytes in primary culture also expressed bestrophin-3 protein, the amount of which was reduced by endoplasmic reticulum stress. Bestrophin-3 protein was detected in astrocytes in the hippocampal region of the human neonatal brain which had patchy white matter gliosis and neuronal loss in the Sommer’s sector of the Ammon’s horn (CA1). Analysis of bestrophin-3 mRNA in mouse brain with and without injury showed the presence of two truncated spliced variants, but no full-length mRNA. Total amount of bestrophin-3 mRNA increased after HI, but showed only minor injury-related change. However, the splice variants of bestrophin-3 mRNA were differentially regulated after HI depending on the presence of tissue injury. Our results show that bestrophin-3 is expressed in neonatal mouse brain after injury and in the human neonatal brain with pathology. In mouse brain bestrophin-3 protein is upregulated in a specific astrocyte population after injury and is co-expressed with nestin. Splice variants of bestrophin-3 mRNA respond differently to HI, which might indicate their different roles in tissue injury.
Collapse
Affiliation(s)
- Veronika Golubinskaya
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Regina Vontell
- Division of Imaging Sciences & Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St Thomas' Hospital, London, United Kingdom
| | - Veena Supramaniam
- Division of Imaging Sciences & Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St Thomas' Hospital, London, United Kingdom
| | - Josephine Wyatt-Ashmead
- Wigglesworth Perinatal-Padiatric Pathology Service, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Helena Gustafsson
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Holger Nilsson
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
12
|
Mislocalisation of BEST1 in iPSC-derived retinal pigment epithelial cells from a family with autosomal dominant vitreoretinochoroidopathy (ADVIRC). Sci Rep 2016; 6:33792. [PMID: 27653836 PMCID: PMC5031956 DOI: 10.1038/srep33792] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/26/2016] [Indexed: 12/31/2022] Open
Abstract
Autosomal dominant vitreoretinochoroidopathy (ADVIRC) is a rare, early-onset retinal dystrophy characterised by distinct bands of circumferential pigmentary degeneration in the peripheral retina and developmental eye defects. ADVIRC is caused by mutations in the Bestrophin1 (BEST1) gene, which encodes a transmembrane protein thought to function as an ion channel in the basolateral membrane of retinal pigment epithelial (RPE) cells. Previous studies suggest that the distinct ADVIRC phenotype results from alternative splicing of BEST1 pre-mRNA. Here, we have used induced pluripotent stem cell (iPSC) technology to investigate the effects of an ADVIRC associated BEST1 mutation (c.704T > C, p.V235A) in patient-derived iPSC-RPE. We found no evidence of alternate splicing of the BEST1 transcript in ADVIRC iPSC-RPE, however in patient-derived iPSC-RPE, BEST1 was expressed at the basolateral membrane and the apical membrane. During human eye development we show that BEST1 is expressed more abundantly in peripheral RPE compared to central RPE and is also expressed in cells of the developing retina. These results suggest that higher levels of mislocalised BEST1 expression in the periphery, from an early developmental stage, could provide a mechanism that leads to the distinct clinical phenotype observed in ADVIRC patients.
Collapse
|
13
|
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
|
14
|
Golubinskaya V, Elvin J, Ebefors K, Gustafsson H, Mallard C, Nyström J, Nilsson H. Bestrophin-3 is differently expressed in normal and injured mouse glomerular podocytes. Acta Physiol (Oxf) 2015; 214:481-96. [PMID: 25912364 DOI: 10.1111/apha.12516] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 03/17/2015] [Accepted: 04/20/2015] [Indexed: 12/13/2022]
Abstract
AIM Bestrophins are putative calcium-activated chloride channels. Recently, cell-protective functions for Bestrophin-3 (Best3) were proposed. Best3 exists in different splice variants. We have here examined expression, alternative splicing and localization of Best3 in mouse podocytes under normal conditions and during endoplasmic reticulum (ER) stress. METHODS Best3 expression was determined on the mRNA level using quantitative PCR and on the protein level by immunohistochemistry and Western blotting. RESULTS Staining for Best3 was pronounced in glomeruli and was detected in cultured mouse podocytes. Best3 did not co-localize with markers for endothelial cells (CD31), podocyte foot processes (synaptopodin) or microtubules (actin). However, immunogold-based electron microscopy and co-localization with nestin showed Best3 presence in podocyte primary processes and cell bodies. Only two splice variants of Best3 mRNA (both lacking exons 2 and 3, and one also lacking exon 6), but no full-length variant, were detected. ER stress induced by lipopolysaccharides in vivo transiently elevated mRNA levels of total Best3 and its two splice variants with different time courses. In cultured podocytes under ER stress induced by thapsigargin, the expression of total Best3, its splice variants and nestin transiently increased with similar time courses. The ER stress marker C/EBP homologous protein (CHOP) and nestin mRNA increased during ER stress in vivo and in vitro. CONCLUSIONS Best3 is localized intracellularly in cell bodies and primary processes of mouse podocytes and is co-localized with nestin. Two splice variants of Best3 are expressed in glomeruli and in cultured podocytes, and their expression is differentially regulated in ER stress.
Collapse
Affiliation(s)
- V. Golubinskaya
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| | - J. Elvin
- Department of Molecular and Clinical Medicine; Institute of Medicine; The Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| | - K. Ebefors
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| | - H. Gustafsson
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| | - C. Mallard
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| | - J. Nyström
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| | - H. Nilsson
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| |
Collapse
|
15
|
Leblanc N, Forrest AS, Ayon RJ, Wiwchar M, Angermann JE, Pritchard HAT, Singer CA, Valencik ML, Britton F, Greenwood IA. Molecular and functional significance of Ca(2+)-activated Cl(-) channels in pulmonary arterial smooth muscle. Pulm Circ 2015; 5:244-68. [PMID: 26064450 DOI: 10.1086/680189] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/22/2014] [Indexed: 12/31/2022] Open
Abstract
Increased peripheral resistance of small distal pulmonary arteries is a hallmark signature of pulmonary hypertension (PH) and is believed to be the consequence of enhanced vasoconstriction to agonists, thickening of the arterial wall due to remodeling, and increased thrombosis. The elevation in arterial tone in PH is attributable, at least in part, to smooth muscle cells of PH patients being more depolarized and displaying higher intracellular Ca(2+) levels than cells from normal subjects. It is now clear that downregulation of voltage-dependent K(+) channels (e.g., Kv1.5) and increased expression and activity of voltage-dependent (Cav1.2) and voltage-independent (e.g., canonical and vanilloid transient receptor potential [TRPC and TRPV]) Ca(2+) channels play an important role in the functional remodeling of pulmonary arteries in PH. This review focuses on an anion-permeable channel that is now considered a novel excitatory mechanism in the systemic and pulmonary circulations. It is permeable to Cl(-) and is activated by a rise in intracellular Ca(2+) concentration (Ca(2+)-activated Cl(-) channel, or CaCC). The first section outlines the biophysical and pharmacological properties of the channel and ends with a description of the molecular candidate genes postulated to encode for CaCCs, with particular emphasis on the bestrophin and the newly discovered TMEM16 and anoctamin families of genes. The second section provides a review of the various sources of Ca(2+) activating CaCCs, which include stimulation by mobilization from intracellular Ca(2+) stores and Ca(2+) entry through voltage-dependent and voltage-independent Ca(2+) channels. The third and final section summarizes recent findings that suggest a potentially important role for CaCCs and the gene TMEM16A in PH.
