1
|
McCann T, Sundaramurthi H, Walsh C, Virdi S, Alvarez Y, Sapetto-Rebow B, Collery RF, Carter SP, Moran A, Mulholland R, O'Connor JJ, Taylor MR, Rauch N, Starostik MR, English MA, Swaroop A, Geisler R, Reynolds AL, Kennedy BN. Emc1 is essential for vision and zebrafish photoreceptor outer segment morphogenesis. FASEB J 2024; 38:e70086. [PMID: 39360639 DOI: 10.1096/fj.202401977r] [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: 08/23/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
Inherited retinal diseases (IRDs) are a rare group of eye disorders characterized by progressive dysfunction and degeneration of retinal cells. In this study, we characterized the raifteirí (raf) zebrafish, a novel model of inherited blindness, identified through an unbiased ENU mutagenesis screen. A mutation in the largest subunit of the endoplasmic reticulum membrane protein complex, emc1 was subsequently identified as the causative raf mutation. We sought to elucidate the cellular and molecular phenotypes in the emc1-/- knockout model and explore the association of emc1 with retinal degeneration. Visual behavior and retinal electrophysiology assays demonstrated that emc1-/- mutants had severe visual impairments. Retinal histology and morphometric analysis revealed extensive abnormalities, including thinning of the photoreceptor layer, in addition to large gaps surrounding the lens. Notably, photoreceptor outer segments were drastically smaller, outer segment protein expression was altered and hyaloid vasculature development was disrupted. Transcriptomic profiling identified cone and rod-specific phototransduction genes significantly downregulated by loss of emc1. These data shed light on why emc1 is a causative gene in inherited retinal disease and how outer segment morphogenesis is regulated.
Collapse
Affiliation(s)
- Tess McCann
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Husvinee Sundaramurthi
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Ciara Walsh
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Sanamjeet Virdi
- Karlsruhe Institute of Technology (KIT) Institute of Biological and Chemical Systems -Biological Information Processing, Eggenstein-Leopoldshafen, Germany
- Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Yolanda Alvarez
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Beata Sapetto-Rebow
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Ross F Collery
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
- Medical College of Wisconsin Eye Institute, Milwaukee, Wisconsin, USA
| | - Stephen P Carter
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Ailis Moran
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Ruth Mulholland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - John J O'Connor
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Michael R Taylor
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nora Rauch
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| | - Margaret R Starostik
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Milton A English
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Robert Geisler
- Karlsruhe Institute of Technology (KIT) Institute of Biological and Chemical Systems -Biological Information Processing, Eggenstein-Leopoldshafen, Germany
| | - Alison L Reynolds
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
- School of Veterinary Medicine, Veterinary Science Centre, University College Dublin, Dublin, Ireland
| | - Breandán N Kennedy
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Wen L, Man X, Luan J, Zhang S, Zhao C, Bao Y, Liu C, Feng X. Early-life exposure to five biodegradable plastics impairs eye development and visually-mediated behavior through disturbing hypothalamus-pituitary-thyroid (HPT) axis in zebrafish larvae. Comp Biochem Physiol C Toxicol Pharmacol 2024; 284:109981. [PMID: 39033795 DOI: 10.1016/j.cbpc.2024.109981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/09/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
Biodegradable plastics have been commonly developed and applied as an alternative to traditional plastics, which cause environmental plastic pollution. However, biodegradable plastics still present limitations such as stringent degradation conditions and slow degradation rate, and may cause harm to the environment and organisms. Consequently, in this study, zebrafish was used to evaluate the effects of five biodegradable microplastics (MPs), polyglycolic acid (PGA), polylactic acid (PLA), polybutylene succinate (PBS), polyhydroxyalkanoate (PHA) and polybutylene adipate terephthalate (PBAT) exposure on the early development, retina morphology, visually-mediated behavior, and thyroid signaling at concentrations of 1 mg/L and 100 mg/L. The results indicated that all MPs induced decreased survival rate, reduced body length, smaller eyes, and smaller heads, affecting the early development of zebrafish larvae. Moreover, the thickness of retinal layers, including inner plexiform layer (IPL), outer nuclear layer (ONL), and retinal ganglion layer (RGL) was decreased, and the expression of key genes related to eye and retinal development was abnormally altered after all MPs exposure. Exposure to PBS and PBAT led to abnormal visually-mediated behavior, indicating likely affected the visual function. All MPs could also cause thyroid system disorders, among which alterations in the thyroid hormone receptors (TRs) genes could affect the retinal development of zebrafish larvae. In summary, biodegradable MPs exhibited eye developmental toxicity and likely impaired the visual function in zebrafish larvae. This provided new evidence for revealing the effects of biodegradable plastics on aquatic organism development and environmental risks to aquatic ecosystems.
