1
|
Lin SJ, Huang K, Petree C, Qin W, Varshney P, Varshney G. Optimizing gRNA selection for high-penetrance F0 CRISPR screening for interrogating disease gene function. Nucleic Acids Res 2025; 53:gkaf180. [PMID: 40103232 PMCID: PMC11915512 DOI: 10.1093/nar/gkaf180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Genes and genetic variants associated with human disease are continually being discovered, but validating their causative roles and mechanisms remains a significant challenge. CRISPR/Cas9 genome editing in model organisms like zebrafish can enable phenotypic characterization of founder generation (F0) knockouts (Crispants), but existing approaches are not amenable to high-throughput genetic screening due to high variability, cost, and low phenotype penetrance. To overcome these challenges, here we provide guide RNA (gRNA) selection rules that enable high phenotypic penetrance of up to three simultaneous knockouts in F0 animals following injection of 1-2 gRNAs per gene. We demonstrate a strong transcriptomic overlap in our F0 knockouts and stable knockout lines that take several months to generate. We systematically evaluated this approach across 324 gRNAs targeting 125 genes and demonstrated its utility in studying epistasis, characterizing paralogous genes, and validating human disease gene phenotypes across multiple tissues. Applying our approach in a high-throughput manner, we screened and identified 10 novel neurodevelopmental disorders and 50 hearing genes not previously studied in zebrafish. Altogether, our approach achieves high phenotypic penetrance using low numbers of gRNAs per gene in F0 zebrafish, offering a robust pipeline for rapidly characterizing candidate human disease genes.
Collapse
Affiliation(s)
- Sheng-Jia Lin
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Kevin Huang
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Cassidy Petree
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Wei Qin
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Pratishtha Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Gaurav K Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| |
Collapse
|
2
|
Smaili W, Pezet C, Marlin S, Ernest S. R391 human dominant mutation does not affect TubB4b localization and sensory hair cells structure in zebrafish inner ear and lateral line. Dev Biol 2025; 517:301-316. [PMID: 39515407 DOI: 10.1016/j.ydbio.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 10/24/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Heterozygous R391 TUBB4B pathogenic variations are responsible for an association of hearing loss and retinal dystrophy in human. With the goal of understanding the functions of TuBB4b and the pathogenic role of R391 variations, we characterized tubB4B in zebrafish and identified the gene regulatory elements necessary and sufficient for expression of TubB4b as in endogenous tissues. Using knock-out and transgenic approaches, we determined that R391 mutations impair neither localization of TubB4B within sensory hair cells (SHC) nor their structure, but induced to a small decrease in SHC number from anterior crista. Expression of R391 mutations in sensory hair cells has no effect on zebrafish audition, suggesting a different equilibrium between various tubulin isotypes in zebrafish possibly due to compensatory mechanisms. The careful expression analysis and transgenic tools generated in this study could help understand how recently described pathogenic variants lead to more severe clinical forms of TUBB4B-related diseases.
Collapse
Affiliation(s)
- Wiam Smaili
- Université Paris Cité, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, F-75015, Paris, France
| | - Camille Pezet
- Université Paris Cité, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, F-75015, Paris, France
| | - Sandrine Marlin
- Université Paris Cité, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, F-75015, Paris, France; Centre de référence des surdités génétiques, Hôpital Necker-Enfants Malades, AP-HP, F-75015, Paris, France
| | - Sylvain Ernest
- Université Paris Cité, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, F-75015, Paris, France.
| |
Collapse
|
3
|
Luo C, Zhang Q, Zheng S, Wang D, Huang W, Huang Y, Shi X, Xie H, Wu K. Visual toxicity in zebrafish larvae following exposure to 2,2',4,4'-tetrabromodiphenyl ether (BDE-47), triphenyl phosphate (TPhP), and isopropyl phenyl diphenyl phosphate (IPPP). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175131. [PMID: 39127212 DOI: 10.1016/j.scitotenv.2024.175131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024]
Abstract
TPhP and IPPP, alternatives to PBDEs as flame retardants, have been studied for their developmental toxicity, but their visual toxicities are less understood. In this study, zebrafish larvae were exploited to evaluate the potential ocular impairments following exposure to BDE-47, TPhP, and IPPP. The results revealed a range of ocular abnormalities, including malformation, vascular issues within the eyes, and histopathological changes in the retina. Notably, the visually mediated behavioral changes were primarily observed in IPPP and TPhP, indicating that they caused more severe eye malformations and vision impairment than BDE-47. Molecular docking and MD simulations showed stronger binding affinity of TPhP and IPPP to RAR and RBP receptors. Elevated ROS and T3 levels induced by these compounds led to apoptosis in larvae eyes, and increased GABA levels induced by TPhP and IPPP hindered retinal repair. In summary, our results indicate TPhP and IPPP exhibit severer visual toxicity than BDE-47, affecting eye development and visually guided behaviors. The underlying mechanism involves disruptions in RA signaling, retinal neurotransmitters imbalance, thyroid hormones up-regulation, and apoptosis in larvae eyes. This work highlights novel insights into the need for cautious use of these flame retardants due to their potential biological hazards, thereby offering valuable guidance for their safer applications.
Collapse
Affiliation(s)
- Congying Luo
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Qiong Zhang
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Shukai Zheng
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Dinghui Wang
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Wenlong Huang
- Department of Forensic Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Yanhong Huang
- Mental Health Center of Shantou University, Shantou, Guangdong, China
| | - Xiaoling Shi
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Han Xie
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Kusheng Wu
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515041, Guangdong, China.
| |
Collapse
|
4
|
Giffen KP, Liu H, Yamane KL, Li Y, Chen L, Kramer KL, Zallocchi M, He DZ. Molecular specializations underlying phenotypic differences in inner ear hair cells of zebrafish and mice. Front Neurol 2024; 15:1437558. [PMID: 39484049 PMCID: PMC11524865 DOI: 10.3389/fneur.2024.1437558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction Hair cells (HCs) are the sensory receptors of the auditory and vestibular systems in the inner ears of vertebrates that selectively transduce mechanical stimuli into electrical activity. Although all HCs have the hallmark stereocilia bundle for mechanotransduction, HCs in non-mammals and mammals differ in their molecular specialization in the apical, basolateral, and synaptic membranes. HCs of non-mammals, such as zebrafish (zHCs), are electrically tuned to specific frequencies and possess an active process in the stereocilia bundle to amplify sound signals. Mammalian HCs, in contrast, are not electrically tuned and achieve amplification by somatic motility of outer HCs (OHCs). Methods To understand the genetic mechanisms underlying differences between adult zebrafish and mammalian HCs, we compared their RNA-seq-characterized transcriptomes, focusing on protein-coding orthologous genes related to HC specialization. Results There was considerable shared expression of gene orthologs among the HCs, including those genes associated with mechanotransduction, ion transport/channels, and synaptic signaling. However, there were some notable differences in expression among zHCs, OHCs, and inner HCs (IHCs), which likely underlie the distinctive physiological properties of each cell type. For example, OHCs highly express Slc26a5 which encodes the motor protein prestin that contributes to OHC electromotility. However, zHCs have only weak expression of slc26a5, and subsequently showed no voltage-dependent electromotility when measured. Notably, the zHCs expressed more paralogous genes including those associated with HC-specific functions and transcriptional activity, though it is unknown whether they have functions similar to their mammalian counterparts. There was overlap in the expressed genes associated with a known hearing phenotype. Discussion Our analyses unveil substantial differences in gene expression patterns that may explain phenotypic specialization of zebrafish and mouse HCs. This dataset also includes several protein-coding genes to further the functional characterization of HCs and study of HC evolution from non-mammals to mammals.
Collapse
Affiliation(s)
- Kimberlee P. Giffen
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Department of Basic Sciences, Augusta University/University of Georgia Medical Partnership, Athens, GA, United States
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Huizhan Liu
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Kacey L. Yamane
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Yi Li
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
- Department of Otorhinolaryngology, Beijing Tongren Hospital, Beijing Capital Medical University, Beijing, China
| | - Lei Chen
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Kenneth L. Kramer
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Marisa Zallocchi
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - David Z. He
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
5
|
Maniaci A, Briglia M, Allia F, Montalbano G, Romano GL, Zaouali MA, H’mida D, Gagliano C, Malaguarnera R, Lentini M, Graziano ACE, Giurdanella G. The Role of Pericytes in Inner Ear Disorders: A Comprehensive Review. BIOLOGY 2024; 13:802. [PMID: 39452111 PMCID: PMC11504721 DOI: 10.3390/biology13100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024]
Abstract
Inner ear disorders, including sensorineural hearing loss, Meniere's disease, and vestibular neuritis, are prevalent conditions that significantly impact the quality of life. Despite their high incidence, the underlying pathophysiology of these disorders remains elusive, and current treatment options are often inadequate. Emerging evidence suggests that pericytes, a type of vascular mural cell specialized to maintain the integrity and function of the microvasculature, may play a crucial role in the development and progression of inner ear disorders. The pericytes are present in the microvasculature of both the cochlea and the vestibular system, where they regulate blood flow, maintain the blood-labyrinth barrier, facilitate angiogenesis, and provide trophic support to neurons. Understanding their role in inner ear disorders may provide valuable insights into the pathophysiology of these conditions and lead to the development of novel diagnostic and therapeutic strategies, improving the standard of living. This comprehensive review aims to provide a detailed overview of the role of pericytes in inner ear disorders, highlighting the anatomy and physiology in the microvasculature, and analyzing the mechanisms that contribute to the development of the disorders. Furthermore, we explore the potential pericyte-targeted therapies, including antioxidant, anti-inflammatory, and angiogenic approaches, as well as gene therapy strategies.