Collapse
Affiliation(s)
- Normand Leblanc
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Abigail S Forrest
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Ramon J Ayon
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Michael Wiwchar
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Jeff E Angermann
- School of Community Health Sciences, University of Nevada, Reno, Nevada, USA
| | - Harry A T Pritchard
- Vascular Biology Research Centre, Institute of Cardiovascular and Cell Sciences, St. George's University of London, London, United Kingdom
| | - Cherie A Singer
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Maria L Valencik
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Fiona Britton
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Iain A Greenwood
- Vascular Biology Research Centre, Institute of Cardiovascular and Cell Sciences, St. George's University of London, London, United Kingdom
| |
Collapse
|
16
|
Bestrophin 1 is indispensable for volume regulation in human retinal pigment epithelium cells. Proc Natl Acad Sci U S A 2015; 112:E2630-9. [PMID: 25941382 DOI: 10.1073/pnas.1418840112] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In response to cell swelling, volume-regulated anion channels (VRACs) participate in a process known as regulatory volume decrease (RVD). Only recently, first insight into the molecular identity of mammalian VRACs was obtained by the discovery of the leucine-rich repeats containing 8A (LRRC8A) gene. Here, we show that bestrophin 1 (BEST1) but not LRRC8A is crucial for volume regulation in human retinal pigment epithelium (RPE) cells. Whole-cell patch-clamp recordings in RPE derived from human-induced pluripotent stem cells (hiPSC) exhibit an outwardly rectifying chloride current with characteristic functional properties of VRACs. This current is severely reduced in hiPSC-RPE cells derived from macular dystrophy patients with pathologic BEST1 mutations. Disruption of the orthologous mouse gene (Best1(-/-)) does not result in obvious retinal pathology but leads to a severe subfertility phenotype in agreement with minor endogenous expression of Best1 in murine RPE but highly abundant expression in mouse testis. Sperm from Best1(-/-) mice showed reduced motility and abnormal sperm morphology, indicating an inability in RVD. Together, our data suggest that the molecular identity of VRACs is more complex--that is, instead of a single ubiquitous channel, VRACs could be formed by cell type- or tissue-specific subunit composition. Our findings provide the basis to further examine VRAC diversity in normal and diseased cell physiology, which is key to exploring novel therapeutic approaches in VRAC-associated pathologies.
Collapse
|
17
|
In-depth characterisation of Retinal Pigment Epithelium (RPE) cells derived from human induced pluripotent stem cells (hiPSC). Neuromolecular Med 2014; 16:551-64. [PMID: 24801942 PMCID: PMC4119585 DOI: 10.1007/s12017-014-8308-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/22/2014] [Indexed: 12/24/2022]
Abstract
Induced pluripotent stem cell (iPSC)-derived retinal pigment epithelium (RPE) has widely been appreciated as a promising tool to model human ocular disease emanating from primary RPE pathology. Here, we describe the successful reprogramming of adult human dermal fibroblasts to iPSCs and their differentiation to pure expandable RPE cells with structural and functional features characteristic for native RPE. Fibroblast cultures were established from skin biopsy material and subsequently reprogrammed following polycistronic lentiviral transduction with OCT4, SOX2, KLF4 and L-Myc. Fibroblast-derived iPSCs showed typical morphology, chromosomal integrity and a distinctive stem cell marker profile. Subsequent differentiation resulted in expandable pigmented hexagonal RPE cells. The cells revealed stable RNA expression of mature RPE markers RPE65, RLBP and BEST1. Immunolabelling verified localisation of BEST1 at the basolateral plasma membrane, and scanning electron microscopy showed typical microvilli at the apical side of iPSC-derived RPE cells. Transepithelial resistance was maintained at high levels during cell culture indicating functional formation of tight junctions. Secretion capacity was demonstrated for VEGF-A. Feeding of porcine photoreceptor outer segments revealed the proper ability of these cells for phagocytosis. IPSC-derived RPE cells largely maintained these properties after cryopreservation. Together, our study underlines that adult dermal fibroblasts can serve as a valuable resource for iPSC-derived RPE with characteristics highly reminiscent of true RPE cells. This will allow its broad application to establish cellular models for RPE-related human diseases.
Collapse
|
18
|
Effects of alternative splicing on the function of bestrophin-1 calcium-activated chloride channels. Biochem J 2014; 458:575-83. [PMID: 24341532 DOI: 10.1042/bj20121546] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The proposed Ca2+-activated Cl- channel protein Best1 (bestrophin 1) is expressed and functionally important in the retina and in the brain. Human BEST1 has two known splice variants, Best1V1 and Best1V2, which arise from alternative splicing of two exons: exon 2 splicing results in a unique N-terminal domain, whereas alternative splicing of exon 11 produces two mutually exclusive C-termini. Prior studies were limited to Best1V1 and its clinically relevant mutations. In the present work, we cloned a novel splice variant of Best1V1 missing exon 2 (Best1V1Δex2) and differing from each of the two previously identified isoforms by one alternatively spliced domain. This finding allowed us to determine the role for alternative splicing of the Best1 N- and C-termini. We heteroexpressed Best1V1Δex2 in HEK (human embryonic kidney)-293 cells, and compared its properties with Best1V1 and Best1V2. Western blot analysis confirmed protein expression from all three splice variants. Both Best1V1 and Best1V1Δex2 successfully formed Ca2+-activated Cl- channels, demonstrating that the N-terminus encoded by exon 2 is not essential for channel function. In contrast, Best1V2-expressing cells had no detectable Ca2+-activated Cl- currents, pointing to a critical role for splicing of the C-terminus. Surface protein biotinylation demonstrated that Best1V1 and Best1V1Δex2 are trafficked to the plasma membrane, whereas Best1V2 is not. These results define the impact of alternative splicing on Best1 function, and should be taken into consideration in future modelling of the Best1 protein structure.
Collapse
|
19
|
Abstract
Human Bestrophin 1 (hBest1) is a calcium-activated chloride channel that regulates neuronal excitability, synaptic activity, and retinal homeostasis. Mutations in hBest1 cause the autosomal-dominant Best macular dystrophy (BMD). Because hBest1 mutations cause BMD, but a knockout does not, we wondered if hBest1 mutants exert a dominant negative effect through interaction with other calcium-activated chloride channels, such as hBest2, 3, or 4, or transmembrane member 16A (TMEM16A), a member of another channel family. The subunit architecture of Best channels is debated, and their ability to form heteromeric channel assemblies is unclear. Using single-molecule subunit analysis, we find that each of hBest1, 2, 3, and 4 forms a homotetrameric channel. Despite considerable conservation among hBests, hBest1 has little or no interaction with other hBests or mTMEM16A. We identify the domain responsible for assembly specificity. This domain also plays a role in channel function. Our results indicate that Best channels preferentially self-assemble into homotetramers.