Collapse
Affiliation(s)
- Liang Wen
- China Shenhua Coal to Liquid and Chemical CO., LTD. of China Energy, Beijing 100011, China
| | - Xiaoting Man
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Jialu Luan
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Shuhui Zhang
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Chengtian Zhao
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Yehua Bao
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Congzhi Liu
- China Shenhua Coal to Liquid and Chemical CO., LTD. of China Energy, Beijing 100011, China.
| | - Xizeng Feng
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
3
|
Ávila-Mendoza J, Urban-Sosa VA, Lazcano I, Orozco A, Luna M, Martínez-Moreno CG, Arámburo C. Comparative analysis of Krüppel-like factors expression in the retinas of zebrafish and mice during development and after injury. Gen Comp Endocrinol 2024; 356:114579. [PMID: 38964422 DOI: 10.1016/j.ygcen.2024.114579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
The Krüppel-like factors (KLFs) have emerged as important transcriptional regulators of various cellular processes, including neural development. Some of them have been described as intrinsic factors involved in axon regeneration in the central nervous system (CNS) of vertebrates. Zebrafish are known for their ability to regenerate several tissues in adulthood, including the CNS, a capability lost during vertebrate evolution and absent in adult mammals. The role that KLFs could play in this differential ability remains unknown. Therefore, in this study, we analyzed the endogenous response of certain KLFs implicated in axon regeneration (KLFs 6, 7, 9, and 13) during retina development and after axon injury. The results showed that the expression of Klfs 6, 7, and 13 decreases in the developing retina of mice but not in zebrafish, while the mRNA levels of Klf9 strongly increase in both species. The response to injury was further analyzed using optic nerve crush (ONC) as a model of lesion. Our analysis during the acute phase (hours) demonstrated an induction of Klfs 6 and 7 expression exclusively in the zebrafish retina, while Klfs 9 and 13 mRNA levels increased in both species. Further analysis of the chronic response (days) showed that mRNA levels of Klf6 transiently increase in the retinas of both zebrafish and mice, whereas those of Klf7 decrease later after optic nerve injury. In addition, the analysis revealed that the expression of Klf9 decreases, while that of Klf13 increases in the retinas of zebrafish in response to optic nerve injury but remains unaltered in mice. Altogether, these findings support the hypothesis that KLFs may play a role in the differential axon regeneration abilities exhibited by fish and mice.
Collapse
Affiliation(s)
- José Ávila-Mendoza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico.
| | - Valeria A Urban-Sosa
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Iván Lazcano
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Aurea Orozco
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Maricela Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Carlos G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico.
| |
Collapse
|
4
|
Safrina O, Vorontsova I, Donaldson PJ, Schilling TF. Zebrafish Optical Development Requires Regulated Water Permeability by Aquaporin 0. Invest Ophthalmol Vis Sci 2024; 65:42. [PMID: 39330988 PMCID: PMC11437712 DOI: 10.1167/iovs.65.11.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024] Open
Abstract
Purpose Optical development of the zebrafish eye relies on the movement of the highly refractive lens nucleus from an anterior to a central location in the optical axis during development. We have shown that this mechanism in turn depends on the function of Aquaporin 0a (Aqp0a), a multifunctional and extremely abundant protein in lens fiber cell membranes. Here, we probe the specific cellular functions necessary for rescuing lens nucleus centralization defects in aqp0a-/- null mutants by stable overexpression of an Aqp0 orthologue from a killifish, MIPfun. Methods We test in vivo requirements for lens transparency and nucleus centralization of MIPfun for auto-adhesion, water permeability (Pf), and Pf sensitivity to regulation by Ca2+ or pH by overexpression of MIPfun mutants previously shown to have defects in these functions in vitro or in silico. Results Water permeability of MIPfun is essential for rescuing lens transparency and nucleus centralization defects, whereas auto-adhesion is not. Furthermore, water permeability regulation by Ca2+ and pH appear residue-dependent, because some Ca2+-insensitive mutants fail to rescue, and pH-insensitive mutants only partially rescue defects. MIPfun lacking Pf sensitivity to both, Ca2+ and pH, also fails to rescue lens nucleus centralization. Conclusion This study shows that regulation of water permeability by Aqp0 plays a key role in the centralization of the zebrafish lens nucleus, providing the first direct evidence for water transport in this aspect of optical development.
Collapse
Affiliation(s)
- Olga Safrina
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States
| | - Irene Vorontsova
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States
- Department of Physiology, The University of Auckland, Aotearoa New Zealand National Eye Centre, Auckland, New Zealand
| | - Paul J Donaldson
- Department of Physiology, The University of Auckland, Aotearoa New Zealand National Eye Centre, Auckland, New Zealand
| | - Thomas F Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States
| |
Collapse
|
5
|
Xie J, Goodbourn PT, Bui BV, Jusuf PR. Establishment and comprehensive characterization of a novel dark-reared zebrafish model for myopia studies. Exp Eye Res 2024; 246:110009. [PMID: 39067805 DOI: 10.1016/j.exer.2024.110009] [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: 04/14/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Myopia is predicted to impact approximately 5 billion people by 2050, necessitating mechanistic understanding of its development. Myopia results from dysregulated genetic mechanisms of emmetropization, caused by over-exposure to aberrant visual environments; however, these genetic mechanisms remain unclear. Recent human genome-wide association studies have identified a range of novel myopia-risk genes. To facilitate large-scale in vivo mechanistic examination of gene-environment interactions, this study aims to establish a myopia model platform that allows efficient environmental and genetic manipulations. We established an environmental zebrafish myopia model by dark-rearing. Ocular biometrics including relative ocular refraction were quantified using optical coherence tomography images. Spatial vision was assessed using optomotor response (OMR). Retinal function was analyzed via electroretinography (ERG). Myopia-associated molecular contents or distributions were examined using RT-qPCR or immunohistochemistry. Our model produces robust phenotypic changes, showing myopia after 2 weeks of dark-rearing, which were recoverable within 2 weeks after returning animals to normal lighting. 2-week dark-reared zebrafish have reduced spatial-frequency tuning function. ERG showed reduced photoreceptor and bipolar cell function (a- and b-waves) after only 2 days of dark-rearing, which worsened after 2 weeks of dark-rearing. We also found dark-rearing-induced changes to expression of myopia-risk genes, including egr1, vegfaa, vegfab, rbp3, gjd2a and gjd2b, inner retinal distribution of EFEMP1, TIMP2 and MMP2, as well as transiently reduced PSD95 density in the inner plexiform layer. Coupled with the gene editing tools available for zebrafish, our environmental myopia model provides an excellent platform for large-scale investigation of gene-environment interactions in myopia development.