Collapse
Affiliation(s)
- Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Marilena Briglia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Fabio Allia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Laboratory, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| | - Giovanni Luca Romano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mohamed Amine Zaouali
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Avicenne Street, 5019 Monastir, Tunisia;
| | - Dorra H’mida
- Department of Cytogenetics and Reproductive Biology, Farhat Hached Hospital, 4021 Sousse, Tunisia;
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Roberta Malaguarnera
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mario Lentini
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Adriana Carol Eleonora Graziano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| |
Collapse
|
6
|
Wu LY, Zhang JL, Zeeshan M, Zhou Y, Zhang YT, He WT, Jin N, Dai Y, Chi W, Ou Z, Dong GH, Lin LZ. Caspase-8 promotes NLRP3 inflammasome activation mediates eye development defects in zebrafish larvae exposed to perfulorooctane sulfonate (PFOS). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 356:124252. [PMID: 38815886 DOI: 10.1016/j.envpol.2024.124252] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/18/2024] [Accepted: 05/26/2024] [Indexed: 06/01/2024]
Abstract
Epidemiological evidence showed that serum high perfluorooctane sulfonate (PFOS) levels are associated with multiple eye related diseases, but the potential underlying molecular mechanisms remain poorly understood. Zebrafish and photoreceptor cell (661w) models were used to investigate the molecular mechanism of PFOS induced eye development defects. Our results showed a novel molecular mechanism of PFOS-induced inflammation response-mediated photoreceptor cell death associated with eye development defects. Inhibition of Caspase-8 activation significantly decreased photoreceptor cell death in PFOS exposure. Mechanistically, Toll-like receptor 4 (TLR4) mediates activation of Caspase-8 promote activation of NLR family pyrin domain-containing 3 (NLRP3) inflammasome to elicit maturation of interleukin-1 beta (IL-1β) via Caspase-1 activation, facilitating photoreceptor cell inflammation damage in PFOS exposure. In addition, we also made a novel finding that Caspase-3 activation was increased via Caspase-8 activation and directly intensified cell death. Our results show the important role of Caspase-8 activation in PFOS induced eye development defects and highlight Caspase-8 mediated activation of the NLRP3 inflammation triggers activation of Caspase-1 and promote the maturation of IL-1β in retinal inflammatory injury.
Collapse
Affiliation(s)
- Lu-Yin Wu
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing-Lin Zhang
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mohammed Zeeshan
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yang Zhou
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Environmental Protection, Guangzhou, 510655, China
| | - Yun-Ting Zhang
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wan-Ting He
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Nanxiang Jin
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70210, Kuopio, Finland
| | - Ye Dai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, Guangdong, China
| | - Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, Guangdong, China
| | - Zejin Ou
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Guang-Hui Dong
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Zi Lin
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Niksirat H, Siino V, Steinbach C, Levander F. The quantification of zebrafish ocular-associated proteins provides hints for sex-biased visual impairments and perception. Heliyon 2024; 10:e33057. [PMID: 38994070 PMCID: PMC11238053 DOI: 10.1016/j.heliyon.2024.e33057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Biochemical differences between sexes can also be seen in non-sexual organs and may affect organ functions and susceptibility to diseases. It has been shown that there are sex-biased visual perceptions and impairments. Abundance differences of eye proteins could provide explanations for some of these. Exploration of the ocular proteome was performed to find sex-based protein abundance differences in zebrafish Danio rerio. A label-free protein quantification workflow using high-resolution mass spectrometry was employed to find proteins with significant differences between the sexes. In total, 3740 unique master proteins were identified and quantified, and 49 proteins showed significant abundance differences between the eyes of male and female zebrafish. Those proteins belong to lipoproteins, immune system, blood coagulation, antioxidants, iron and heme-binding proteins, ion channels, pumps and exchangers, neuronal and photoreceptor proteins, and the cytoskeleton. An extensive literature review provided clues for the possible links between the sex-biased level of proteins and visual perception and impairments. In conclusion, sexual dimorphism at the protein level was discovered for the first time in the eye of zebrafish and should be accounted for in ophthalmological studies. Data are available via ProteomeXchange with identifier PXD033338.
Collapse
Affiliation(s)
- Hamid Niksirat
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Valentina Siino
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Christoph Steinbach
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund, Sweden
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
8
|
Han S, Wang Q, Cheng M, Hu Y, Liu P, Hou W, Liang L. The effects of ush2a gene knockout on vesicle transport in photoreceptors. Gene 2024; 892:147885. [PMID: 37813209 DOI: 10.1016/j.gene.2023.147885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 10/11/2023]
Abstract
USH2A (Usher syndrome type 2A) gene mutations are the predominant cause of Usher syndrome type 2, characterized by sensorineural hearing loss and retinitis pigmentosa (RP), and also significant contributors to non-syndromic RP. To date, there is a lack of definitive therapeutic interventions to mitigate the associated disorders caused by USH2A mutations, and the precise pathogenic mechanisms underlying their onset remain unclear. In the present study, we utilized the ush2a knockout zebrafish model to investigate the pathological mechanisms of RP. In late-stage ush2a-/- zebrafish, the outer segments of rods displayed shortened length and decreased number. Anomalous vesicle accumulation was observed at the junction between the inner and outer segments, accompanied by reduced expression and structural damage of actin filaments in the photoreceptor cells. Furthermore, we discovered that Rab8 expression was downregulated and exhibited aberrant localization in ush2a-/- zebrafish. Additionally, we identified an interaction between USH2A and Rab8. Therefore, the knockout of ush2a may potentially affect vesicle transport through the regulation of Rab8, providing a novel target for maintaining the survival of photoreceptor cells. These findings also contribute to our understanding of the potential molecular pathogenesis underlying RP caused by USH2A gene mutations.
Collapse
Affiliation(s)
- Shanshan Han
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Qiong Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Meiqi Cheng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Yue Hu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Pei Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Wanle Hou
- Department of Clinical Laboratory, the First Hospital of Laohekou City, Laohekou 441800, Hubei, China.
| | - Liang Liang
- Department of Ophthalmology, the First Clinical Hospital of China Three Gorges University, Yichang 443003, Hubei, China.
| |
Collapse
|
9
|
Loiseau A, Raîche-Marcoux G, Maranda C, Bertrand N, Boisselier E. Animal Models in Eye Research: Focus on Corneal Pathologies. Int J Mol Sci 2023; 24:16661. [PMID: 38068983 PMCID: PMC10706114 DOI: 10.3390/ijms242316661] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 09/27/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
The eye is a complex sensory organ that enables visual perception of the world. The dysfunction of any of these tissues can impair vision. Conduction studies on laboratory animals are essential to ensure the safety of therapeutic products directly applied or injected into the eye to treat ocular diseases before eventually proceeding to clinical trials. Among these tissues, the cornea has unique homeostatic and regenerative mechanisms for maintaining transparency and refraction of external light, which are essential for vision. However, being the outermost tissue of the eye and directly exposed to the external environment, the cornea is particularly susceptible to injury and diseases. This review highlights the evidence for selecting appropriate animals to better understand and treat corneal diseases, which rank as the fifth leading cause of blindness worldwide. The development of reliable and human-relevant animal models is, therefore, a valuable research tool for understanding and translating fundamental mechanistic findings, as well as for assessing therapeutic potential in humans. First, this review emphasizes the unique characteristics of animal models used in ocular research. Subsequently, it discusses current animal models associated with human corneal pathologies, their utility in understanding ocular disease mechanisms, and their role as translational models for patients.
Collapse
Affiliation(s)
- Alexis Loiseau
- Faculty of Medicine, Department of Ophthalmology and Otolaryngology—Head and Neck Surgery, CHU de Québec Research Center, Université Laval, Québec, QC G1S 4L8, Canada; (G.R.-M.); (C.M.)
| | - Gabrielle Raîche-Marcoux
- Faculty of Medicine, Department of Ophthalmology and Otolaryngology—Head and Neck Surgery, CHU de Québec Research Center, Université Laval, Québec, QC G1S 4L8, Canada; (G.R.-M.); (C.M.)
| | - Cloé Maranda
- Faculty of Medicine, Department of Ophthalmology and Otolaryngology—Head and Neck Surgery, CHU de Québec Research Center, Université Laval, Québec, QC G1S 4L8, Canada; (G.R.-M.); (C.M.)
| | - Nicolas Bertrand
- Faculty of Pharmacy, CHU de Quebec Research Center, Université Laval, Québec, QC G1V 4G2, Canada;
| | - Elodie Boisselier
- Faculty of Medicine, Department of Ophthalmology and Otolaryngology—Head and Neck Surgery, CHU de Québec Research Center, Université Laval, Québec, QC G1S 4L8, Canada; (G.R.-M.); (C.M.)
| |
Collapse
|
10
|
Yi J, Ma Y, Ma J, Yu H, Zhang K, Jin L, Yang Q, Sun D, Wu D. Rapid Assessment of Ocular Toxicity from Environmental Contaminants Based on Visually Mediated Zebrafish Behavior Studies. TOXICS 2023; 11:706. [PMID: 37624211 PMCID: PMC10459940 DOI: 10.3390/toxics11080706] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
The presence of contaminants in the environment has increased in recent years, and studies have demonstrated that these contaminants have the ability to penetrate the blood-retinal barrier and directly affect the visual systems of organisms. Zebrafish are recognized as an ideal model for human eye diseases due to their anatomical and functional similarities to the human eye, making them an efficient and versatile organism for studying ocular toxicity caused by environmental contaminants in the field of environmental toxicology. Meanwhile, zebrafish exhibit a diverse repertoire of visually mediated behaviors, and their visual system undergoes complex changes in behavioral responses when exposed to environmental contaminants, enabling rapid assessment of the ocular toxicity induced by such pollutants. Therefore, this review aimed to highlight the effectiveness of zebrafish as a model for examining the effects of environmental contaminants on ocular development. Special attention is given to the visually mediated behavior of zebrafish, which allows for a rapid assessment of ocular toxicity resulting from exposure to environmental contaminants. Additionally, the potential mechanisms by which environmental contaminants may induce ocular toxicity are briefly outlined.
Collapse
Affiliation(s)
- Jia Yi
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yilei Ma
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Ma
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Kun Zhang
- Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Libo Jin
- National and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China;
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China;
| | - Da Sun
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
- National and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China;
| | - Dejun Wu
- Emergency Department, Quzhou People’s Hospital, Quzhou 324000, China
| |
Collapse
|
11
|
Bielak K, Benkowska-Biernacka D, Ptak M, Stolarski J, Kalka M, Ożyhar A, Dobryszycki P. Otolin-1, an otolith- and otoconia-related protein, controls calcium carbonate bioinspired mineralization. Biochim Biophys Acta Gen Subj 2023; 1867:130327. [PMID: 36791829 DOI: 10.1016/j.bbagen.2023.130327] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Otoliths and otoconia are calcium carbonate biomineral structures that form in the inner ear of fish and humans, respectively. The formation of these structures is tightly linked to the formation of an organic matrix framework with otolin-1, a short collagen-like protein from the C1q family as one of its major constituents. METHODS In this study, we examined the activity of recombinant otolin-1 originating from Danio rerio and Homo sapiens on calcium carbonate bioinspired mineralization with slow-diffusion method and performed crystals characterization with scanning electron microscopy, two-photon excited fluorescence microscopy, confocal laser scanning microscopy and micro-Raman spectroscopy. RESULTS We show that both proteins are embedded in the core of CaCO3 crystals that form through the slow-diffusion mineralization method. Both of them influence the morphology but do not change the polymorphic mineral phase. D.rerio otolin-1 also closely adheres to the crystal surface. GENERAL SIGNIFICANCE The results suggest, that otolin-1 is not a passive scaffold, but is directly involved in regulating the morphology of the resulting calcium carbonate biocrystals.