Collapse
|
20
|
Pasquay C, Wang LF, Lorenz B, Preising MN. Bestrophin 1 – Phenotypes and Functional Aspects in Bestrophinopathies. Ophthalmic Genet 2013; 36:193-212. [DOI: 10.3109/13816810.2013.863945] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
21
|
Ito G, Okamoto R, Murano T, Shimizu H, Fujii S, Nakata T, Mizutani T, Yui S, Akiyama-Morio J, Nemoto Y, Okada E, Araki A, Ohtsuka K, Tsuchiya K, Nakamura T, Watanabe M. Lineage-specific expression of bestrophin-2 and bestrophin-4 in human intestinal epithelial cells. PLoS One 2013; 8:e79693. [PMID: 24223998 PMCID: PMC3818177 DOI: 10.1371/journal.pone.0079693] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 09/24/2013] [Indexed: 01/21/2023] Open
Abstract
Intestinal epithelial cells (IECs) regulate the absorption and secretion of anions, such as HCO3- or Cl-. Bestrophin genes represent a newly identified group of calcium-activated Cl- channels (CaCCs). Studies have suggested that, among the four human bestrophin-family genes, bestrophin-2 (BEST2) and bestrophin-4 (BEST4) might be expressed within the intestinal tissue. Consistently, a study showed that BEST2 is expressed by human colonic goblet cells. However, their precise expression pattern along the gastrointestinal tract, or the lineage specificity of the cells expressing these genes, remains largely unknown. Here, we show that BEST2 and BEST4 are expressed in vivo, each in a distinct, lineage-specific manner, in human IECs. While BEST2 was expressed exclusively in colonic goblet cells, BEST4 was expressed in the absorptive cells of both the small intestine and the colon. In addition, we found that BEST2 expression is significantly down-regulated in the active lesions of ulcerative colitis, where goblet cells were depleted, suggesting that BEST2 expression is restricted to goblet cells under both normal and pathologic conditions. Consistently, the induction of goblet cell differentiation by a Notch inhibitor, LY411575, significantly up-regulated the expression of not BEST4 but BEST2 in MUC2-positive HT-29 cells. Conversely, the induction of absorptive cell differentiation up-regulated the expression of BEST4 in villin-positive Caco-2 cells. In addition, we found that the up- or down-regulation of Notch activity leads to the preferential expression of either BEST4 or BEST2, respectively, in LS174T cells. These results collectively confirmed that BEST2 and BEST4 could be added to the lineage-specific genes of humans IECs due to their abilities to clearly identify goblet cells of colonic origin and a distinct subset of absorptive cells, respectively.
Collapse
Affiliation(s)
- Go Ito
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Advanced GI therapeutics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail:
| | - Tatsuro Murano
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiromichi Shimizu
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoru Fujii
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toru Nakata
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomohiro Mizutani
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shiro Yui
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Junko Akiyama-Morio
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuhiro Nemoto
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Eriko Okada
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akihiro Araki
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuo Ohtsuka
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kiichiro Tsuchiya
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Advanced GI therapeutics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tetsuya Nakamura
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Advanced GI therapeutics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
22
|
|
23
|
Identification of colorectal cancer related genes with mRMR and shortest path in protein-protein interaction network. PLoS One 2012; 7:e33393. [PMID: 22496748 PMCID: PMC3319543 DOI: 10.1371/journal.pone.0033393] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 02/13/2012] [Indexed: 11/19/2022] Open
Abstract
One of the most important and challenging problems in biomedicine and genomics is how to identify the disease genes. In this study, we developed a computational method to identify colorectal cancer-related genes based on (i) the gene expression profiles, and (ii) the shortest path analysis of functional protein association networks. The former has been used to select differentially expressed genes as disease genes for quite a long time, while the latter has been widely used to study the mechanism of diseases. With the existing protein-protein interaction data from STRING (Search Tool for the Retrieval of Interacting Genes), a weighted functional protein association network was constructed. By means of the mRMR (Maximum Relevance Minimum Redundancy) approach, six genes were identified that can distinguish the colorectal tumors and normal adjacent colonic tissues from their gene expression profiles. Meanwhile, according to the shortest path approach, we further found an additional 35 genes, of which some have been reported to be relevant to colorectal cancer and some are very likely to be relevant to it. Interestingly, the genes we identified from both the gene expression profiles and the functional protein association network have more cancer genes than the genes identified from the gene expression profiles alone. Besides, these genes also had greater functional similarity with the reported colorectal cancer genes than the genes identified from the gene expression profiles alone. All these indicate that our method as presented in this paper is quite promising. The method may become a useful tool, or at least plays a complementary role to the existing method, for identifying colorectal cancer genes. It has not escaped our notice that the method can be applied to identify the genes of other diseases as well.
Collapse
|
24
|
Milenkovic VM, Röhrl E, Weber BHF, Strauss O. Disease-associated missense mutations in bestrophin-1 affect cellular trafficking and anion conductance. J Cell Sci 2012; 124:2988-96. [PMID: 21878505 DOI: 10.1242/jcs.085878] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Bestrophin-1, an integral membrane protein encoded by the BEST1 gene, is localized predominantly to the basolateral membrane of the retinal pigment epithelium. Mutations in the BEST1 gene have been associated with Best vitelliforme macular dystrophy (BMD), a central retinopathy with autosomal dominant inheritance and variable penetrance. Over 120 disease-causing mutations are known, the majority of which result in amino acid substitutions within four mutational hot-spot regions in the highly conserved N-terminal half of the protein. Although initially thought to impair Cl⁻ channel function, the molecular pathology of BEST1 mutations is still controversial. We have analyzed the subcellular localization of 13 disease-associated BEST1 mutant proteins in polarized MDCK II cells, an established model of apical to basolateral protein sorting. Immunostaining demonstrated that nine of the 13 mutant proteins failed to integrate into the cell membrane. The defective proteins were predominantly retained in the cytoplasm, whereas wild-type bestrophin-1 revealed cell membrane localization. Functional analysis of I⁻ fluxes in HEK-293 cells showed that all mutants exhibited a significant reduction in anion conductance. Our data indicate that defective intracellular trafficking could be a common cause of BMD accompanied by impaired anion conductance, representing a loss of anion channel function that is probably due to mistargeting of mutant protein.
Collapse
Affiliation(s)
- Vladimir M Milenkovic
- Experimental Ophthalmology, Eye Hospital, University Medical Center Regensburg, 93054 Regensburg, Germany
| | | | | | | |
Collapse
|
25
|
Wittström E, Ponjavic V, Bondeson ML, Andréasson S. Anterior Segment Abnormalities and Angle-Closure Glaucoma in a Family with a Mutation in theBEST1Gene and Best Vitelliform Macular Dystrophy. Ophthalmic Genet 2011; 32:217-27. [DOI: 10.3109/13816810.2011.567884] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Abstract
PURPOSE The purpose of this study was to investigate the BEST1 gene mutations in Chinese patients with Best vitelliform macular dystrophy (BVMD). METHODS Twenty-six subjects from 7 Chinese families with BVMD and 100 unrelated healthy Chinese subjects without a family history of BVMD were screened for mutations in the BEST1 gene by direct sequencing. The subjects underwent complete ophthalmologic examination and BEST1 gene screening. RESULTS Six novel missense mutations (Thr2Asn, Leu75Phe, Ser144Asn, Arg255Trp, Pro297Thr, and Asp301Gly) and 1 previously reported mutation (Arg218Cys) were identified. Each family was found to have a unique BEST1 mutation that segregated with the disease. Two of the six novel mutations are located within the four previously reported common mutation clusters within the BEST1 gene. One family with patients having homozygous Leu75Phe mutations did not have the more severe BVMD phenotype. None of the patients with mutations was identified among the 100 healthy control subjects. CONCLUSION A large number of unique novel missense mutations was found in Chinese patients with BVMD, suggesting considerable interethnic differences between the mutation sites in the BEST1 gene in different populations. The few truncating BEST1 mutations and the lack of a more severe phenotype in homozygous patients suggest that the missense BEST1 mutation may produce a dominant negative effect on wild-type BEST1 gene.