Collapse
Affiliation(s)
- Jiaheng Xie
- School of Biosciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Patrick T Goodbourn
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| | - Patricia Regina Jusuf
- School of Biosciences, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| |
Collapse
|
6
|
Saied-Santiago K, Baxter M, Mathiaparanam J, Granato M. Serotonin neuromodulation directs optic nerve regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607648. [PMID: 39185204 PMCID: PMC11343150 DOI: 10.1101/2024.08.12.607648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Optic nerve (ON) regeneration in mammalian systems is limited by an overshadowing dominance of inhibitory factors. This has severely hampered the identification of pro-regenerative pathways. Here, we take advantage of the regenerative capacity of larval zebrafish to identify pathways that promote ON regeneration. From a small molecule screen, we identified modulators of serotonin (5-HT) signaling that inhibit ON regeneration. We find several serotonin type-1 receptor genes are expressed in RGC neurons during regeneration and that inhibiting 5-HT1 receptors or components of the 5-HT pathway selectively impedes ON regeneration. We show that 5-HT1 receptor signaling is dispensable during ON development yet is critical for regenerating axons to emerge from the injury site. Blocking 5-HT receptors once ON axons have crossed the chiasm does not inhibit regeneration, suggesting a selective role for 5-HT receptor signaling early during ON regeneration. Finally, we show that agonist-mediated activation of 5-HT1 receptors leads to enhanced and ectopic axonal regrowth. Combined, our results provide evidence for mechanisms through which serotonin-dependent neuromodulation directs ON regeneration in vivo.
Collapse
Affiliation(s)
- Kristian Saied-Santiago
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Melissa Baxter
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Jaffna Mathiaparanam
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| |
Collapse
|
7
|
Saputra F, Kishida M, Hu SY. Oxidative stress induced by hydrogen peroxide disrupts zebrafish visual development by altering apoptosis, antioxidant and estrogen related genes. Sci Rep 2024; 14:14454. [PMID: 38914633 PMCID: PMC11196719 DOI: 10.1038/s41598-024-64933-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/14/2024] [Indexed: 06/26/2024] Open
Abstract
Hydrogen peroxide is considered deleterious molecule that cause cellular damage integrity and function. Its key redox signaling molecule in oxidative stress and exerts toxicity on a wide range of organisms. Thus, to understand whether oxidative stress alters visual development, zebrafish embryos were exposed to H2O2 at concentration of 0.02 to 62.5 mM for 7 days. Eye to body length ratio (EBR) and apoptosis in retina at 48 hpf, and optomotor response (OMR) at 7 dpf were all measured. To investigate whether hydrogen peroxide-induced effects were mediated by oxidative stress, embryos were co-incubated with the antioxidant, glutathione (GSH) at 50 μM. Results revealed that concentrations of H2O2 at or above 0.1 mM induced developmental toxicity, leading to increased mortality and hatching delay. Furthermore, exposure to 0.1 mM H2O2 decreased EBR at 48 hpf and impaired OMR visual behavior at 7 dpf. Additionally, exposure increased the area of apoptotic cells in the retina at 48 hpf. The addition of GSH reversed the effects of H2O2, suggesting the involvement of oxidative stress. H2O2 decreased the expression of eye development-related genes, pax6α and pax6β. The expression of apoptosis-related genes, tp53, casp3 and bax, significantly increased, while bcl2α expression decreased. Antioxidant-related genes sod1, cat and gpx1a showed decreased expression. Expression levels of estrogen receptors (ERs) (esr1, esr2α, and esr2β) and ovarian and brain aromatase genes (cyp19a1a and cyp19a1b, respectively) were also significantly reduced. Interestingly, co-incubation of GSH effectivity reversed the impact of H2O2 on most parameters. Overall, these results demonstrate that H2O2 induces adverse effects on visual development via oxidative stress, which leads to alter apoptosis, diminished antioxidant defenses and reduced estrogen production.
Collapse
Affiliation(s)
| | - Mitsuyo Kishida
- Graduate School of Science and Technology, Kumamoto University, Kumamoto, Japan.
| | - Shao-Yang Hu
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, Taiwan.