Collapse
Affiliation(s)
- Klaudia Bielak
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspiańskiego 27, 50-370 Wroclaw, Poland
| | - Dominika Benkowska-Biernacka
- Institute of Advanced Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Maciej Ptak
- Division of Optical Spectroscopy, Institute of Low Temperature and Structure Research, Polish Academy of Sciences, ul. Okólna 2, 50-422 Wroclaw, Poland
| | - Jarosław Stolarski
- Institute of Paleobiology, Polish Academy of Sciences, ul. Twarda 51/55, 00-818 Warsaw, Poland
| | - Marta Kalka
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspiańskiego 27, 50-370 Wroclaw, Poland
| | - Andrzej Ożyhar
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspiańskiego 27, 50-370 Wroclaw, Poland
| | - Piotr Dobryszycki
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspiańskiego 27, 50-370 Wroclaw, Poland.
| |
Collapse
|
12
|
Vöcking O, Famulski JK. A temporal single cell transcriptome atlas of zebrafish anterior segment development. Sci Rep 2023; 13:5656. [PMID: 37024546 PMCID: PMC10079958 DOI: 10.1038/s41598-023-32212-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
Anterior segment dysgenesis (ASD), resulting in vision impairment, stems from maldevelopment of anterior segment (AS) tissues. Incidence of ASD has been linked to malfunction of periocular mesenchyme cells (POM). POM cells specify into anterior segment mesenchyme (ASM) cells which colonize and produce AS tissues. In this study we uncover ASM developmental trajectories associated with formation of the AS. Using a transgenic line of zebrafish that fluorescently labels the ASM throughout development, Tg[foxc1b:GFP], we isolated GFP+ ASM cells at several developmental timepoints (48-144 hpf) and performed single cell RNA sequencing. Clustering analysis indicates subdifferentiation of ASM as early as 48 hpf and subsequent diversification into corneal epithelium/endothelium/stroma, or annular ligament (AL) lineages. Tracking individual clusters reveals common developmental pathways, up to 72 hpf, for the AL and corneal endothelium/stroma and distinct pathways for corneal epithelium starting at 48 hpf. Spatiotemporal validation of over 80 genes found associated with AS development demonstrates a high degree of conservation with mammalian trabecular meshwork and corneal tissues. In addition, we characterize thirteen novel genes associated with annular ligament and seven with corneal development. Overall, the data provide a molecular verification of the long-standing hypothesis that POM derived ASM give rise to AS tissues and highlight the high degree of conservation between zebrafish and mammals.
Collapse
Affiliation(s)
- Oliver Vöcking
- Department of Biology, University of Kentucky, Lexington, USA
| | - J K Famulski
- Department of Biology, University of Kentucky, Lexington, USA.
| |
Collapse
|
13
|
Ma X, Dai Y, Qiu T, Chen X, Xiao P, Li W. Effects of acute exposure to amisulbrom on retinal development in zebrafish (Danio rerio) embryos. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:46248-46256. [PMID: 36715803 DOI: 10.1007/s11356-023-25584-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023]
Abstract
Amisulbrom is an oomycete-specific fungicide that was developed by Nissan Chemical Industries Limited. The exposure of developing zebrafish embryo to amisulbrom caused disorders in the visual phototransduction system. However, the potential toxic mechanisms of amisulbrom on retinal development remains unclear. The research purpose of this study was to evaluate the adverse effects of amisulbrom on retinal development in a model organism, the zebrafish. Zebrafish embryos were treated with 0, 0.0075, 0.075, or 0.75 μM amisulbrom from 3 h post-fertilization (hpf) to 72 hpf. Compared with the control group, amisulbrom-treated zebrafish embryos displayed phenotypic microphthalmia, dysregulation of gene transcription levels (alcama, prox1a, sox2, vsx1, rho, bluops, rdops, uvops, and grops) related to the retinal cell layer differentiation, and increased retinal apoptosis. In addition, the content of glutathione and malondialdehyde increased significantly after exposure to amisulbrom. Overall, our data demonstrate the toxicity of amisulbrom to eye development, which will help to assess the potential ecotoxicological impacts posed by amisulbrom to aquatic species.
Collapse
Affiliation(s)
- Xueying Ma
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Yizhe Dai
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Tiantong Qiu
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Xin Chen
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Peng Xiao
- National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution, Zhejiang Provincial Key Lab for Water Environment and Marine Biological Resources Protection, Institute for Eco-Environmental Research of Sanyang Wetland, College of Life and Environmental Science, Wenzhou University, 325035, Wenzhou, People's Republic of China
| | - Wenhua Li
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China.
| |
Collapse
|
14
|
Wu L, Zeeshan M, Dang Y, Liang LY, Gong YC, Li QQ, Tan YW, Fan YY, Lin LZ, Zhou Y, Liu RQ, Hu LW, Yang BY, Zeng XW, Yu Y, Dong GH. Environmentally relevant concentrations of F-53B induce eye development disorders-mediated locomotor behavior in zebrafish larvae. CHEMOSPHERE 2022; 308:136130. [PMID: 36049635 DOI: 10.1016/j.chemosphere.2022.136130] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/19/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
The perfluorooctane sulfonate alternative, F-53B, induces multiple physiological defects but whether it can disrupt eye development is unknown. We exposed zebrafish to F-53B at four different concentrations (0, 0.15, 1.5, and 15 μg/L) for 120 h post-fertilization (hpf). Locomotor behavior, neurotransmitters content, histopathological alterations, morphological changes, cell apoptosis, and retinoic acid signaling were studied. Histology and morphological analyses showed that F-53B induced pathological changes in lens and retina of larvae and eye size were significantly reduced as compared to control. Acridine orange (AO) staining revealed a dose-dependent increase in early apoptosis, accompanied by upregulation of p53, casp-9 and casp-3 genes. Genes related to retinoic acid signaling (aldh1a2), lens developmental (cryaa, crybb, crygn, and mipa) and retinal development (pax6, rx1, gant1, rho, opn1sw and opn1lw) were significantly downregulated. In addition, behavioral responses (swimming speed) were significantly increased, while no significant changes in the neurotransmitters (dopamine and acetylcholine) level were observed. Therefore, in this study we observed that exposure to F-53B inflicted histological and morphological changes in zebrafish larvae eye, induced visual motor dysfunctions, perturbed retinoid signaling and retinal development and ultimately triggering apoptosis.
Collapse
Affiliation(s)
- Luyin Wu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mohammed Zeeshan
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yao Dang
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou, 510655, China
| | - Li-Ya Liang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yan-Chen Gong
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing-Qing Li
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ya-Wen Tan
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuan-Yuan Fan
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Zi Lin
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yang Zhou
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou, 510655, China
| | - Ru-Qing Liu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Wen Hu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Bo-Yi Yang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao-Wen Zeng
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yunjiang Yu
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou, 510655, China.
| | - Guang-Hui Dong
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
15
|
Gao G, Guo S, Zhang Q, Zhang H, Zhang C, Peng G. Kiaa1024L/Minar2 is essential for hearing by regulating cholesterol distribution in hair bundles. eLife 2022; 11:e80865. [PMID: 36317962 PMCID: PMC9714970 DOI: 10.7554/elife.80865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/31/2022] [Indexed: 12/05/2022] Open
Abstract
Unbiased genetic screens implicated a number of uncharacterized genes in hearing loss, suggesting some biological processes required for auditory function remain unexplored. Loss of Kiaa1024L/Minar2, a previously understudied gene, caused deafness in mice, but how it functioned in the hearing was unclear. Here, we show that disruption of kiaa1024L/minar2 causes hearing loss in the zebrafish. Defects in mechanotransduction, longer and thinner hair bundles, and enlarged apical lysosomes in hair cells are observed in the kiaa1024L/minar2 mutant. In cultured cells, Kiaa1024L/Minar2 is mainly localized to lysosomes, and its overexpression recruits cholesterol and increases cholesterol labeling. Strikingly, cholesterol is highly enriched in the hair bundle membrane, and loss of kiaa1024L/minar2 reduces cholesterol localization to the hair bundles. Lowering cholesterol levels aggravates, while increasing cholesterol levels rescues the hair cell defects in the kiaa1024L/minar2 mutant. Therefore, cholesterol plays an essential role in hair bundles, and Kiaa1024L/Minar2 regulates cholesterol distribution and homeostasis to ensure normal hearing.