Collapse
|
27
|
Romanenko VG, Catalán MA, Brown DA, Putzier I, Hartzell HC, Marmorstein AD, Gonzalez-Begne M, Rock JR, Harfe BD, Melvin JE. Tmem16A encodes the Ca2+-activated Cl- channel in mouse submandibular salivary gland acinar cells. J Biol Chem 2010; 285:12990-3001. [PMID: 20177062 PMCID: PMC2857126 DOI: 10.1074/jbc.m109.068544] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 01/30/2010] [Indexed: 12/28/2022] Open
Abstract
Activation of an apical Ca(2+)-dependent Cl(-) channel (CaCC) is the rate-limiting step for fluid secretion in many exocrine tissues. Here, we compared the properties of native CaCC in mouse submandibular salivary gland acinar cells to the Ca(2+)-gated Cl(-) currents generated by Tmem16A and Best2, members from two distinct families of Ca(2+)-activated Cl(-) channels found in salivary glands. Heterologous expression of Tmem16A and Best2 transcripts in HEK293 cells produced Ca(2+)-activated Cl(-) currents with time and voltage dependence and inhibitor sensitivity that resembled the Ca(2+)-activated Cl(-) current found in native salivary acinar cells. Best2(-/-) and Tmem16A(-/-) mice were used to further characterize the role of these channels in the exocrine salivary gland. The amplitude and the biophysical footprint of the Ca(2+)-activated Cl(-) current in submandibular gland acinar cells from Best2-deficient mice were the same as in wild type cells. Consistent with this observation, the fluid secretion rate in Best2 null mice was comparable with that in wild type mice. In contrast, submandibular gland acinar cells from Tmem16A(-/-) mice lacked a Ca(2+)-activated Cl(-) current and a Ca(2+)-mobilizing agonist failed to stimulate Cl(-) efflux, requirements for fluid secretion. Furthermore, saliva secretion was abolished by the CaCC inhibitor niflumic acid in wild type and Best2(-/-) mice. Our results demonstrate that both Tmem16A and Best2 generate Ca(2+)-activated Cl(-) current in vitro with similar properties to those expressed in native cells, yet only Tmem16A appears to be a critical component of the acinar Ca(2+)-activated Cl(-) channel complex that is essential for saliva production by the submandibular gland.
Collapse
Affiliation(s)
| | | | | | - Ilva Putzier
- the
Department of Cell Biology, Emory University, Atlanta, Georgia 30322
| | - H. Criss Hartzell
- the
Department of Cell Biology, Emory University, Atlanta, Georgia 30322
| | - Alan D. Marmorstein
- the
Department of Ophthalmology and Vision Science and
- College of Optical Sciences, University of Arizona, Tucson, Arizona 85711, and the
| | | | - Jason R. Rock
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida 32611
| | - Brian D. Harfe
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida 32611
| | | |
Collapse
|
28
|
Dace DS, Khan AA, Stark JL, Kelly J, Cross AH, Apte RS. Interleukin-10 overexpression promotes Fas-ligand-dependent chronic macrophage-mediated demyelinating polyneuropathy. PLoS One 2009; 4:e7121. [PMID: 19771172 PMCID: PMC2743195 DOI: 10.1371/journal.pone.0007121] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 08/19/2009] [Indexed: 12/20/2022] Open
Abstract
Background Demyelinating polyneuropathy is a debilitating, poorly understood disease that can exist in acute (Guillain-Barré syndrome) or chronic forms. Interleukin-10 (IL-10), although traditionally considered an anti-inflammatory cytokine, has also been implicated in promoting abnormal angiogenesis in the eye and in the pathobiology of autoimmune diseases such as lupus and encephalomyelitis. Principal Findings Overexpression of IL-10 in a transgenic mouse model leads to macrophage-mediated demyelinating polyneuropathy. IL-10 upregulates ICAM-1 within neural tissues, promoting massive macrophage influx, inflammation-induced demyelination, and subsequent loss of neural tissue resulting in muscle weakness and paralysis. The primary insult is to perineural myelin followed by secondary axonal loss. Infiltrating macrophages within the peripheral nerves demonstrate a highly pro-inflammatory signature. Macrophages are central players in the pathophysiology, as in vivo depletion of macrophages using clodronate liposomes reverses the phenotype, including progressive nerve loss and paralysis. Macrophage-mediate demyelination is dependent on Fas-ligand (FasL)-mediated Schwann cell death. Significance These findings mimic the human disease chronic idiopathic demyelinating polyneuropathy (CIDP) and may also promote further understanding of the pathobiology of related conditions such as acute idiopathic demyelinating polyneuropathy (AIDP) or Guillain-Barré syndrome.
Collapse
Affiliation(s)
- Dru S. Dace
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
| | - Aslam A. Khan
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
| | - Jennifer L. Stark
- The National Multiple Sclerosis Society, New York, New York, United States of America
| | - Jennifer Kelly
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
| | - Anne H. Cross
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
| | - Rajendra S. Apte
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
29
|
Onuma Y, Haramoto Y, Nejigane S, Takahashi S, Asashima M. Bestrophin genes are expressed in Xenopus development. Biochem Biophys Res Commun 2009; 384:290-5. [DOI: 10.1016/j.bbrc.2009.04.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 04/22/2009] [Indexed: 10/20/2022]
|
30
|
Sohn EH, Francis PJ, Duncan JL, Weleber RG, Saperstein DA, Farrell DF, Stone EM. Phenotypic variability due to a novel Glu292Lys variation in exon 8 of the BEST1 gene causing best macular dystrophy. ARCHIVES OF OPHTHALMOLOGY (CHICAGO, ILL. : 1960) 2009; 127:913-20. [PMID: 19597114 PMCID: PMC2711525 DOI: 10.1001/archophthalmol.2009.148] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
OBJECTIVE To study the phenotypic characteristics of patients with a novel p.E292K mutation in BEST1. METHODS Affected individuals underwent ophthalmic examination and testing that included photography, autofluorescence, optical coherence tomography, and electrophysiological testing. Their DNA was analyzed for BEST1 mutations. RESULTS Five patients aged 5 to 59 years who expressed the p.E292K mutation in BEST1 were identified in 3 families. Electro-oculographic light-rise was subnormal in all probands and carriers. Carriers had normal findings from fundus examination, multifocal electroretinography, and visual acuity, and were emmetropic or myopic. Only probands had hyperopia and fundus findings typical of Best macular dystrophy. Optical coherence tomography of vitelliform lesions demonstrated retinal pigment epithelium elevation without subretinal fluid; atrophic lesions exhibited disruption of the hyperreflective outer retina-retinal pigment epithelium complex. Intense hyperautofluorescence correlated with the vitelliform lesion. CONCLUSIONS Patients with the Glu292Lys variation in BEST1 exhibit intrafamilial and interfamilial phenotypic variability. A disproportionate fraction (26%) of Best disease-causing mutations occurs in exon 8, suggesting that the portion of protein encoded by this exon (amino acids 290-316) may be especially important to bestrophin's function. Relatively good visual acuity with vitelliform lesions can be explained by preservation of the outer retina, demonstrated by optical coherence tomography. Clinical Relevance A novel mutation in this region of BEST1 carries implications for disease pathogenesis.