| |
Collapse
|
8
|
Gölz L, Pannetier P, Fagundes T, Knörr S, Behnstedt L, Coordes S, Matthiessen P, Morthorst J, Vergauwen L, Knapen D, Holbech H, Braunbeck T, Baumann L. Development of the integrated fish endocrine disruptor test-Part B: Implementation of thyroid-related endpoints. INTEGRATED ENVIRONMENTAL ASSESSMENT AND MANAGEMENT 2024; 20:830-845. [PMID: 37578010 DOI: 10.1002/ieam.4828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/21/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Given the vital role of thyroid hormones (THs) in vertebrate development, it is essential to identify chemicals that interfere with the TH system. Whereas, among nonmammalian laboratory animals, fish are the most frequently utilized test species in endocrine disruptor research, for example, in guidelines for the detection of effects on the sex hormone system, there is no test guideline (TG) using fish as models for thyroid-related effects; rather, amphibians are used. Therefore, the objective of the present project was to integrate thyroid-related endpoints for fish into a test protocol combining OECD TGs 229 (Fish Short-Term Reproduction Assay) and 234 (Fish Sexual Development Test). The resulting integrated Fish Endocrine Disruption Test (iFEDT) was designed as a comprehensive approach to covering sexual differentiation, early development, and reproduction and to identifying disruption not only of the sexual and/or reproductive system but also the TH system. Two 85-day exposure tests were performed using different well-studied endocrine disruptors: 6-propyl-2-thiouracil (PTU) and 17α-ethinylestradiol (EE2). Whereas the companion Part A of this study presents the findings on effects by PTU and EE2 on endpoints established in existing TGs, the present Part B discusses effects on novel thyroid-related endpoints such as TH levels, thyroid follicle histopathology, and eye development. 6-Propyl-2-thiouracil induced a massive proliferation of thyroid follicles in any life stage, and histopathological changes in the eyes proved to be highly sensitive for TH system disruption especially in younger life stages. For measurement of THs, further methodological development is required. 17-α-Ethinylestradiol demonstrated not only the well-known disruption of the hypothalamic-pituitary-gonadal axis, but also induced effects on thyroid follicles in adult zebrafish (Danio rerio) exposed to higher EE2 concentrations, suggesting crosstalk between endocrine axes. The novel iFEDT has thus proven capable of simultaneously capturing endocrine disruption of both the steroid and thyroid endocrine systems. Integr Environ Assess Manag 2024;20:830-845. © 2023 The Authors. Integrated Environmental Assessment and Management published by Wiley Periodicals LLC on behalf of Society of Environmental Toxicology & Chemistry (SETAC).
Collapse
Affiliation(s)
- Lisa Gölz
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Pauline Pannetier
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
- Laboratoire de Ploufragan-Plouzané-Niort, Site de Plouzané, Agence nationale de sécurité sanitaire de l'alimentation, de l'environnement et du travail, Plouzané, France
| | - Teresa Fagundes
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Susanne Knörr
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Laura Behnstedt
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Sara Coordes
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | | | - Jane Morthorst
- Department of Biology, University of Southern Denmark, Odense, Denmark
| | - Lucia Vergauwen
- Department of Veterinary Sciences, Veterinary Physiology and Biochemistry, Zebrafishlab, University of Antwerp, Wilrijk, Belgium
| | - Dries Knapen
- Department of Veterinary Sciences, Veterinary Physiology and Biochemistry, Zebrafishlab, University of Antwerp, Wilrijk, Belgium
| | - Henrik Holbech
- Department of Biology, University of Southern Denmark, Odense, Denmark
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Lisa Baumann
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
- Amsterdam Institute for Life and Environment (A-LIFE), Section Environmental Health & Toxicology, Vrije Universiteit Amsterdam, HV Amsterdam, The Netherlands
| |
Collapse
|
9
|
Roy D, Subramaniam B, Chong WC, Bornhorst M, Packer RJ, Nazarian J. Zebrafish-A Suitable Model for Rapid Translation of Effective Therapies for Pediatric Cancers. Cancers (Basel) 2024; 16:1361. [PMID: 38611039 PMCID: PMC11010887 DOI: 10.3390/cancers16071361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Pediatric cancers are the leading cause of disease-related deaths in children and adolescents. Most of these tumors are difficult to treat and have poor overall survival. Concerns have also been raised about drug toxicity and long-term detrimental side effects of therapies. In this review, we discuss the advantages and unique attributes of zebrafish as pediatric cancer models and their importance in targeted drug discovery and toxicity assays. We have also placed a special focus on zebrafish models of pediatric brain cancers-the most common and difficult solid tumor to treat.
Collapse
Affiliation(s)
- Debasish Roy
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Bavani Subramaniam
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Wai Chin Chong
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Miriam Bornhorst
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Roger J. Packer
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Javad Nazarian
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
- DIPG/DMG Research Center Zurich, Children’s Research Center, Department of Pediatrics, University Children’s Hospital Zürich, 8032 Zurich, Switzerland
| |
Collapse
|
10
|
Peng X, Jia X, Shang G, Xue M, Jiang M, Chen D, Zhang F, Hu Y. The generation and characterization of a transgenic zebrafish line with lens-specific Cre expression. Mol Vis 2024; 30:123-136. [PMID: 38601019 PMCID: PMC11006009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 03/17/2024] [Indexed: 04/12/2024] Open
Abstract
Purpose Danio rerio zebrafish constitute a popular model for studying lens development and congenital cataracts. However, the specific deletion of a gene with a Cre/LoxP system in the zebrafish lens is unavailable because of the lack of a lens-Cre-transgenic zebrafish. This study aimed to generate a transgenic zebrafish line in which Cre recombinase was specifically expressed in the lens. Methods The pTol2 cryaa:Cre-polyA-cryaa:EGFP (enhanced green fluorescent protein) plasmid was constructed and co-injected with Tol2-transposase into one-to-two-cell-stage wild-type (WT) zebrafish embryos. Whole-mount in situ hybridization (ISH), tissue section, hematoxylin and eosin staining, a Western blot, a split-lamp observation, and a grid transmission assay were used to analyze the Cre expression, lens structure, and lens transparency of the transgenic zebrafish. Results In this study, we generated a transgenic zebrafish line, zTg(cryaa:Cre-cryaa:EGFP), in which Cre recombinase and EGFP were driven by the lens-specific cryaa promoter. zTg(cryaa:Cre-cryaa:EGFP) began to express Cre and EGFP specifically in the lens at the 22 hpf stage, and this ectopic Cre could efficiently and specifically delete the red fluorescent protein (RFP) signal from the lens when zTg(cryaa:Cre-cryaa:EGFP) embryos were injected with the loxP-flanked RFP plasmid. The overexpression of Cre and EGFP did not impair zebrafish development or lens transparency. Accordingly, this zTg(cryaa:Cre-cryaa:EGFP) zebrafish line is a useful tool for gene editing, specifically with zebrafish lenses. Conclusions We established a zTg(cryaa:Cre-cryaa:EGFP) zebrafish line that can specifically express an active Cre recombinase in lens tissues. This transgenic zebrafish line can be used as a tool to specifically manipulate a gene in zebrafish lenses.