Collapse
Affiliation(s)
- Ge Gao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Shuyu Guo
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Quan Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Hefei Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Cuizhen Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Gang Peng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| |
Collapse
|
16
|
Cheng Y, Cao P, Geng C, Chu X, Li Y, Cui J. The adenosine A (2A) receptor antagonist SCH58261 protects photoreceptors by inhibiting microglial activation and the inflammatory response. Int Immunopharmacol 2022; 112:109245. [PMID: 36150227 DOI: 10.1016/j.intimp.2022.109245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/18/2022] [Accepted: 09/09/2022] [Indexed: 11/05/2022]
Abstract
Photoreceptor degeneration is a principal event in a variety of human retinal diseases. Progressive apoptosis of photoreceptors leads to impaired vision and blindness, for which there is no curative treatment. Adenosine 2A receptors (A2AR) are expressed in microglia. Blockade of A2AR has been shown to protect neurons via suppression of inflammation. However, the therapeutic effects of A2AR antagonists on photoreceptor degeneration have not been characterized. In this study, adult zebrafish were exposed to short term high-intensity light to induce photoreceptor death. SCH58261, a selective A2AR antagonist, was immediately injected into the vitreous body. Photoreceptor degeneration and microglia-induced inflammation were evaluated using immunohistochemistry, quantitative real-time polymerase chain reaction, polarization sensitive optical coherence tomography, and optomotor response. Co-culture of BV2 and 661W cells was used to investigate the interaction between microglia and photoreceptors. The results showed that A2AR was over-expressed during photoreceptor degeneration. Following intraocular SCH58261 injection, microglial activation and release of inflammatory factors were inhibited, and photoreceptor survival increased. Inactivation of microglia prevented apoptosis and autophagy in photoreceptors. Our results showed that SCH58261 intervention at the early stage of photoreceptor degeneration protected photoreceptors through inhibition of the inflammatory response, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Yajia Cheng
- Medical International Collaborative Innovation Center, Nankai University School of Medicine, Tianjin, China; Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Peipei Cao
- Medical International Collaborative Innovation Center, Nankai University School of Medicine, Tianjin, China
| | - Chao Geng
- Medical International Collaborative Innovation Center, Nankai University School of Medicine, Tianjin, China
| | - Xiaoqi Chu
- Medical International Collaborative Innovation Center, Nankai University School of Medicine, Tianjin, China
| | - Yuhao Li
- Beijing Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Jianlin Cui
- Medical International Collaborative Innovation Center, Nankai University School of Medicine, Tianjin, China.
| |
Collapse
|
17
|
Sabharwal A, Campbell JM, Schwab TL, WareJoncas Z, Wishman MD, Ata H, Liu W, Ichino N, Hunter DE, Bergren JD, Urban MD, Urban RM, Holmberg SR, Kar B, Cook A, Ding Y, Xu X, Clark KJ, Ekker SC. A Primer Genetic Toolkit for Exploring Mitochondrial Biology and Disease Using Zebrafish. Genes (Basel) 2022; 13:1317. [PMID: 35893052 PMCID: PMC9331066 DOI: 10.3390/genes13081317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
Mitochondria are a dynamic eukaryotic innovation that play diverse roles in biology and disease. The mitochondrial genome is remarkably conserved in all vertebrates, encoding the same 37-gene set and overall genomic structure, ranging from 16,596 base pairs (bp) in the teleost zebrafish (Danio rerio) to 16,569 bp in humans. Mitochondrial disorders are amongst the most prevalent inherited diseases, affecting roughly 1 in every 5000 individuals. Currently, few effective treatments exist for those with mitochondrial ailments, representing a major unmet patient need. Mitochondrial dysfunction is also a common component of a wide variety of other human illnesses, ranging from neurodegenerative disorders such as Huntington's disease and Parkinson's disease to autoimmune illnesses such as multiple sclerosis and rheumatoid arthritis. The electron transport chain (ETC) component of mitochondria is critical for mitochondrial biology and defects can lead to many mitochondrial disease symptoms. Here, we present a publicly available collection of genetic mutants created in highly conserved, nuclear-encoded mitochondrial genes in Danio rerio. The zebrafish system represents a potentially powerful new opportunity for the study of mitochondrial biology and disease due to the large number of orthologous genes shared with humans and the many advanced features of this model system, from genetics to imaging. This collection includes 15 mutant lines in 13 different genes created through locus-specific gene editing to induce frameshift or splice acceptor mutations, leading to predicted protein truncation during translation. Additionally, included are 11 lines created by the random insertion of the gene-breaking transposon (GBT) protein trap cassette. All these targeted mutant alleles truncate conserved domains of genes critical to the proper function of the ETC or genes that have been implicated in human mitochondrial disease. This collection is designed to accelerate the use of zebrafish to study many different aspects of mitochondrial function to widen our understanding of their role in biology and human disease.
Collapse
Affiliation(s)
- Ankit Sabharwal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Jarryd M. Campbell
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Tanya L. Schwab
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Zachary WareJoncas
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Mark D. Wishman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Hirotaka Ata
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Wiebin Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
- Division of Cardiovascular Diseases, Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Noriko Ichino
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Danielle E. Hunter
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Jake D. Bergren
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Mark D. Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Rhianna M. Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Shannon R. Holmberg
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Bibekananda Kar
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Alex Cook
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
- Division of Cardiovascular Diseases, Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
- Division of Cardiovascular Diseases, Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Karl J. Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| | - Stephen C. Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; (A.S.); (J.M.C.); (T.L.S.); (Z.W.); (M.D.W.); (H.A.); (W.L.); (N.I.); (D.E.H.); (J.D.B.); (M.D.U.); (R.M.U.); (S.R.H.); (B.K.); (A.C.); (Y.D.); (X.X.); (K.J.C.)
| |
Collapse
|
18
|
Tang D, Zheng S, Zheng Z, Liu C, Zhang J, Yan R, Wu C, Zuo N, Wu L, Xu H, Liu S, He Y. Dnmt1 is required for the development of auditory organs via cell cycle arrest and Fgf signalling. Cell Prolif 2022; 55:e13225. [PMID: 35352419 PMCID: PMC9136517 DOI: 10.1111/cpr.13225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 11/30/2022] Open
Abstract
Objectives To explore the role of DNA methyltransferase 1 (DNMT1) in the development of auditory system using zebrafish as experimental model. Methods Morpholino oligonucleotide was used to induce Dnmt1 deficiency. RNA sequencing, in situ hybridization (ISH), whole genomic bisulfide sequencing (WGBS) and immunostaining were used to investigate the morphologic alterations and mechanisms. Results We found that downregulation of Dnmt1 induced decreased number of neuromasts and repressed cell proliferation of primordium in the developing posterior lateral line system of zebrafish. The ISH data uncovered that Fgf signalling pathway was inhibited and the expression of chemokine members cxcr4b, cxcr7b and cxcl12a were interfered, while lef1 expression was increased after inhibiting Dnmt1. Additionally, Dnmt1 downregulation led to malformed otoliths and deformed semicircular canals, and hair cell differentiation in utricle and saccule was inhibited severely. The in situ staining of otic placode markers pax2/5 and fgf 3/8/10 was decreased when Dnmt1 downregulated. The WGBS analysis demonstrated that the global methylation status was markedly downregulated, and cell cycle genes were among those most differently expressed between Dnmt1 morphants and the controls. Further ISH analysis confirmed the findings by RNA‐seq and WGBS assay that cdkn1a and tp53 were both upregulated after knockdown of Dnmt1. Conclusion Our results revealed that Dnmt1 is essential for the development of zebrafish auditory organ through regulating cell cycle genes together with Wnt and Fgf signalling pathways.
Collapse
Affiliation(s)
- Dongmei Tang
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, China
| | - Shimei Zheng
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Zhiwei Zheng
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, China
| | - Chang Liu
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, China
| | - Jiner Zhang
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Renchun Yan
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Cheng Wu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Na Zuo
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Lijuan Wu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Hongfei Xu
- Department of Forensic Medicine, Soochow University, Suzhou, China
| | - Shaofeng Liu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Yingzi He
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Zheng S, Tang D, Wang X, Liu C, Zuo N, Yan R, Wu C, Ma J, Wang C, Xu H, He Y, Liu D, Liu S. Kif15 Is Required in the Development of Auditory System Using Zebrafish as a Model. Front Mol Neurosci 2022; 15:844568. [PMID: 35370541 PMCID: PMC8971910 DOI: 10.3389/fnmol.2022.844568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/21/2022] [Indexed: 11/30/2022] Open
Abstract
Kif15, a kinesin family member, is powerful in the formation of bipolar spindles. There is emerging evidence indicating that Kif15 plays vital roles in influencing the growth of axons and interference with the progression of the tumor. However, the function of Kif15 in the auditory organs remains unknown. The Western blotting test was used to examine the effect of Kif15 downregulation by specific morpholino targeting Kif15 (Kif15-MO). The development of the inner ear and posterior lateral line (PLL) system in zebrafish was under continuous observation from spawns to 96 h postfertilization (hpf) to investigate the potential role of Kif15 in the auditory and vestibular system. We uncovered that Kif15 inhibition induced otic organ deformities in zebrafish, including malformed semicircular canals, abnormal number and location of otoliths, and reduced number of hair cells (HCs) both in utricle and saccule. Furthermore, a remarkable reduction in the number of PLL neuromasts was also explored in Kif15-MO morphants compared to the normal larvae. We also detected notably reduced activity in locomotion after Kif15 was knocked down. Additionally, we performed rescue experiments with co-injection of Kif15 mRNA and found that the Kif15 splicing MO-induced deformities in otic vesicle and PLL of zebrafish were successfully rescued, and the severely reduced locomotor activity caused by Kif15-MO was partially rescued compared to the control-MO (Con-MO) embryos. Our findings uncover that Kif15 is essential in the early development of auditory and vestibular organs using zebrafish as models.
Collapse
Affiliation(s)
- Shimei Zheng
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Dongmei Tang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Xin Wang
- Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and MOE, Nantong University, Nantong, China
| | - Chang Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Na Zuo
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Renchun Yan
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Cheng Wu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Jun Ma
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Chuanxi Wang
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Hongfei Xu
- Department of Forensic Medicine, Soochow University, Suzhou, China
| | - Yingzi He
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
- *Correspondence: Yingzi He,
| | - Dong Liu
- Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and MOE, Nantong University, Nantong, China
- Dong Liu, ,
| | - Shaofeng Liu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
- Shaofeng Liu,
| |
Collapse
|
20
|
Aragona M, Porcino C, Guerrera MC, Montalbano G, Laurà R, Cometa M, Levanti M, Abbate F, Cobo T, Capitelli G, Vega JA, Germanà A. The BDNF/TrkB Neurotrophin System in the Sensory Organs of Zebrafish. Int J Mol Sci 2022; 23:ijms23052621. [PMID: 35269763 PMCID: PMC8910639 DOI: 10.3390/ijms23052621] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022] Open
Abstract
The brain-derived neurotrophic factor (BDNF) was discovered in the last century, and identified as a member of the neurotrophin family. BDNF shares approximately 50% of its amino acid with other neurotrophins such as NGF, NT-3 and NT-4/5, and its linear amino acid sequences in zebrafish (Danio rerio) and human are 91% identical. BDNF functions can be mediated by two categories of receptors: p75NTR and Trk. Intriguingly, BDNF receptors were highly conserved in the process of evolution, as were the other NTs’ receptors. In this review, we update current knowledge about the distribution and functions of the BDNF-TrkB system in the sensory organs of zebrafish. In fish, particularly in zebrafish, the distribution and functions of BDNF and TrkB in the brain have been widely studied. Both components of the system, associated or segregated, are also present outside the central nervous system, especially in sensory organs including the inner ear, lateral line system, retina, taste buds and olfactory epithelium.