Collapse
Affiliation(s)
- Elliott H Sohn
- Doheny Eye Institute and Department of Ophthalmology, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Boon CJ, Klevering BJ, Leroy BP, Hoyng CB, Keunen JE, den Hollander AI. The spectrum of ocular phenotypes caused by mutations in the BEST1 gene. Prog Retin Eye Res 2009; 28:187-205. [DOI: 10.1016/j.preteyeres.2009.04.002] [Citation(s) in RCA: 242] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
32
|
Abstract
There are four members of the bestrophin family of proteins in the human genome, of which two are known to be expressed in the eye. The gene BEST1 (formerly VMD2) which encodes the protein bestrophin-1 (Best1) was first identified in 1998. Mutations in this gene have now been associated with four clinically distinguishable human eye diseases, collectively referred to as "bestrophinopathies". Over the last decade, laboratories have sought to understand how Best1 mutations could result in eye diseases that range in presentation from macular degeneration to nanophthalmos. The majority of our knowledge comes from studies that have sought to understand how Best1 mutations or dysfunction could induce the classical symptoms of the most common of these diseases: Best vitelliform macular dystrophy (BVMD). BVMD is a dominant trait that is characterized electrophysiologically by a diminished electrooculogram light peak with a normal clinical electroretinogram. This together with the localization of Best1 to the retinal pigment epithelium (RPE) basolateral plasma membrane and data from heterologous expression studies, have led to the proposal that Best1 generates the light peak, and that bestrophins are a family of Ca(2+) activated Cl(-) channels (CaCCs). However, data from Best1 knock-out and knock-in mice, coupled with the recent discovery of a recessive bestrophinopathy suggest that Best1 does not generate the light peak. Recently Best2 was found to be expressed in non-pigmented epithelia in the ciliary body. However, aqueous dynamics in Best2 knock-out mice do not support a role for Best2 as a Cl(-) channel. Thus, the purported CaCC function of the bestrophins and how loss of this function relates to clinical disease needs to be reassessed. In this article, we examine data obtained from tissue-type and animal models and discuss the current state of bestrophin research, what roles Best1 and Best2 may play in ocular epithelia and ocular electrophysiology, and how perturbation of these functions may result in disease.
Collapse
Affiliation(s)
- Alan D Marmorstein
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ 85711, USA.
| | | | | |
Collapse
|
33
|
Kranjc A, Grillo FW, Rievaj J, Boccaccio A, Pietrucci F, Menini A, Carloni P, Anselmi C. Regulation of bestrophins by Ca2+: a theoretical and experimental study. PLoS One 2009; 4:e4672. [PMID: 19262692 PMCID: PMC2650406 DOI: 10.1371/journal.pone.0004672] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 01/26/2009] [Indexed: 01/06/2023] Open
Abstract
Bestrophins are a recently discovered family of Cl(-) channels, for which no structural information is available. Some family members are activated by increased intracellular Ca2+ concentration. Bestrophins feature a well conserved Asp-rich tract in their COOH terminus (Asp-rich domain), which is homologous to Ca2+-binding motifs in human thrombospondins and in human big-conductance Ca2+- and voltage-gated K+ channels (BK(Ca)). Consequently, the Asp-rich domain is also a candidate for Ca2+ binding in bestrophins. Based on these considerations, we constructed homology models of human bestrophin-1 (Best1) Asp-rich domain using human thrombospondin-1 X-ray structure as a template. Molecular dynamics simulations were used to identify Asp and Glu residues binding Ca2+ and to predict the effects of their mutations to alanine. We then proceeded to test selected mutations in the Asp-rich domain of the highly homologous mouse bestrophin-2. The mutants expressed in HEK-293 cells were investigated by electrophysiological experiments using the whole-cell voltage-clamp technique. Based on our molecular modeling results, we predicted that Asp-rich domain has two defined binding sites and that D301A and D304A mutations may impact the binding of the metal ions. The experiments confirmed that these mutations do actually affect the function of the protein causing a large decrease in the Ca2+-activated Cl(-) current, fully consistent with our predictions. In addition, other studied mutations (E306A, D312A) did not decrease Ca2+-activated Cl(-) current in agreement with modeling results.
Collapse
Affiliation(s)
- Agata Kranjc
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| | | | - Juraj Rievaj
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| | - Anna Boccaccio
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
- Italian Institute of Technology (IIT), SISSA-Unit, Trieste, Italy
| | - Fabio Pietrucci
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| | - Anna Menini
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
- Italian Institute of Technology (IIT), SISSA-Unit, Trieste, Italy
| | - Paolo Carloni
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
- Italian Institute of Technology (IIT), SISSA-Unit, Trieste, Italy
- CNR-INFM-DEMOCRITOS Modeling Center for Research in Atomistic Simulation, Trieste, Italy
| | - Claudio Anselmi
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
- CNR-INFM-DEMOCRITOS Modeling Center for Research in Atomistic Simulation, Trieste, Italy
- * E-mail:
| |
Collapse
|
34
|
Marsey LL, Winpenny JP. Bestrophin expression and function in the human pancreatic duct cell line, CFPAC-1. J Physiol 2009; 587:2211-24. [PMID: 19237432 DOI: 10.1113/jphysiol.2008.159087] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pancreatic duct epithelial cells (PDECs) have been shown to express calcium activated chloride channels (CaCCs) and there is evidence for their involvement in fluid secretion from these cells. The molecular identity of the CaCC in PDECs remains unknown. Recently, the bestrophin family of proteins have been proposed as a potential molecular candidate for CaCCs. Expression of bestrophins is strongly correlated with the function of CaCCs in a variety of tissues. In the present study, the expression of bestrophins has been investigated in the cystic fibrosis pancreatic duct cell line, CFPAC-1. Iodide efflux analysis was used to characterise native CaCCs in CFPAC-1 cell monolayers. Efflux was induced with the addition of UTP (100 microM, 10.2 +/- 1.5 nmol min(-1)), which was blocked by the chloride channel blockers niflumic acid (81%) and DIDS (90%). The UTP-stimulated iodide efflux was shown to be Ca(2+) dependent and cAMP independent. RT-PCR analysis of RNA isolated from CFPAC-1 cells demonstrated positive identification of all four human bestrophin mRNAs. Western blot of CFPAC-1 cell protein isolates with antibodies specific to human bestrophin 1 (hBest1) showed that hBest1 protein was expressed in this cell line. HBest1 was present on the cell surface, demonstrated using biotinylation and confocal imaging, as well as in the cytoplasm. SiRNA-mediated silencing of hBest1 in CFPAC-1 cells reduced the UTP-stimulated iodide efflux by around 40%. This study provides evidence that the bestrophins are expressed in pancreatic duct cells and, more specifically, that hBest1 plays a role in the CaCCs found in these cells.
Collapse
Affiliation(s)
- Laura L Marsey
- Biomedicine Group, Biomedical Research Centre, School of Medicine, Health Policy and Practice, Faculty of Health, University of East Anglia, Norwich NR4 7TJ, UK
| | | |
Collapse
|
35
|
O'Driscoll KE, Hatton WJ, Burkin HR, Leblanc N, Britton FC. Expression, localization, and functional properties of Bestrophin 3 channel isolated from mouse heart. Am J Physiol Cell Physiol 2008; 295:C1610-24. [PMID: 18945938 DOI: 10.1152/ajpcell.00461.2008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bestrophins are a novel family of proteins that encode calcium-activated chloride channels. In this study we establish that Bestrophin transcripts are expressed in the mouse and human heart. Native mBest3 protein expression and localization in heart was demonstrated by using a specific polyclonal mBest3 antibody. Immunostaining of isolated cardiac myocytes indicates that mBest3 is present at the membrane. Using the patch-clamp technique, we characterized the biophysical and pharmacological properties of mBest3 cloned from heart. Whole cell chloride currents were evoked in both HEK293 and COS-7 cells expressing mBest3 by elevation of intracellular calcium. mBest3 currents displayed a K(D) for Ca(2+) of approximately 175 nM. The calcium-activated chloride current was found to be time and voltage independent and displayed slight outward rectification. The anion permeability sequence of the channel was SCN(-)>I(-)>Cl(-), and the current was inhibited by niflumic acid and DIDS in the micromolar range. In addition, we generated a site-specific mutation (F80L) in the putative pore region of mBest3 that significantly altered the ion conduction and pharmacology of this channel. Our functional and mutational studies examining the biophysical properties of mBest3 indicate that it functions as a pore-forming chloride channel that is activated by physiological levels of calcium. This study reports novel findings regarding the molecular expression, tissue localization, and functional properties of mBest3 cloned from heart.