Collapse
Affiliation(s)
- Xuyan Peng
- The Laboratory of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zheng Zhou, China
| | - Xiaolin Jia
- The Laboratory of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zheng Zhou, China
| | - Guohui Shang
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zheng Zhou, China
| | - Mengjiao Xue
- The Laboratory of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zheng Zhou, China
| | - Mingjun Jiang
- The Laboratory of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zheng Zhou, China
| | - Dandan Chen
- The Laboratory of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zheng Zhou, China
| | - Fengyan Zhang
- The Laboratory of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zheng Zhou, China
| | - Yanzhong Hu
- The Laboratory of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zheng Zhou, China
- Joint National Laboratory for Antibody Drug Engineering, Henan University School of Basic Medical Sciences. Kaifeng, China
- Kaifeng Key Lab for Cataract and Myopia, Institute of Eye Disease, Kaifeng Central Hospital, Kaifeng, China
| |
Collapse
|
11
|
Deering MJ, Paradis H, Ahmad R, Al-Mehiawi AS, Gendron RL. The role of dietary vitamin A in mechanisms of cataract development in the teleost lumpfish (Cyclopterus lumpus L). JOURNAL OF FISH DISEASES 2024; 47:e13899. [PMID: 38041393 DOI: 10.1111/jfd.13899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/15/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023]
Abstract
Lumpfish (Cyclopterus lumpus L) are highly prone to cataract development in the wild and in culture. There is evidence that cataract in farmed fish is related to nutrition. However, both the nutrients and the mechanisms involved in cataract development in lumpfish are not clear. Here we investigated the mechanisms involved and the role of dietary vitamin A in cataract development in a cultured lumpfish population. Cultured lumpfish were fed three diets differing only in vitamin A supplementation level (5000, 15,000 and 120,000 IU/kg) over an 18-month period, and fish weight, cataract frequencies and severities were determined. Western blotting and immunohistochemistry were performed on lens tissue to measure the levels of oxidative stress, and apoptosis. The lowest levels of vitamin A significantly reduced cataract frequencies in adult lumpfish and resulted in less severe cataract and increased weight in males. Oxidative stress levels in the lens were positively correlated with vitamin A intake. Apoptosis was observed at high levels in lenses with severe cataract. Oxidative stress and apoptosis levels were the highest in regions of the lens with severe, advanced cataract pathology when compared to regions with no visible pathology. These results suggest that higher vitamin A intake contributes to cataract development through an oxidative stress pathway, and that both oxidative stress and apoptosis are involved in advanced stages of cataract in lumpfish.
Collapse
Affiliation(s)
- Margret J Deering
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Hélène Paradis
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Raahyma Ahmad
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Adil S Al-Mehiawi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Robert L Gendron
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
12
|
Verma SK, Nandi A, Sinha A, Patel P, Mohanty S, Jha E, Jena S, Kumari P, Ghosh A, Jerman I, Chouhan RS, Dutt A, Samal SK, Mishra YK, Varma RS, Panda PK, Kaushik NK, Singh D, Suar M. The posterity of Zebrafish in paradigm of in vivo molecular toxicological profiling. Biomed Pharmacother 2024; 171:116160. [PMID: 38237351 DOI: 10.1016/j.biopha.2024.116160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
The aggrandised advancement in utility of advanced day-to-day materials and nanomaterials has raised serious concern on their biocompatibility with human and other biotic members. In last few decades, understanding of toxicity of these materials has been given the centre stage of research using many in vitro and in vivo models. Zebrafish (Danio rerio), a freshwater fish and a member of the minnow family has garnered much attention due to its distinct features, which make it an important and frequently used animal model in various fields of embryology and toxicological studies. Given that fertilization and development of zebrafish eggs take place externally, they serve as an excellent model organism for studying early developmental stages. Moreover, zebrafish possess a comparable genetic composition to humans and share almost 70% of their genes with mammals. This particular model organism has become increasingly popular, especially for developmental research. Moreover, it serves as a link between in vitro studies and in vivo analysis in mammals. It is an appealing choice for vertebrate research, when employing high-throughput methods, due to their small size, swift development, and relatively affordable laboratory setup. This small vertebrate has enhanced comprehension of pathobiology and drug toxicity. This review emphasizes on the recent developments in toxicity screening and assays, and the new insights gained about the toxicity of drugs through these assays. Specifically, the cardio, neural, and, hepatic toxicology studies inferred by applications of nanoparticles have been highlighted.
Collapse
Affiliation(s)
- Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar, India.