Collapse
Affiliation(s)
- Marialuisa Aragona
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (C.P.); (M.C.G.); (G.M.); (R.L.); (M.C.); (M.L.); (F.A.)
| | - Caterina Porcino
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (C.P.); (M.C.G.); (G.M.); (R.L.); (M.C.); (M.L.); (F.A.)
| | - Maria Cristina Guerrera
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (C.P.); (M.C.G.); (G.M.); (R.L.); (M.C.); (M.L.); (F.A.)
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (C.P.); (M.C.G.); (G.M.); (R.L.); (M.C.); (M.L.); (F.A.)
| | - Rosaria Laurà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (C.P.); (M.C.G.); (G.M.); (R.L.); (M.C.); (M.L.); (F.A.)
| | - Marzio Cometa
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (C.P.); (M.C.G.); (G.M.); (R.L.); (M.C.); (M.L.); (F.A.)
| | - Maria Levanti
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (C.P.); (M.C.G.); (G.M.); (R.L.); (M.C.); (M.L.); (F.A.)
| | - Francesco Abbate
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (C.P.); (M.C.G.); (G.M.); (R.L.); (M.C.); (M.L.); (F.A.)
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, 33006 Oviedo, Spain;
| | - Gabriel Capitelli
- Faculty of Medical Sciences, University of Buenos Aires, Viamonte 1053, CABA, Buenos Aires 1056, Argentina;
| | - José A. Vega
- Grupo SINPOS, Universidad de Oviedo, 33003 Oviedo, Spain;
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 7500912, Chile
| | - Antonino Germanà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (C.P.); (M.C.G.); (G.M.); (R.L.); (M.C.); (M.L.); (F.A.)
- Correspondence:
| |
Collapse
|
21
|
Hong Y, Luo Y. Zebrafish Model in Ophthalmology to Study Disease Mechanism and Drug Discovery. Pharmaceuticals (Basel) 2021; 14:ph14080716. [PMID: 34451814 PMCID: PMC8400593 DOI: 10.3390/ph14080716] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 12/14/2022] Open
Abstract
Visual impairment and blindness are common and seriously affect people’s work and quality of life in the world. Therefore, the effective therapies for eye diseases are of high priority. Zebrafish (Danio rerio) is an alternative vertebrate model as a useful tool for the mechanism elucidation and drug discovery of various eye disorders, such as cataracts, glaucoma, diabetic retinopathy, age-related macular degeneration, photoreceptor degeneration, etc. The genetic and embryonic accessibility of zebrafish in combination with a behavioral assessment of visual function has made it a very popular model in ophthalmology. Zebrafish has also been widely used in ocular drug discovery, such as the screening of new anti-angiogenic compounds or neuroprotective drugs, and the oculotoxicity test. In this review, we summarized the applications of zebrafish as the models of eye disorders to study disease mechanism and investigate novel drug treatments.
Collapse
Affiliation(s)
| | - Yan Luo
- Correspondence: ; Tel.: +86-020-87335931
| |
Collapse
|
22
|
Fuster-García C, García-Bohórquez B, Rodríguez-Muñoz A, Aller E, Jaijo T, Millán JM, García-García G. Usher Syndrome: Genetics of a Human Ciliopathy. Int J Mol Sci 2021; 22:6723. [PMID: 34201633 PMCID: PMC8268283 DOI: 10.3390/ijms22136723] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/21/2022] Open
Abstract
Usher syndrome (USH) is an autosomal recessive syndromic ciliopathy characterized by sensorineural hearing loss, retinitis pigmentosa and, sometimes, vestibular dysfunction. There are three clinical types depending on the severity and age of onset of the symptoms; in addition, ten genes are reported to be causative of USH, and six more related to the disease. These genes encode proteins of a diverse nature, which interact and form a dynamic protein network called the "Usher interactome". In the organ of Corti, the USH proteins are essential for the correct development and maintenance of the structure and cohesion of the stereocilia. In the retina, the USH protein network is principally located in the periciliary region of the photoreceptors, and plays an important role in the maintenance of the periciliary structure and the trafficking of molecules between the inner and the outer segments of photoreceptors. Even though some genes are clearly involved in the syndrome, others are controversial. Moreover, expression of some USH genes has been detected in other tissues, which could explain their involvement in additional mild comorbidities. In this paper, we review the genetics of Usher syndrome and the spectrum of mutations in USH genes. The aim is to identify possible mutation associations with the disease and provide an updated genotype-phenotype correlation.
Collapse
Affiliation(s)
- Carla Fuster-García
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Belén García-Bohórquez
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
| | - Ana Rodríguez-Muñoz
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
| | - Elena Aller
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Teresa Jaijo
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - José M. Millán
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Gema García-García
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| |
Collapse
|
23
|
Nicolson T. Navigating Hereditary Hearing Loss: Pathology of the Inner Ear. Front Cell Neurosci 2021; 15:660812. [PMID: 34093131 PMCID: PMC8172992 DOI: 10.3389/fncel.2021.660812] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Inherited forms of deafness account for a sizable portion of hearing loss among children and adult populations. Many patients with sensorineural deficits have pathological manifestations in the peripheral auditory system, the inner ear. Within the hearing organ, the cochlea, most of the genetic forms of hearing loss involve defects in sensory detection and to some extent, signaling to the brain via the auditory cranial nerve. This review focuses on peripheral forms of hereditary hearing loss and how these impairments can be studied in diverse animal models or patient-derived cells with the ultimate goal of using the knowledge gained to understand the underlying biology and treat hearing loss.
Collapse
Affiliation(s)
- Teresa Nicolson
- Department of Otolaryngology, Stanford University, Stanford, CA, United States
| |
Collapse
|
24
|
Baldridge D, Wangler MF, Bowman AN, Yamamoto S, Schedl T, Pak SC, Postlethwait JH, Shin J, Solnica-Krezel L, Bellen HJ, Westerfield M. Model organisms contribute to diagnosis and discovery in the undiagnosed diseases network: current state and a future vision. Orphanet J Rare Dis 2021; 16:206. [PMID: 33962631 PMCID: PMC8103593 DOI: 10.1186/s13023-021-01839-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
Decreased sequencing costs have led to an explosion of genetic and genomic data. These data have revealed thousands of candidate human disease variants. Establishing which variants cause phenotypes and diseases, however, has remained challenging. Significant progress has been made, including advances by the National Institutes of Health (NIH)-funded Undiagnosed Diseases Network (UDN). However, 6000-13,000 additional disease genes remain to be identified. The continued discovery of rare diseases and their genetic underpinnings provides benefits to affected patients, of whom there are more than 400 million worldwide, and also advances understanding the mechanisms of more common diseases. Platforms employing model organisms enable discovery of novel gene-disease relationships, help establish variant pathogenicity, and often lead to the exploration of underlying mechanisms of pathophysiology that suggest new therapies. The Model Organism Screening Center (MOSC) of the UDN is a unique resource dedicated to utilizing informatics and functional studies in model organisms, including worm (Caenorhabditis elegans), fly (Drosophila melanogaster), and zebrafish (Danio rerio), to aid in diagnosis. The MOSC has directly contributed to the diagnosis of challenging cases, including multiple patients with complex, multi-organ phenotypes. In addition, the MOSC provides a framework for how basic scientists and clinicians can collaborate to drive diagnoses. Customized experimental plans take into account patient presentations, specific genes and variant(s), and appropriateness of each model organism for analysis. The MOSC also generates bioinformatic and experimental tools and reagents for the wider scientific community. Two elements of the MOSC that have been instrumental in its success are (1) multidisciplinary teams with expertise in variant bioinformatics and in human and model organism genetics, and (2) mechanisms for ongoing communication with clinical teams. Here we provide a position statement regarding the central role of model organisms for continued discovery of disease genes, and we advocate for the continuation and expansion of MOSC-type research entities as a Model Organisms Network (MON) to be funded through grant applications submitted to the NIH, family groups focused on specific rare diseases, other philanthropic organizations, industry partnerships, and other sources of support.
Collapse
Affiliation(s)
- Dustin Baldridge
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, 77030, USA.
- Department of Pediatrics, BCM, Houston, TX, 77030, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA.
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX, 77030, USA.
| | - Angela N Bowman
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX, 77030, USA
- Department of Neuroscience, BCM, Houston, TX, 77030, USA
| | - Tim Schedl
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Stephen C Pak
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Jimann Shin
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX, 77030, USA
- Department of Neuroscience, BCM, Houston, TX, 77030, USA
- Howard Hughes Medical Institute, Houston, TX, 77030, USA
| | - Monte Westerfield
- Institute of Neuroscience, University of Oregon, Eugene, OR, 97403, USA
| |
Collapse
|
25
|
Parkinson L, Stawicki TM. alms1 mutant zebrafish do not show hair cell phenotypes seen in other cilia mutants. PLoS One 2021; 16:e0246844. [PMID: 33793549 PMCID: PMC8016283 DOI: 10.1371/journal.pone.0246844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/26/2021] [Indexed: 11/18/2022] Open
Abstract
Multiple cilia-associated genes have been shown to affect hair cells in zebrafish (Danio rerio), including the human deafness gene dcdc2, the radial spoke gene rsph9, and multiple intraflagellar transport (IFT) and transition zone genes. Recently a zebrafish alms1 mutant was generated. The ALMS1 gene is the gene mutated in the ciliopathy Alström Syndrome a disease that causes hearing loss among other symptoms. The hearing loss seen in Alström Syndrome may be due in part to hair cell defects as Alms1 mutant mice show stereocilia polarity defects and a loss of hair cells. Hair cell loss is also seen in postmortem analysis of Alström patients. The zebrafish alms1 mutant has metabolic defects similar to those seen in Alström syndrome and Alms1 mutant mice. We wished to investigate if it also had hair cell defects. We, however, failed to find any hair cell related phenotypes in alms1 mutant zebrafish. They had normal lateral line hair cell numbers as both larvae and adults and normal kinocilia formation. They also showed grossly normal swimming behavior, response to vibrational stimuli, and FM1-43 loading. Mutants also showed a normal degree of sensitivity to both short-term neomycin and long-term gentamicin treatment. These results indicate that cilia-associated genes differentially affect different hair cell types.