Collapse
Affiliation(s)
- Kate E O'Driscoll
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | | | | | | | | |
Collapse
|
36
|
Srivastava A, Romanenko VG, Gonzalez-Begne M, Catalán MA, Melvin JE. A variant of the Ca2+-activated Cl channel Best3 is expressed in mouse exocrine glands. J Membr Biol 2008; 222:43-54. [PMID: 18414923 DOI: 10.1007/s00232-008-9098-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2007] [Accepted: 02/21/2008] [Indexed: 11/27/2022]
Abstract
Fluid secretion by exocrine glands requires the activation of an apical Ca2+-dependent Cl channel, the molecular identity of which is unknown. We found that mouse exocrine glands expressed an alternately spliced variant of Best3, a member of the Bestrophin (Vmd2) Ca2+-activated Cl channel gene family, whereas the heart expressed full-length Best3. The spliced transcript lacked exons 2, 3 and 6 (Best3-Delta2,3,6) and is predicted to generate an in-frame protein missing the entire cytoplasmic N terminus, the initial two transmembrane domains and part of the first intracellular loop. In addition to exocrine glands, the Best3-Delta2,3,6 splice variant transcript was detected in lung, testis and kidney. The parotid gland and heart expressed proteins of the predicted size for Best3-Delta2,3,6 and full-length Best3, respectively, that targeted to the plasma membrane in HEK293 cells. HEK293 cells expressing Best3 displayed Ca2+-dependent Cl(-) currents that were sensitive to the Cl channel blocker DIDS. In contrast, no Ca2+-dependent Cl(-) currents were detected in cells expressing Best3-Delta2,3,6. Cotransfection of Best3-Delta2,3,6 with Best3 or Best2 (also expressed in salivary gland acinar cells) had no significant effects on the currents generated by either of these Ca2+-dependent Cl channels. Our results demonstrate that exocrine glands express a unique splice variant of Best3. Nevertheless, Best3-Delta2,3,6 does not produce Ca2+-dependent Cl(-) currents, nor does it regulate the activity of Best2 or the full-length Best3 channel.
Collapse
Affiliation(s)
- Alaka Srivastava
- Center for Oral Biology, Medical Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 611, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
37
|
Bakall B, McLaughlin P, Stanton JB, Zhang Y, Hartzell HC, Marmorstein LY, Marmorstein AD. Bestrophin-2 is involved in the generation of intraocular pressure. Invest Ophthalmol Vis Sci 2008; 49:1563-70. [PMID: 18385076 PMCID: PMC2832837 DOI: 10.1167/iovs.07-1338] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The bestrophin family of proteins has been demonstrated to generate or regulate Ca2+-activated Cl(-) conductances. Mutations in bestrophin-1 (Best1) cause several blinding eye diseases, but little is known about other bestrophin family members. This study involved disruption of the Best2 gene in mice. METHODS The mouse Best2 gene was disrupted by replacing exons 1, 2, and part of exon 3 with a Lac Z. The expression profile of Bestrophin-2 (Best2) was examined using RT-PCR, X-gal staining, and immunohistochemistry. Intraocular pressure (IOP) was measured by anterior chamber cannulation. RESULTS RT-PCR of mouse tissues revealed Best2 mRNA in eye, colon, nasal epithelia, trachea, brain, lung, and kidney. X-gal staining, confirmed expression in colon epithelia and in the eye, in the nonpigmented epithelia (NPE). Best2 was not expressed in RPE cells. Best2 protein was observed only in NPE and colon epithelia. The absence of Best2 had no obvious deleterious effect on the mice. However, the Best2-/- mice were found to have significantly (P < 0.02) diminished IOP with respect to the Best2+/+ and Best2+/- littermates. The Best2-/- and Best2+/- mice responded better to the carbonic anhydrase inhibitor brinzolamide than did their Best2+/+ littermates, although the beta-blocker timolol brought IOP to the same level, regardless of genotype. CONCLUSIONS Best2 plays a role in the generation of IOP by regulating formation of aqueous humor, and inhibition of Best2 function represents an attractive new avenue for regulating IOP in individuals with glaucoma.
Collapse
Affiliation(s)
- Benjamin Bakall
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona
| | - Precious McLaughlin
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona
| | - J. Brett Stanton
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona
| | - Youwen Zhang
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona
| | | | - Lihua Y. Marmorstein
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Alan D. Marmorstein
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona
- College of Optical Sciences, University of Arizona, Tucson, Arizona
| |
Collapse
|
38
|
Hartzell HC, Qu Z, Yu K, Xiao Q, Chien LT. Molecular physiology of bestrophins: multifunctional membrane proteins linked to best disease and other retinopathies. Physiol Rev 2008; 88:639-72. [PMID: 18391176 DOI: 10.1152/physrev.00022.2007] [Citation(s) in RCA: 265] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This article reviews the current state of knowledge about the bestrophins, a newly identified family of proteins that can function both as Cl(-) channels and as regulators of voltage-gated Ca(2+) channels. The founding member, human bestrophin-1 (hBest1), was identified as the gene responsible for a dominantly inherited, juvenile-onset form of macular degeneration called Best vitelliform macular dystrophy. Mutations in hBest1 have also been associated with a small fraction of adult-onset macular dystrophies. It is proposed that dysfunction of bestrophin results in abnormal fluid and ion transport by the retinal pigment epithelium, resulting in a weakened interface between the retinal pigment epithelium and photoreceptors. There is compelling evidence that bestrophins are Cl(-) channels, but bestrophins remain enigmatic because it is not clear that the Cl(-) channel function can explain Best disease. In addition to functioning as a Cl(-) channel, hBest1 also is able to regulate voltage-gated Ca(2+) channels. Some bestrophins are activated by increases in intracellular Ca(2+) concentration, but whether bestrophins are the molecular counterpart of Ca(2+)-activated Cl(-) channels remains in doubt. Bestrophins are also regulated by cell volume and may be a member of the volume-regulated anion channel family.
Collapse
Affiliation(s)
- H Criss Hartzell
- Department of Cell Biology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | | | | | | |
Collapse
|
39
|
Fuller CM, Kovacs G, Anderson SJ, Benos DJ. The CLCAs: Proteins with Ion Channel, Cell Adhesion and Tumor Suppressor Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007. [DOI: 10.1007/0-387-23250-8_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
40
|
Qu ZQ, Yu K, Cui YY, Ying C, Hartzell C. Activation of bestrophin Cl- channels is regulated by C-terminal domains. J Biol Chem 2007; 282:17460-7. [PMID: 17442670 DOI: 10.1074/jbc.m701043200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bestrophins (VMD2, VMD2L1, VMD2L2, and VMD2L3) are a new family of anion channels. The mechanisms of their regulation are not yet well understood. Recently, we found that a domain (amino acids 356-364) in the C terminus of mouse VMD2L3 (mBest3) inhibited channel activity when it was expressed in HEK293 cells (Qu, Z., Cui, Y., and Hartzell, H. C. (2006) FEBS Lett. 580, 2141-2214). Here we show that this auto-inhibitory (AI) domain in mBest3 and human (h)Best3 is composed of seven critical residues, (356)IPSFLGS(362). Replacement of any residue (except Pro(357)) in the domain with alanine activated Cl(-) currents. Substitution of Pro(357) with other amino acids, especially phenylalanine, did activate currents. Membrane biotinylation demonstrated that nonfunctional mBest3 protein was trafficked to the plasma membrane, implying that the AI domain inhibited channel gating but not trafficking. mBest3-F359A and hBest3-G361A mutations induced outwardly rectifying currents, suggesting that the AI domain is associated with the channel pore or gating mechanism. Supporting this suggestion, the mBest3 AI domain was demonstrated to be located within a membrane-associated region. When the wild-type mBest3 C terminus (amino acids 292-669) was expressed in HEK293 cells, the protein was located mainly in the particulate fraction, but it became soluble when a sequence containing the AI domain was deleted (Delta353-404). There is an AI domain ((357)QPSFQGS(363)) in mouse VMD2L1 (mBest2) as well, but its inhibitory effect is competed by a downstream facilitatory sequence (amino acids 405-454). These results suggest that an auto-inhibitory mechanism in C termini may be universal among bestrophins investigated in the study.