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Adrija Sinha
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Paritosh Patel
- School of Biotechnology, KIIT University, Bhubaneswar, India; Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897, Seoul, South Korea
| | | | - Ealisha Jha
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Snehasmita Jena
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Puja Kumari
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno 61137, Czech Republic
| | - Aishee Ghosh
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Ivan Jerman
- National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Raghuraj Singh Chouhan
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Ateet Dutt
- Instituto de Investigaciones en Materiales, UNAM, CDMX, Mexico
| | - Shailesh Kumar Samal
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, Alsion 2, Sønderborg DK-6400, Denmark
| | - Rajender S Varma
- Institute for Nanomaterials, Advanced Technologies and Innovation (CxI), Technical University of Liberec (TUL), Studentská 1402/2, Liberec 1 461 17, Czech Republic
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897, Seoul, South Korea.
| | - Deobrat Singh
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar, India.
| |
Collapse
|
13
|
de Arruda Leite B, Meireles G, Abe FR, Gravato C, Dorta DJ, de Oliveira DP. Do zebrafish become blind or is it too much red dye in water? Distinguishing the embryo-larval development and physiology effects of DR 60, 73, and 78. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168062. [PMID: 37884151 DOI: 10.1016/j.scitotenv.2023.168062] [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: 05/22/2023] [Revised: 09/20/2023] [Accepted: 10/21/2023] [Indexed: 10/28/2023]
Abstract
Some dyes currently used by the textile, pharmaceutical, food, cosmetic, and photographic industries have been shown to be toxic and/or mutagenic to aquatic life. Most of these dyes resist degradation processes available for treating wastewater, and these processes might generate even more toxic by-products. Despite the large number of available dyes and the large quantity of dyes released into the environment, studies on their toxicity are still scarce. We evaluated and compared the effects in the animal model Danio rerio (zebrafish) of environmentally relevant concentrations of Disperse Red 60 (DR 60), 73 (DR 73), and 78 (DR 78) using the fish embryo acute toxicity (FET) test, morphometric analysis, immunofluorescence imaging, and behavioral parameters. DR 60 caused ocular modifications, while the DR 73 caused non-inflation of the swim bladder (NISB), pericardial edema (PE), scoliosis (S) and abnormal yolk sac (AYS) from at 0.125 mg/L. In behavioral tests, all the dyes induced changes in velocity and time spent swimming of exposed larvae. However, these alterations in behavior seem to be caused by different factors dependent on the dye and its concentration. Nevertheless, behavior seems to add valuable information concerning the hazards analysis of dyes, since it reveals to be the most sensitive group of parameters tested in the current study. In conclusion, of the behavioral and developmental alterations caused by these dyes should be interpreted as an alert for greater attention when registering new dyes and releasing them into the environment. In the particular case of DR 60 the possibility that directly affects the eye of larvae is of great environmental concern, but also from the human health perspective.
Collapse
Affiliation(s)
- Bianca de Arruda Leite
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, 14040-903 Ribeirão Preto, São Paulo, Brazil; National Institute of Science and Technology for Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactive Substances (INCT-DATREM), Brazil
| | - Gabriela Meireles
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, 14040-903 Ribeirão Preto, São Paulo, Brazil; Faculty of Sciences, University of Lisbon, Campo Grande, 1749-016 Lisboa, Portugal
| | - Flávia Renata Abe
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, 14040-903 Ribeirão Preto, São Paulo, Brazil; National Institute of Science and Technology for Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactive Substances (INCT-DATREM), Brazil
| | - Carlos Gravato
- Faculty of Sciences, University of Lisbon, Campo Grande, 1749-016 Lisboa, Portugal
| | - Daniel Junqueira Dorta
- National Institute of Science and Technology for Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactive Substances (INCT-DATREM), Brazil; Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Departamento de Química, Universidade de São Paulo, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, São Paulo CEP 14040901, Brazil
| | - Danielle P de Oliveira
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, 14040-903 Ribeirão Preto, São Paulo, Brazil; National Institute of Science and Technology for Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactive Substances (INCT-DATREM), Brazil.
| |
Collapse
|
14
|
Xie J, Goodbourn P, Sztal T, Jusuf PR. Neural Endophenotype Assessment in Zebrafish Larvae Using Optomotor and ZebraBox Locomotion Assessment. Methods Mol Biol 2024; 2746:213-224. [PMID: 38070092 DOI: 10.1007/978-1-0716-3585-8_17] [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] [Indexed: 12/18/2023]
Abstract
Due to the highly conserved genetics across the central nervous system, the easily probed visual system can act as an endophenotype for assessing neurological function. Here, we describe a psychophysics approach to assess visually driven swimming behavior in the high-throughput zebrafish genetic model system. We use the optomotor response test together with general locomotion behavior to assess neural processing while excluding motor defects related to muscle function.
Collapse
Affiliation(s)
- Jiaheng Xie
- School of Biosciences, The University of Melbourne, Parkville, VIC, Australia
| | - Patrick Goodbourn
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Tamar Sztal
- School of Biological Sciences, Monash University, Melbourne, VIC, Australia
| | - Patricia R Jusuf
- School of Biosciences, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
15
|
Binter M, Langer F, Hu X, Lindziute M, Framme C, Tode J, Fuchs H. A simple dissection method for the isolation of mouse trabecular meshwork cells. PLoS One 2023; 18:e0296124. [PMID: 38128042 PMCID: PMC10734917 DOI: 10.1371/journal.pone.0296124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
PURPOSE The outflow pathway, especially trabecular meshwork (TM), plays an essential role in glaucoma, and the availability of TM cells is crucial for in vitro research. So far, the isolation of TM cells from mice has been anything but manageable due to the small size of the eye. Direct isolation using a stereomicroscope and forceps requires a high grade of dexterity. Indirect isolation is based on the phagocytic properties of TM cells and involves injecting magnetic microspheres into the anterior chamber of live mice followed by isolation. Therefore, a simpler, less expensive, and nonexperimental strategy for isolating mouse TM cells would be desirable. METHODS After enucleation, the eyes were cut in half anterior-to-posteriorly. The lens and posterior segment were removed. Iris and the attached ciliary body were gently pulled backward and disconnected from the remaining tissue to expose the TM. By incising through the cornea anteriorly and posteriorly of the TM, the cornea/TM stripe could be isolated. The cornea/TM stripe was cultured with the pigmented side down in a 6-well. The outgrowing pigmented cells were analyzed by immunocytochemistry and mRNA expression for previously described TM cell markers. The phagocytic properties of the cells were additionally confirmed using fluorescent microspheres. RESULTS Pigmented phagocytic cells were the first to grow out of the cornea/TM strips after approximately 4-7 days. Cells were positive for Collagen IV, Fibronectin1, Vimentin, and Actin alpha 2 and could phagocytize fluorescent microbeads. Cross-linked actin networks were visible after 9 days of exposure to TGFB2 (transforming growth factor-beta 2). Additionally, treatment with 500 nM Dexamethasone for one week increased myocilin expression, as previously reported for TM cells. In addition, we proved that this method can also be used in albino mice, which lack pigmentation of the trabecular meshwork. CONCLUSIONS The isolated cells show phagocytic properties and specific expression of markers reported in TM cells. Therefore, our dissection-based method is inexpensive and reproducible for isolating TM cells in mice.