Collapse
Affiliation(s)
- Lauren Parkinson
- Neuroscience Program, Lafayette College, Easton, Pennsylvania, United States of America
| | - Tamara M. Stawicki
- Neuroscience Program, Lafayette College, Easton, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
26
|
Li M, Yang T, Gao L, Xu H. An inadvertent issue of human retina exposure to endocrine disrupting chemicals: A safety assessment. CHEMOSPHERE 2021; 264:128484. [PMID: 33022499 DOI: 10.1016/j.chemosphere.2020.128484] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/07/2020] [Accepted: 09/27/2020] [Indexed: 06/11/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are a group of chemical compounds that present a considerable public health problem due to their pervasiveness and associations with chronic diseases. EDCs can interrupt the endocrine system and interfere with hormone homeostasis, leading to abnormalities in human physiology. Much attention has been focused on the adverse effects EDCs have on the reproductive system, neurogenesis, neuroendocrine system, and thyroid dysfunction. The eye is usually directly exposed to the surrounding environment; however, the influences of EDCs on the eye have received comparatively little attention. Ocular diseases, such as ocular surface diseases and retinal diseases, have been implicated in hormone deficiency or excess. Epidemiologic studies have shown that EDC exposure not only causes ocular surface disorders, such as dry eye, but also associates with visual deficits and retinopathy. EDCs can pass through the human blood-retinal barrier and enter the neural retina, and can then accumulate in the retina. The retina is an embryologic extension of the central nervous system, and is extremely sensitive and vulnerable to EDCs that could be passed across the placenta during critical periods of retinal development. Subtle alterations in the retinal development process usually result in profound immediate, long-term, and delayed effects late in life. This review, based on extensive literature survey, briefly summarizes the current knowledge about the impact of representative manufactured EDCs on retinal toxicity, including retinal structure alterations and dysfunction. We also highlight the potential mechanism of action of EDCs on the retina, and the predictive retinal models of EDC exposure.
Collapse
Affiliation(s)
- Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Tian Yang
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of PLA General Hospital, Beijing, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China.
| |
Collapse
|
27
|
Vona B, Doll J, Hofrichter MAH, Haaf T, Varshney GK. Small fish, big prospects: using zebrafish to unravel the mechanisms of hereditary hearing loss. Hear Res 2020; 397:107906. [PMID: 32063424 PMCID: PMC7415493 DOI: 10.1016/j.heares.2020.107906] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/13/2020] [Accepted: 01/29/2020] [Indexed: 12/16/2022]
Abstract
Over the past decade, advancements in high-throughput sequencing have greatly enhanced our knowledge of the mutational signatures responsible for hereditary hearing loss. In its present state, the field has a largely uncensored view of protein coding changes in a growing number of genes that have been associated with hereditary hearing loss, and many more that have been proposed as candidate genes. Sequencing data can now be generated using methods that have become widespread and affordable. The greatest hurdles facing the field concern functional validation of uncharacterized genes and rapid application to human diseases, including hearing and balance disorders. To date, over 30 hearing-related disease models exist in zebrafish. New genome editing technologies, including CRISPR/Cas9 will accelerate the functional validation of hearing loss genes and variants in zebrafish. Here, we discuss current progress in the field and recent advances in genome editing approaches.
Collapse
Affiliation(s)
- Barbara Vona
- Department of Otolaryngology--Head & Neck Surgery, Tübingen Hearing Research Centre, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Julia Doll
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | | | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | - Gaurav K Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States.
| |
Collapse
|
28
|
Cheng X, Zhang JJ, Shi DL. Loss of Rbm24a causes defective hair cell development in the zebrafish inner ear and neuromasts. J Genet Genomics 2020; 47:403-406. [PMID: 33036919 DOI: 10.1016/j.jgg.2020.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/29/2020] [Accepted: 07/23/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Xiaoning Cheng
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jing-Jing Zhang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, 524001, China.
| | - De-Li Shi
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, 524001, China; Develompental Biology Laboratory, CNRS UMR7622, Institut de Biologie Paris-Seine, Sorbonne University, 75005, Paris, France.
| |
Collapse
|
29
|
Germanà A, Guerrera MC, Laurà R, Levanti M, Aragona M, Mhalhel K, Germanà G, Montalbano G, Abbate F. Expression and Localization of BDNF/TrkB System in the Zebrafish Inner Ear. Int J Mol Sci 2020; 21:ijms21165787. [PMID: 32806650 PMCID: PMC7460859 DOI: 10.3390/ijms21165787] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, is involved in multiple and fundamental functions of the central and peripheral nervous systems including sensory organs. Despite recent advances in knowledge on the functional significance of BDNF and TrkB in the regulation of the acoustic system of mammals, the localization of BDNF/TrkB system in the inner ear of zebrafish during development, is not well known. Therefore, the goal of the present study is to analyze the age-dependent changes using RT-PCR, Western Blot and single and double immunofluorescence of the BDNF and its specific receptor in the zebrafish inner ear. The results showed the mRNA expression and the cell localization of BDNF and TrkB in the hair cells of the crista ampullaris and in the neuroepithelium of the utricle, saccule and macula lagena, analyzed at different ages. Our results demonstrate that the BDNF/TrkB system is present in the sensory cells of the inner ear, during whole life. Therefore, this system might play a key role in the development and maintenance of the hair cells in adults, suggesting that the zebrafish inner ear represents an interesting model to study the involvement of the neurotrophins in the biology of sensory cells
Collapse
|
30
|
Grifone R, Shao M, Saquet A, Shi DL. RNA-Binding Protein Rbm24 as a Multifaceted Post-Transcriptional Regulator of Embryonic Lineage Differentiation and Cellular Homeostasis. Cells 2020; 9:E1891. [PMID: 32806768 PMCID: PMC7463526 DOI: 10.3390/cells9081891] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
RNA-binding proteins control the metabolism of RNAs at all stages of their lifetime. They are critically required for the post-transcriptional regulation of gene expression in a wide variety of physiological and pathological processes. Rbm24 is a highly conserved RNA-binding protein that displays strongly regionalized expression patterns and exhibits dynamic changes in subcellular localization during early development. There is increasing evidence that it acts as a multifunctional regulator to switch cell fate determination and to maintain tissue homeostasis. Dysfunction of Rbm24 disrupts cell differentiation in nearly every tissue where it is expressed, such as skeletal and cardiac muscles, and different head sensory organs, but the molecular events that are affected may vary in a tissue-specific, or even a stage-specific manner. Recent works using different animal models have uncovered multiple post-transcriptional regulatory mechanisms by which Rbm24 functions in key developmental processes. In particular, it represents a major splicing factor in muscle cell development, and plays an essential role in cytoplasmic polyadenylation during lens fiber cell terminal differentiation. Here we review the advances in understanding the implication of Rbm24 during development and disease, by focusing on its regulatory roles in physiological and pathological conditions.
Collapse
Affiliation(s)
- Raphaëlle Grifone
- Developmental Biology Laboratory, CNRS-UMR7622, IBPS, Sorbonne University, 75005 Paris, France; (R.G.); (A.S.)
| | - Ming Shao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao 266237, China;
| | - Audrey Saquet
- Developmental Biology Laboratory, CNRS-UMR7622, IBPS, Sorbonne University, 75005 Paris, France; (R.G.); (A.S.)
| | - De-Li Shi
- Developmental Biology Laboratory, CNRS-UMR7622, IBPS, Sorbonne University, 75005 Paris, France; (R.G.); (A.S.)
| |
Collapse
|
31
|
Wang Y, Liu W, Yuan B, Yin X, Li Y, Li Z, Cui J, Yuan X, Li Y. The Application of Methylprednisolone Nanoscale Zirconium-Porphyrin Metal-Organic Framework (MPS-NPMOF) in the Treatment of Photoreceptor Degeneration. Int J Nanomedicine 2019; 14:9763-9776. [PMID: 31849467 PMCID: PMC6911332 DOI: 10.2147/ijn.s225992] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/13/2019] [Indexed: 12/23/2022] Open
Abstract
Background Photoreceptor degeneration is one of the most refractory oculopathy in the world, leading to vision loss in severe cases. Methyprednisolone is one of the most commonly prescribed medications for the treatment of retinal degenerative diseases, either by oral administration or repeated intraocular injections. However, the efficacy was unsatisfactory due to its systemic or local side effects and short retention time within the retina. Methods Nanoscale zirconium-porphyrin metal-organic framework (NPMOF) was synthesized and characterized. The biotoxicity and imaging capability of NPMOF were evaluated using zebrafish embryos and larvae. NPMOF was then used as a skeleton and loaded with methylprednisolone (MPS) to prepare a novel kind of nanoparticle, MPS-NPMOF. Photoreceptor degeneration was induced by high-intensity light exposure in adult zebrafish. MPS-NPMOF was delivered to the injured retina by intraocular injection. The photoreceptor regeneration and its underlying mechanism were explored by immunohistochemistry, quantitative real-time polymerase chain reaction and behavioral test. Results NPMOF not only had low biotoxicity but also emitted bright fluorescence. Following a single MPS-NPMOF intraocular injection, the injured retina exhibited the faster photoreceptor regeneration with better visual function by promoting the cell proliferation. Conclusion NPMOF is an ideal carrier and could be applied in tracking and delivering medications. By intraocular injection, the novel drug delivery system, MPS-NPMOF, accomplishes the sustained release of drug and plays a therapeutic role in photoreceptor degeneration.
Collapse
Affiliation(s)
- Yajie Wang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine, Tianjin 300071, People's Republic of China.,Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Hospital, Tianjin 300020, People's Republic of China
| | - Wei Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Nankai University College of Chemistry, Tianjin 300071, People's Republic of China.,Tianjin University School of Science, Tianjin 300072, People's Republic of China
| | - Bo Yuan
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine, Tianjin 300071, People's Republic of China.,Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Hospital, Tianjin 300020, People's Republic of China
| | - Xuebo Yin
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Nankai University College of Chemistry, Tianjin 300071, People's Republic of China
| | - Yiming Li
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine, Tianjin 300071, People's Republic of China
| | - Zongjin Li
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine, Tianjin 300071, People's Republic of China
| | - Jianlin Cui
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine, Tianjin 300071, People's Republic of China
| | - Xiaoyong Yuan
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Hospital, Tianjin 300020, People's Republic of China
| | - Yuhao Li
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine, Tianjin 300071, People's Republic of China.,Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, People's Republic of China
| |
Collapse
|
32
|
Cavodeassi F, Wilson SW. Looking to the future of zebrafish as a model to understand the genetic basis of eye disease. Hum Genet 2019; 138:993-1000. [PMID: 31422478 PMCID: PMC6710215 DOI: 10.1007/s00439-019-02055-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
Abstract
In this brief commentary, we provide some of our thoughts and opinions on the current and future use of zebrafish to model human eye disease, dissect pathological progression and advance in our understanding of the genetic bases of microphthalmia, andophthalmia and coloboma (MAC) in humans. We provide some background on eye formation in fish and conservation and divergence across vertebrates in this process, discuss different approaches for manipulating gene function and speculate on future research areas where we think research using fish may prove to be particularly effective.