Collapse
Affiliation(s)
- Zhi Qiang Qu
- Department of Cell Biology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | | | | | | |
Collapse
|
41
|
Marchant D, Yu K, Bigot K, Roche O, Germain A, Bonneau D, Drouin-Garraud V, Schorderet DF, Munier F, Schmidt D, Le Neindre P, Marsac C, Menasche M, Dufier JL, Fischmeister R, Hartzell C, Abitbol M. New VMD2 gene mutations identified in patients affected by Best vitelliform macular dystrophy. J Med Genet 2007; 44:e70. [PMID: 17287362 PMCID: PMC2598027 DOI: 10.1136/jmg.2006.044511] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE The mutations responsible for Best vitelliform macular dystrophy (BVMD) are found in a gene called VMD2. The VMD2 gene encodes a transmembrane protein named bestrophin-1 (hBest1) which is a Ca(2+)-sensitive chloride channel. This study was performed to identify disease-specific mutations in 27 patients with BVMD. Because this disease is characterised by an alteration in Cl(-) channel function, patch clamp analysis was used to test the hypothesis that one of the VMD2 mutated variants causes the disease. METHODS Direct sequencing analysis of the 11 VMD2 exons was performed to detect new abnormal sequences. The mutant of hBest1 was expressed in HEK-293 cells and the associated Cl(-) current was examined using whole-cell patch clamp analysis. RESULTS Six new VMD2 mutations were identified, located exclusively in exons four, six and eight. One of these mutations (Q293H) was particularly severe. Patch clamp analysis of human embryonic kidney cells expressing the Q293H mutant showed that this mutant channel is non-functional. Furthermore, the Q293H mutant inhibited the function of wild-type bestrophin-1 channels in a dominant negative manner. CONCLUSIONS This study provides further support for the idea that mutations in VMD2 are a necessary factor for Best disease. However, because variable expressivity of VMD2 was observed in a family with the Q293H mutation, it is also clear that a disease-linked mutation in VMD2 is not sufficient to produce BVMD. The finding that the Q293H mutant does not form functional channels in the membrane could be explained either by disruption of channel conductance or gating mechanisms or by improper trafficking of the protein to the plasma membrane.
Collapse
Affiliation(s)
- D Marchant
- Centre de recherche thérapeutique en ophtalmologie, équipe d'accueil 2502 MENRT, Université René Descartes Paris V, Faculté de Médecine Necker-Enfants Malades, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Milenkovic VM, Rivera A, Horling F, Weber BHF. Insertion and topology of normal and mutant bestrophin-1 in the endoplasmic reticulum membrane. J Biol Chem 2006; 282:1313-21. [PMID: 17110374 DOI: 10.1074/jbc.m607383200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The vitelliform macular dystrophy type 2 (VMD2) gene mutated in Best macular dystrophy encodes a 585-amino acid putative transmembrane protein termed bestrophin-1. The vast majority of known disease-associated alterations are of the missense type, which cluster near predicted transmembrane domains (TMDs). To investigate bestrophin-1 membrane topology and to assess consequences of point mutations on membrane integration, we have analyzed the insertion of putative TMDs into the endoplasmic reticulum (ER) membrane. Out of six potential TMDs, our data suggest a topological model of bestrophin-1 with four transmembrane-spanning segments and one large cytoplasmatic loop between putative TMD2 and TMD5. Consequently, a relatively hydrophobic segment containing putative TMD3 (aa 130-149) and TMD4 (aa 179-201) is located within the cytoplasm. Furthermore, we show that three out of 18 disease-associated alterations investigated (I73N, Y85H, F281del) reveal measurable effects on membrane insertion suggesting that defective membrane integration of bestrophin-1 may represent a potential disease mechanism for a small subset of Best macular dystrophy-related mutations.
Collapse
Affiliation(s)
- Vladimir M Milenkovic
- Institute of Physiology, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053 Regensburg, Germany
| | | | | | | |
Collapse
|
43
|
Hagen AR, Barabote RD, Saier MH. The bestrophin family of anion channels: identification of prokaryotic homologues. Mol Membr Biol 2006; 22:291-302. [PMID: 16154901 DOI: 10.1080/09687860500129711] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The human disease protein, Bestrophin-1, associated with vitelliform macular dystrophy, has recently been shown to be an integral membrane anion channel-forming protein. In this study we have recovered all bestrophin homologues from the NCBI database and analyzed their sequences using bioinformatic approaches. Eukaryotic homologues were found in animals and fungi but not in plants or protozoans, and prokaryotic homologues distantly related to the eukaryotic proteins, were identified in certain Gram-negative bacterial kingdoms but not in Gram-positive bacteria or archaea. Our analyses suggest a uniform 4 TMS topology for most of these homologues with regions of conservation overlapping and preceding the odd numbered TMSs and overlapping and following the even numbered TMSs. Well-conserved motifs were identified in both the eukaryotic and the prokaryotic homologues, and these proved to overlap, suggesting common structural and functional properties. Phylogenetic analyses revealed that the eukaryotic proteins cluster according to organismal type, and that the prokaryotic proteins sometimes (but not always) do so. This suggests that eukaryotic paralogues arose exclusively by recent gene duplication events although both early and late gene duplication events occurred in prokaryotes.
Collapse
Affiliation(s)
- Andrew R Hagen
- Division of Biological Sciences, University of California at San Diego, La Jolla, CA, 92093-0116, USA
| | | | | |
Collapse
|
44
|
Abstract
The retinal pigment epithelium (RPE) lying distal to the retina regulates the extracellular environment and provides metabolic support to the outer retina. RPE abnormalities are closely associated with retinal death and it has been claimed several of the most important diseases causing blindness are degenerations of the RPE. Therefore, the study of the RPE is important in Ophthalmology. Although visualisation of the RPE is part of clinical investigations, there are a limited number of methods which have been used to investigate RPE function. One of the most important is a study of the current generated by the RPE. In this it is similar to other secretory epithelia. The RPE current is large and varies as retinal activity alters. It is also affected by drugs and disease. The RPE currents can be studied in cell culture, in animal experimentation but also in clinical situations. The object of this review is to summarise this work, to relate it to the molecular membrane mechanisms of the RPE and to possible mechanisms of disease states.
Collapse
Affiliation(s)
- Geoffrey B Arden
- Department of Optometry and Visual Science, Henry Wellcome Laboratiories for Visual Sciences, City University, London, UK.
| | | |
Collapse
|
45
|
Qu Z, Cui Y, Hartzell C. A short motif in the C-terminus of mouse bestrophin 3 [corrected] inhibits its activation as a Cl channel. FEBS Lett 2006; 580:2141-6. [PMID: 16563389 DOI: 10.1016/j.febslet.2006.03.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 03/07/2006] [Accepted: 03/08/2006] [Indexed: 11/19/2022]
Abstract
Bestrophins are a new family of anion channels. Here, we examined the Cl channel activity of mBest4. Surprisingly, wild type mouse bestrophin-4 (mBest4) did not induce functional Cl channels when over-expressed in HEK293 cells. However, deletion of part of the C-terminus (residues 353-669) produced large Cl currents, suggesting the presence of a C-terminal motif that inhibited Cl channel function. Deletion of a short motif (356-364) or substitution of certain residues in this motif with alanines also resulted in expression of robust Cl currents. The channel activity of the mBest4 protein lacking the C-terminus (residues 353-669) was specifically inhibited by co-expression of C-terminal fragments of mBest4 having the inhibitory motif, suggesting that the C-terminal motif blocked mBest4 channel activity probably by interacting with the channel pore.