Collapse
Affiliation(s)
- Maximilian Binter
- Institute of Ophthalmology, Hannover Medical School, University Eye Hospital, Hannover, Germany
| | - Fridolin Langer
- Institute of Ophthalmology, Hannover Medical School, University Eye Hospital, Hannover, Germany
| | - Xiaonan Hu
- Institute of Ophthalmology, Hannover Medical School, University Eye Hospital, Hannover, Germany
| | - Migle Lindziute
- Institute of Ophthalmology, Hannover Medical School, University Eye Hospital, Hannover, Germany
| | - Carsten Framme
- Institute of Ophthalmology, Hannover Medical School, University Eye Hospital, Hannover, Germany
| | - Jan Tode
- Institute of Ophthalmology, Hannover Medical School, University Eye Hospital, Hannover, Germany
| | - Heiko Fuchs
- Institute of Ophthalmology, Hannover Medical School, University Eye Hospital, Hannover, Germany
| |
Collapse
|
16
|
Lee HS, Jang S, Eom Y, Kim KT. Comparing Ocular Toxicity of Legacy and Alternative Per- and Polyfluoroalkyl Substances in Zebrafish Larvae. TOXICS 2023; 11:1021. [PMID: 38133422 PMCID: PMC10747198 DOI: 10.3390/toxics11121021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Studies comparing the ocular toxicity potential between legacy and alternative PFAS are lacking. To address this research gap, zebrafish larvae were exposed to both legacy PFAS (i.e., perfluorooctanesulfonic acid [PFOS] and perfluorooctanoic acid [PFOA]) and their corresponding alternatives (i.e., perfluorobutanesulfonic acid [PFBS] and perfluorobutanoic acid [PFBA]). Alterations in their visual behaviors, such as phototactic and optomotor responses (OMR), were assessed at sublethal concentrations. Gene expression variations in visual function-associated pathways were also measured. Visual behavioral assessment revealed that PFOS exposure resulted in concentration-dependent reductions in phototactic responses at 10-1000 μg/L, with PFOA exerting reduction effects only at 100 mg/L. However, their two alternatives had no effect at all tested concentrations. Following an improved contrast-OMR (C-OMR) assessment, PFOS decreased the OMR to a water flow stimulus at 10, 100, and 1000 μg/L. The gene expression analysis revealed that PFOS exposure markedly downregulated most genes involved in the opsins in the photoreceptor and phototransduction cascade, which explains the observed visual behavior changes well. Our findings indicate that PFOS is the most likely PFAS to cause visual toxicity, with PFOA present but less likely, and their substitutes, PFBS and PFBA, cannot be classified as visually toxic to zebrafish.
Collapse
Affiliation(s)
- Han-seul Lee
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Soogyeong Jang
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Youngsub Eom
- Zebrafish Translational Medical Research Center, Korea University, Ansan 15355, Republic of Korea
- Department of Ophthalmology, Korea University Ansan Hospital, Ansan 15355, Republic of Korea
- Department of Ophthalmology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ki-Tae Kim
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
- Department of Environmental Energy Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| |
Collapse
|
17
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
18
|
Gómez Sánchez A, Colucci P, Moran A, Moya López A, Colligris B, Álvarez Y, Kennedy BN. Systemic treatment with cigarette smoke extract affects zebrafish visual behaviour, intraocular vasculature morphology and outer segment phagocytosis. OPEN RESEARCH EUROPE 2023; 3:48. [PMID: 38283058 PMCID: PMC10822043 DOI: 10.12688/openreseurope.15491.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/21/2023] [Indexed: 01/30/2024]
Abstract
Introduction Cigarette smoking adversely affects multiple aspects of human health including eye disorders such as age-related macular degeneration, cataracts and dry eye disease. However, there remains a knowledge gap in how constituents of cigarette smoke affect vision and retinal biology. We used zebrafish to assess effects of short-term acute exposure to cigarette smoke extract (CSE) on visual behaviour and retinal biology. Methods Zebrafish larvae with a developed visual system at three days post-fertilization (dpf) were exposed to CSE for 4, 24 or 48 hours. Visual behaviour, hyaloid vasculature morphology, retinal histology, oxidative stress gene expression and outer segment phagocytosis were investigated using visual behavioural optokinetic and visual motor response assays (OKR and VMR), microscopy (light, fluorescence and transmission electron microscopy), and real-time PCR. Results In zebrafish larvae, 48 hours of CSE treatment resulted in significantly reduced visual behaviour. Larvae treated with 10, 15 or 20 μg/mL CSE showed an average of 13.7, 10.7 or 9.4 saccades per minute, respectively, significantly lower compared with 0.05% DMSO controls (p=0.0093, p=0.0004 and p<0.0001, respectively) that exhibited 19.7 saccades per minute. The diameter of intraocular vessels increased from 4.833 μm in 0.05% DMSO controls to 5.885 μm in the 20 μg/mL CSE-treated larvae (p=0.0333). Biometry analysis highlighted a significant axial length elongation in 20 μg/mL CSE-treated larvae (216.9 μm, p<0.0001) compared to 0.05% dimethyl sulfoxide (DMSO) controls (205.1 μm). Larvae exposed to 20 μg/mL CSE had significantly (p=0.0002) higher numbers of RPE phagosomes compared to vehicle controls (0.1425 and 0.093 phagosomes/μm RPE, respectively). Conclusions Zebrafish larvae with a developed visual system display apparent defects in visual behaviour and retinal biology after acute exposure to CSE, establishing a valuable in vivo model to investigate ocular disorders related to cigarette smoke.