Collapse
Affiliation(s)
- Florencia Cavodeassi
- Institute of Medical and Biomedical Education, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, UK.
| | - Stephen W Wilson
- Department of Cell and Developmental Biology, Biosciences, UCL, Gower St, London, WC1E 6BT, UK
| |
Collapse
|
33
|
Unconventional secretory pathway activation restores hair cell mechanotransduction in an USH3A model. Proc Natl Acad Sci U S A 2019; 116:11000-11009. [PMID: 31097578 DOI: 10.1073/pnas.1817500116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pathogenic variant c.144T>G (p.N48K) in the clarin1 gene (CLRN1) results in progressive loss of vision and hearing in Usher syndrome IIIA (USH3A) patients. CLRN1 is predicted to be an essential protein in hair bundles, the mechanosensory structure of hair cells critical for hearing and balance. When expressed in animal models, CLRN1 localizes to the hair bundle, whereas glycosylation-deficient CLRN1N48K aggregates in the endoplasmic reticulum, with only a fraction reaching the bundle. We hypothesized that the small amount of CLRN1N48K that reaches the hair bundle does so via an unconventional secretory pathway and that activation of this pathway could be therapeutic. Using genetic and pharmacological approaches, we find that clarin1 knockout (clrn1 KO/KO ) zebrafish that express the CLRN1 c.144T>G pathogenic variant display progressive hair cell dysfunction, and that CLRN1N48K is trafficked to the hair bundle via the GRASP55 cargo-dependent unconventional secretory pathway (GCUSP). On expression of GRASP55 mRNA, or on exposure to the drug artemisinin (which activates GCUSP), the localization of CLRN1N48K to the hair bundles was enhanced. Artemisinin treatment also effectively restored hair cell mechanotransduction and attenuated progressive hair cell dysfunction in clrn1 KO/KO larvae that express CLRN1 c.144T>G , highlighting the potential of artemisinin to prevent sensory loss in CLRN1 c.144T>G patients.
Collapse
|
34
|
Kujawski S, Sonawane M, Knust E. penner/lgl2 is required for the integrity of the photoreceptor layer in the zebrafish retina. Biol Open 2019; 8:8/4/bio041830. [PMID: 31015218 PMCID: PMC6503998 DOI: 10.1242/bio.041830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The vertebrate retina is a complex tissue built from multiple neuronal cell types, which develop from a pseudostratified neuroepithelium. These cells are arranged into a highly organized and stereotypic pattern formed by nuclear and plexiform layers. The process of lamination as well as the maturation and differentiation of photoreceptor cells rely on the establishment and maintenance of apico-basal cell polarity and formation of adhesive junctions. Defects in any of these processes can result in impaired vision and are causally related to a variety of human diseases leading to blindness. While the importance of apical polarity regulators in retinal stratification and disease is well established, little is known about the function of basal regulators in retinal development. Here, we analyzed the role of Lgl2, a basolateral polarity factor, in the zebrafish retina. Lgl2 is upregulated in photoreceptor cells and in the retinal pigment epithelium by 72 h post fertilization. In both cell types, Lgl2 is localized basolaterally. Loss of zygotic Lgl2 does not interfere with retinal lamination or photoreceptor cell polarity or maturation. However, knockdown of both maternal and zygotic Lgl2 leads to impaired cell adhesion. As a consequence, severe layering defects occur in the distal retina, manifested by a breakdown of the outer plexiform layer and the outer limiting membrane. These results define zebrafish Lgl2 as an important regulator of retinal lamination, which, given the high degree of evolutionary conservation, may be preserved in other vertebrates, including human. Summary: Knockdown of penner/lgl2 leads to a breakdown of the outer plexiform layer and the outer limiting membrane in the zebrafish retina due to impaired cell adhesion.
Collapse
Affiliation(s)
- Satu Kujawski
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108 01307 Dresden, Germany
| | - Mahendra Sonawane
- Tata Institute of Fundamental Research, Department of Biological Sciences, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai 400005, India
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108 01307 Dresden, Germany
| |
Collapse
|
35
|
Baumann L, Segner H, Ros A, Knapen D, Vergauwen L. Thyroid Hormone Disruptors Interfere with Molecular Pathways of Eye Development and Function in Zebrafish. Int J Mol Sci 2019; 20:E1543. [PMID: 30934780 PMCID: PMC6479403 DOI: 10.3390/ijms20071543] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/12/2019] [Accepted: 03/25/2019] [Indexed: 02/04/2023] Open
Abstract
The effects of thyroid hormone disrupting chemicals (THDCs) on eye development of zebrafish were investigated. We expected THDC exposure to cause transcriptional changes of vision-related genes, which find their phenotypic anchoring in eye malformations and dysfunction, as observed in our previous studies. Zebrafish were exposed from 0 to 5 days post fertilization (dpf) to either propylthiouracil (PTU), a thyroid hormone synthesis inhibitor, or tetrabromobisphenol-A (TBBPA), which interacts with thyroid hormone receptors. Full genome microarray analyses of RNA isolated from eye tissue revealed that the number of affected transcripts was substantially higher in PTU- than in TBBPA-treated larvae. However, multiple components of phototransduction (e.g., phosphodiesterase, opsins) were responsive to both THDC exposures. Yet, the response pattern for the gene ontology (GO)-class "sensory perception" differed between treatments, with over 90% down-regulation in PTU-exposed fish, compared to over 80% up-regulation in TBBPA-exposed fish. Additionally, the reversibility of effects after recovery in clean water for three days was investigated. Transcriptional patterns in the eyes were still altered and partly overlapped between 5 and 8 dpf, showing that no full recovery occurred within the time period investigated. However, pathways involved in repair mechanisms were significantly upregulated, which indicates activation of regeneration processes.
Collapse
Affiliation(s)
- Lisa Baumann
- Centre for Organismal Studies, Aquatic Ecology and Toxicology, University of Heidelberg, Im Neuenheimer Feld 504, 69120 Heidelberg, Germany.
| | - Helmut Segner
- Vetsuisse Faculty, Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland.
| | - Albert Ros
- Fischereiforschungsstelle LAZBW, Argenweg 50/1, 88085 Langenargen, Germany.
| | - Dries Knapen
- Department of Veterinary Sciences, Veterinary Physiology and Biochemistry, Zebrafishlab, University of Antwerp, Universiteitsplein 1, 2160 Wilrijk, Belgium.
| | - Lucia Vergauwen
- Department of Veterinary Sciences, Veterinary Physiology and Biochemistry, Zebrafishlab, University of Antwerp, Universiteitsplein 1, 2160 Wilrijk, Belgium.
- Department of Biology, Systemic Physiological and Ecotoxicological Research (SPHERE), University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium.
| |
Collapse
|
36
|
Singh A, Castillo HA, Brown J, Kaslin J, Dwyer KM, Gibert Y. High glucose levels affect retinal patterning during zebrafish embryogenesis. Sci Rep 2019; 9:4121. [PMID: 30858575 PMCID: PMC6411978 DOI: 10.1038/s41598-019-41009-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 02/12/2019] [Indexed: 02/08/2023] Open
Abstract
Maternal hyperglycaemia has a profound impact on the developing foetus and increases the risk of developing abnormalities like obesity, impaired glucose tolerance and insulin secretory defects in the post-natal life. Increased levels of glucose in the blood stream due to diabetes causes visual disorders like retinopathy. However, the impact of maternal hyperglycaemia due to pre-existing or gestational diabetes on the developing foetal retina is unknown. The aim of this work was to study the effect of hyperglycaemia on the developing retina using zebrafish as a vertebrate model. Wild-type and transgenic zebrafish embryos were exposed to 0, 4 and 5% D-Glucose in a pulsatile manner to mimic the fluctuations in glycaemia experienced by the developing foetus in pregnant women with diabetes. The zebrafish embryos displayed numerous ocular defects associated with altered retinal cell layer thickness, increased presence of macrophages, and decreased number of Müeller glial and retinal ganglion cells following high-glucose exposure. We have developed a model of gestational hyperglycaemia using the zebrafish embryo to study the effect of hyperglycaemia on the developing embryonic retina. The data suggests that glucose exposure is detrimental to the development of embryonic retina and the legacy of this exposure may extend into adulthood. These data suggest merit in retinal assessment in infants born to mothers with pre-existing and gestational diabetes both in early and adult life.
Collapse
Affiliation(s)
- Amitoj Singh
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Road, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Hozana Andrade Castillo
- Monash University, Australian Regenerative Medicine Institute, 23 Innovation Walk, Clayton, VIC, 3800, Australia
- Brazilian Biosciences National Laboratory, Brazilian Centre for Research in Energy and Materials, Campinas, Brazil
| | - Julie Brown
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Road, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Jan Kaslin
- Monash University, Australian Regenerative Medicine Institute, 23 Innovation Walk, Clayton, VIC, 3800, Australia
| | - Karen M Dwyer
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Road, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Yann Gibert
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Road, Waurn Ponds, Geelong, VIC, 3216, Australia.
| |
Collapse
|
37
|
Naert T, Vleminckx K. CRISPR/Cas9 disease models in zebrafish and Xenopus: The genetic renaissance of fish and frogs. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 28:41-52. [PMID: 30205880 DOI: 10.1016/j.ddtec.2018.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/29/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022]
Abstract
The speed by which clinical genomics is currently identifying novel potentially pathogenic variants is outperforming the speed by which these can be functionally (genotype-phenotype) annotated in animal disease models. However, over the past few years the emergence of CRISPR/Cas9 as a straight-forward genome editing technology has revolutionized disease modeling in vertebrate non-mammalian model organisms such as zebrafish, medaka and Xenopus. It is now finally possible, by CRISPR/Cas9, to rapidly establish clinically relevant disease models in these organisms. Interestingly, these can provide both cost-effective genotype-phenotype correlations for gene-(variants) and genomic rearrangements obtained from clinical practice, as well as be exploited to perform translational research to improve prospects of disease afflicted patients. In this review, we show an extensive overview of these new CRISPR/Cas9-mediated disease models and provide future prospects that will allow increasingly accurate modeling of human disease in zebrafish, medaka and Xenopus.