Collapse
Affiliation(s)
- Zhiqiang Qu
- Department of Cell Biology and Center for Neurodegenerative Disease, Emory University School of Medicine, 615 Michael Street, 535 Whitehead Building, Atlanta, GA 30322-3030, USA.
| | | | | |
Collapse
|
46
|
Stanton JB, Goldberg AF, Hoppe G, Marmorstein LY, Marmorstein AD. Hydrodynamic properties of porcine bestrophin-1 in Triton X-100. BIOCHIMICA ET BIOPHYSICA ACTA 2006; 1758:241-7. [PMID: 16600174 PMCID: PMC2832839 DOI: 10.1016/j.bbamem.2006.01.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Revised: 01/24/2006] [Accepted: 01/27/2006] [Indexed: 11/20/2022]
Abstract
Bestrophin-1 (Best-1) is an integral membrane protein, defects in which cause Best vitelliform macular dystrophy. Best-1 is proposed to function as a Cl- channel and/or a regulator of Ca++ channels. A tetrameric (or pentameric) stoichiometry has been reported for recombinant best-1. Using a combination of gel exclusion chromatography and velocity sedimentation we examined the quaternary structure of native best-1 and found that it migrates as a single species with a Stokes radius of 7.3 nm, sedimentation coefficient (S20,w) of 4.9, and partial specific volume (nu) of 0.80 ml/g. The mass of the protein-detergent complex is calculated to be 206 kDa, with the protein component estimated to be approximately 138 kDa. Given a monomeric mass of 68 kDa, we conclude that native best-1 solubilized with Triton X-100 is a homodimer. The differences between this observation and a prior report were examined by comparing recombinant best-1 with tissue derived best-1 using gel exclusion chromatography. Much of the recombinant best-1 eluted in the column void (Vo) fraction, unlike that extracted from RPE cells. We conclude that the minimal functional unit of best-1 is dimeric. This stoichiometry differs from that previously measured for recombinant best-1, suggesting that further studies are necessary to determine the stoichiometry of functional best-1 in RPE membranes.
Collapse
Affiliation(s)
- J. Brett Stanton
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ 85711, USA
| | | | - George Hoppe
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Lihua Y. Marmorstein
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ 85711, USA
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, AZ 85724, USA
| | - Alan D. Marmorstein
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ 85711, USA
- College of Optical Sciences, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
47
|
Rosenthal R, Bakall B, Kinnick T, Peachey N, Wimmers S, Wadelius C, Marmorstein A, Strauss O. Expression of bestrophin‐1, the product of the VMD2 gene, modulates voltage‐dependent Ca
2+
channels in retinal pigment epithelial cells. FASEB J 2006; 20:178-80. [PMID: 16282372 DOI: 10.1096/fj.05-4495fje] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mutations in the VMD2 gene cause Best's disease, an inherited form of macular degeneration. The reduction in the light-peak amplitude in the patient's electro-oculogram suggests that bestrophin-1 influences the membrane conductance of the retinal pigment epithelium (RPE). Systemic application of the L-type Ca2+ channel blocker nimodipine reduced the light-peak amplitude in the rat electroretinogram but not a- and b-waves. Expression of bestrophin-1 in a RPE cell line (RPE-J) led to changes in L-type channel properties. Wild-type bestrophin-1 induced an acceleration of activation kinetics of Ba2+ currents through L-type Ca2+ channels and a shift of the voltage-dependent activation to more negative values, closer to the resting potential of RPE cells. Expression of bestrophin-1 with Best disease-causing mutations led to comparable shifts in voltage-dependent activation but different effects on activation and inactivation kinetics. Bestrophin W93C exhibited slowed activation and inactivation, and bestrophin R218C accelerated the activation and inactivation. Thus, transfection of RPE cells with bestrophin-1 distinctively changed L-type Ca2+ channel kinetics and voltage-dependence. On the basis of these data, we propose that presence of bestrophin-1 influences kinetics and voltage-dependence of voltage-dependent Ca2+ channels and that these effects might open new ways to understand the mechanisms leading to retinal degeneration in Best's disease.
Collapse
Affiliation(s)
- Rita Rosenthal
- Augenklinik, Charité-Universitaetsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Hartzell C, Qu Z, Putzier I, Artinian L, Chien LT, Cui Y. Looking chloride channels straight in the eye: bestrophins, lipofuscinosis, and retinal degeneration. Physiology (Bethesda) 2005; 20:292-302. [PMID: 16174869 DOI: 10.1152/physiol.00021.2005] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent evidence suggests that Cl(-) ion channels are important for retinal integrity. Bestrophin Cl(-) channel mutations in humans are genetically linked to a juvenile form of macular degeneration, and disruption of some ClC Cl(-) channels in mice leads to retinal degeneration. In both cases, accumulation of lipofuscin pigment is a key feature of the cellular degeneration. Because Cl(-) channels regulate the ionic environment inside organelles in the endosomal-lysosomal pathway, retinal degeneration may result from defects in lysosomal trafficking or function.
Collapse
Affiliation(s)
- Criss Hartzell
- Department of Cell Biology, The Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Located between vessels of the choriocapillaris and light-sensitive outer segments of the photoreceptors, the retinal pigment epithelium (RPE) closely interacts with photoreceptors in the maintenance of visual function. Increasing knowledge of the multiple functions performed by the RPE improved the understanding of many diseases leading to blindness. This review summarizes the current knowledge of RPE functions and describes how failure of these functions causes loss of visual function. Mutations in genes that are expressed in the RPE can lead to photoreceptor degeneration. On the other hand, mutations in genes expressed in photoreceptors can lead to degenerations of the RPE. Thus both tissues can be regarded as a functional unit where both interacting partners depend on each other.
Collapse
Affiliation(s)
- Olaf Strauss
- Bereich Experimentelle Ophthalmologie, Klinik und Poliklinik fuer Augenheilkunde, Universitaetsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
50
|
Abstract
CLCA proteins were discovered in bovine trachea and named for a calcium-dependent chloride conductance found in trachea and in other secretory epithelial tissues. At least four closely located gene loci in the mouse and the human code for independent isoforms of CLCA proteins. Full-length CLCA proteins have an unprocessed mass ratio of approximately 100 kDa. Three of the four human loci code for the synthesis of membrane-associated proteins. CLCA proteins affect chloride conductance, epithelial secretion, cell-cell adhesion, apoptosis, cell cycle control, mucus production in asthma, and blood pressure. There is a structural and probable functional divergence between CLCA isoforms containing or not containing beta4-integrin binding domains. Cell cycle control and tumor metastasis are affected by isoforms with the binding domains. These isoforms are expressed prominently in smooth muscle, in some endothelial cells, in the central nervous system, and also in secretory epithelial cells. The isoform with disrupted beta4-integrin binding (hCLCA1, pCLCA1, mCLCA3) alters epithelial mucus secretion and ion transport processes. It is preferentially expressed in secretory epithelial tissues including trachea and small intestine. Chloride conductance is affected by the expression of several CLCA proteins. However, the dependence of the resulting electrical signature on the expression system rather than the CLCA protein suggests that these proteins are not independent Ca2+-dependent chloride channels, but may contribute to the activity of chloride channels formed by, or in conjunction with, other proteins.
Collapse
Affiliation(s)
- Matthew E Loewen
- Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, Canada
| | | |
Collapse
|