Collapse
Affiliation(s)
- Thomas Naert
- Department of Biomedical Molecular Biology, Ghent University, Belgium; Cancer Research Institute Ghent, Belgium
| | - Kris Vleminckx
- Department of Biomedical Molecular Biology, Ghent University, Belgium; Center for Medical Genetics, Ghent University, Belgium; Cancer Research Institute Ghent, Belgium.
| |
Collapse
|
38
|
Dona M, Slijkerman R, Lerner K, Broekman S, Wegner J, Howat T, Peters T, Hetterschijt L, Boon N, de Vrieze E, Sorusch N, Wolfrum U, Kremer H, Neuhauss S, Zang J, Kamermans M, Westerfield M, Phillips J, van Wijk E. Usherin defects lead to early-onset retinal dysfunction in zebrafish. Exp Eye Res 2018; 173:148-159. [PMID: 29777677 DOI: 10.1016/j.exer.2018.05.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 01/24/2023]
Abstract
Mutations in USH2A are the most frequent cause of Usher syndrome and autosomal recessive nonsyndromic retinitis pigmentosa. To unravel the pathogenic mechanisms underlying USH2A-associated retinal degeneration and to evaluate future therapeutic strategies that could potentially halt the progression of this devastating disorder, an animal model is needed. The available Ush2a knock-out mouse model does not mimic the human phenotype, because it presents with only a mild and late-onset retinal degeneration. Using CRISPR/Cas9-technology, we introduced protein-truncating germline lesions into the zebrafish ush2a gene (ush2armc1: c.2337_2342delinsAC; p.Cys780GlnfsTer32 and ush2ab1245: c.15520_15523delinsTG; p.Ala5174fsTer). Homozygous mutants were viable and displayed no obvious morphological or developmental defects. Immunohistochemical analyses with antibodies recognizing the N- or C-terminal region of the ush2a-encoded protein, usherin, demonstrated complete absence of usherin in photoreceptors of ush2armc1, but presence of the ectodomain of usherin at the periciliary membrane of ush2ab1245-derived photoreceptors. Furthermore, defects of usherin led to a reduction in localization of USH2 complex members, whirlin and Adgrv1, at the photoreceptor periciliary membrane of both mutants. Significantly elevated levels of apoptotic photoreceptors could be observed in both mutants when kept under constant bright illumination for three days. Electroretinogram (ERG) recordings revealed a significant and similar decrease in both a- and b-wave amplitudes in ush2armc1 as well as ush2ab1245 larvae as compared to strain- and age-matched wild-type larvae. In conclusion, this study shows that mutant ush2a zebrafish models present with early-onset retinal dysfunction that is exacerbated by light exposure. These models provide a better understanding of the pathophysiology underlying USH2A-associated RP and a unique opportunity to evaluate future therapeutic strategies.
Collapse
Affiliation(s)
- Margo Dona
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Ralph Slijkerman
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Kimberly Lerner
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Sanne Broekman
- Department of Human Genetics, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Jeremy Wegner
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Taylor Howat
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Theo Peters
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Lisette Hetterschijt
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Nanda Boon
- Department of Human Genetics, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| | - Erik de Vrieze
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Nasrin Sorusch
- Institute of Molecular Physiology, Johannes Gutenberg University, Johannes-von-Muellerweg 6, D-55099 Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University, Johannes-von-Muellerweg 6, D-55099 Mainz, Germany
| | - Hannie Kremer
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Department of Human Genetics, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Stephan Neuhauss
- University of Zürich, Institute of Molecular Life Sciences, Winterthurerstrasse 190, Zürich, CH - 8057, Switzerland
| | - Jingjing Zang
- University of Zürich, Institute of Molecular Life Sciences, Winterthurerstrasse 190, Zürich, CH - 8057, Switzerland
| | - Maarten Kamermans
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; Department of Biomedical Physics, Academisch Medisch Centrum, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Monte Westerfield
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Jennifer Phillips
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Erwin van Wijk
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands.
| |
Collapse
|
39
|
Fernandes Y, Buckley DM, Eberhart JK. Diving into the world of alcohol teratogenesis: a review of zebrafish models of fetal alcohol spectrum disorder. Biochem Cell Biol 2018; 96:88-97. [PMID: 28817785 PMCID: PMC7413215 DOI: 10.1139/bcb-2017-0122] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The term fetal alcohol spectrum disorder (FASD) refers to the entire suite of deleterious outcomes resulting from embryonic exposure to alcohol. Along with other reviews in this special issue, we provide insight into how animal models, specifically the zebrafish, have informed our understanding of FASD. We first provide a brief introduction to FASD. We discuss the zebrafish as a model organism and its strengths for alcohol research. We detail how zebrafish has been used to model some of the major defects present in FASD. These include behavioral defects, such as social behavior as well as learning and memory, and structural defects, disrupting organs such as the brain, sensory organs, heart, and craniofacial skeleton. We provide insights into how zebrafish research has aided in our understanding of the mechanisms of ethanol teratogenesis. We end by providing some relatively recent advances that zebrafish has provided in characterizing gene-ethanol interactions that may underlie FASD.
Collapse
Affiliation(s)
- Yohaan Fernandes
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78713, USA
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78713, USA
| | - Desire M Buckley
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78713, USA
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78713, USA
| | - Johann K Eberhart
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78713, USA
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78713, USA
| |
Collapse
|
40
|
Hoenig M, Pannicke U, Gaspar HB, Schwarz K. Recent advances in understanding the pathogenesis and management of reticular dysgenesis. Br J Haematol 2017; 180:644-653. [PMID: 29270983 DOI: 10.1111/bjh.15045] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Reticular Dysgenesis is a rare immunodeficiency which is clinically characterized by the combination of Severe Combined Immunodeficiency (SCID) with agranulocytosis and sensorineural deafness. Mutations in the gene encoding adenylate kinase 2 (AK2) were identified to cause this phenotype. In this review, we will demonstrate important clinical differences between reticular dysgenesis and other SCID entities and summarize recent concepts in the understanding of the pathophysiology of the disease and the management strategies for this difficult condition.
Collapse
Affiliation(s)
- Manfred Hoenig
- Department of Paediatrics, University Medical Centre Ulm, Ulm, Germany
| | - Ulrich Pannicke
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg, Hessen, Germany
| | - Hubert B Gaspar
- UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - Klaus Schwarz
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg, Hessen, Germany
| |
Collapse
|
41
|
Abstract
Our ears are remarkable sensory organs, providing the important senses of balance and hearing. The complex structure of the inner ear, or 'labyrinth', along with the assorted neuroepithelia, have evolved to detect head movements and sounds with impressive sensitivity. The rub is that the inner ear is highly vulnerable to genetic lesions and environmental insults. According to National Institute of Health estimates, hearing loss is one of the most commonly inherited or acquired sensorineural diseases. To understand the causes of deafness and balance disorders, it is imperative to understand the underlying biology of the inner ear, especially the inner workings of the sensory receptors. These receptors, which are termed hair cells, are particularly susceptible to genetic mutations - more than two dozen genes are associated with defects in this cell type in humans. Over the past decade, a substantial amount of progress has been made in working out the molecular basis of hair-cell function using vertebrate animal models. Given the transparency of the inner ear and the genetic tools that are available, zebrafish have become an increasingly popular animal model for the study of deafness and vestibular dysfunction. Mutagenesis screens for larval defects in hearing and balance have been fruitful in finding key components, many of which have been implicated in human deafness. This review will focus on the genes that are required for hair-cell function in zebrafish, with a particular emphasis on mechanotransduction. In addition, the generation of new tools available for the characterization of zebrafish hair-cell mutants will be discussed.
Collapse
Affiliation(s)
- Teresa Nicolson
- Oregon Hearing Research Center and the Vollum Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, Tel: 503-494-3693,
| |
Collapse
|
42
|
Matsubara H, Matsui Y, Miyata R, Nishimura Y, Yamamoto T, Tanaka T, Kondo M. New photic stimulating system with white light-emitting diodes to elicit electroretinograms from zebrafish larvae. Doc Ophthalmol 2017; 135:147-154. [PMID: 28756596 PMCID: PMC5606953 DOI: 10.1007/s10633-017-9602-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/25/2017] [Indexed: 12/24/2022]
Abstract
Purpose The zebrafish is an established animal model commonly used in biological, neuroscience, and genetic research. We have developed a new light stimulating system using white light-emitting diodes (LEDs) to elicit ERGs from zebrafish larvae. The purpose of this study was to record full-field ERGs and to evaluate the inter-trial reliability of the ERGs recorded with our system from zebrafish larvae. Methods The stimulating device used white LEDs that were attached to a stereomicroscope, and the location of the recording electrode on the cornea could be monitored while the eye was being stimulated. Full-field scotopic and photopic ERGs were recorded from larvae at the age of 5–7 days post-fertilization (dpf). Intensity–response curves were constructed from the ERGs. Inter-trial reliability of the ERGs recorded by our system was evaluated. Results This stimulating system could be used for efficient and reliable ERG recordings from 5–7 dpf larvae. The amplitudes, implicit times, and the waveforms of the scotopic and photopic ERGs were similar to those reported in earlier studies. Inter-trial reliability of the amplitudes of the photopic ERG b-waves was excellent with an intra-class correlation coefficient of 0.98. Conclusion We conclude that this new light stimulation system using white LEDs attached to a stereomicroscope will be helpful in recording reliable ERGs from zebrafish larvae.
Collapse
Affiliation(s)
- Hisashi Matsubara
- Department of Ophthalmology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan.
| | - Yoshitsugu Matsui
- Department of Ophthalmology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Ryohei Miyata
- Department of Ophthalmology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Yuhei Nishimura
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Tetsuro Yamamoto
- Department of Neurophysiology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Toshio Tanaka
- Department of Systems Pharmacology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Mineo Kondo
- Department of Ophthalmology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| |
Collapse
|