Collapse
Affiliation(s)
- Alicia Gómez Sánchez
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, D04 V1W8, Ireland
- Ocupharm Diagnostic Group Research, Faculty of Optic and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Patrizia Colucci
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, D04 V1W8, Ireland
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Ailis Moran
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, D04 V1W8, Ireland
| | - Alexandro Moya López
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, D04 V1W8, Ireland
- Ocupharm Diagnostic Group Research, Faculty of Optic and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Basilio Colligris
- Ocupharm Diagnostic Group Research, Faculty of Optic and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Yolanda Álvarez
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, D04 V1W8, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, D04 V1W8, Ireland
| | - Breandán N. Kennedy
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, D04 V1W8, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, D04 V1W8, Ireland
| |
Collapse
|
19
|
Castellini ME, Spagnolli G, Poggi L, Biasini E, Casarosa S, Messina A. Identification of the zebrafish homologues of IMPG2, a retinal proteoglycan. Cell Tissue Res 2023; 394:93-105. [PMID: 37470839 PMCID: PMC10558372 DOI: 10.1007/s00441-023-03808-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
Photoreceptor outer segments are surrounded by a carbohydrate-rich matrix, the interphotoreceptor matrix, necessary for physiological retinal function. Few roles for molecules characterizing the interphotoreceptor matrix have been clearly defined. Recent studies have found the presence of nonsense mutations in the interphotoreceptor matrix proteoglycan 2 (IMPG2) gene in patients affected by retinal dystrophies. IMPG2 encodes for a proteoglycan synthesized by photoreceptors and secreted in the interphotoreceptor matrix. Little is known about the structure and function of this protein, we thus decided to characterize zebrafish impg2. In zebrafish there are two Impg2 proteins, Impg2a and Impg2b. We generated a phylogenetic tree based on IMPG2 protein sequence similarity among vertebrates, showing a significant similarity between humans and teleosts. The human and zebrafish proteins share conserved domains, as also shown by homology models. Expression analyses of impg2a and impg2b show a continued expression in the photoreceptor layer starting from developmental stages and continuing through adulthood. Between 1 and 6 months post-fertilization, there is a significant shift of Impg2 expression toward the outer segment region, suggesting an increase in secretion. This raises intriguing hypotheses about its possible role(s) during retinal maturation, laying the groundwork for the generation of most needed models for the study of IMPG2-related inherited retinal dystrophies.
Collapse
Affiliation(s)
- M E Castellini
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive, 9, 38123, Povo, TN, Italy
| | - G Spagnolli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive, 9, 38123, Povo, TN, Italy
- Sibylla Biotech S.R.L, Piazzetta Chiavica 2 - 37121, Verona, VR, Italy
| | - L Poggi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive, 9, 38123, Povo, TN, Italy
- Centre for Medical Sciences (CISMed), University of Trento, Via S. Maria Maddalena, 1, 38122, Trento, TN, Italy
| | - E Biasini
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive, 9, 38123, Povo, TN, Italy
- Centre for Medical Sciences (CISMed), University of Trento, Via S. Maria Maddalena, 1, 38122, Trento, TN, Italy
| | - S Casarosa
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive, 9, 38123, Povo, TN, Italy.
- Centre for Medical Sciences (CISMed), University of Trento, Via S. Maria Maddalena, 1, 38122, Trento, TN, Italy.
| | - A Messina
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive, 9, 38123, Povo, TN, Italy
- Centre for Mind/Brain Sciences (CIMeC), University of Trento, Piazza Manifattura 1, 38068, Rovereto, TN, Italy
| |
Collapse
|
20
|
Shrestha AP, Rameshkumar N, Boff JM, Rajmanna R, Chandrasegaran T, Frederick CE, Zenisek D, Vaithianathan T. The Effects of Aging on Rod Bipolar Cell Ribbon Synapses. Cells 2023; 12:2385. [PMID: 37830599 PMCID: PMC10572008 DOI: 10.3390/cells12192385] [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: 08/20/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
The global health concern posed by age-related visual impairment highlights the need for further research focused on the visual changes that occur during the process of aging. To date, multiple sensory alterations related to aging have been identified, including morphological and functional changes in inner hair cochlear cells, photoreceptors, and retinal ganglion cells. While some age-related morphological changes are known to occur in rod bipolar cells in the retina, their effects on these cells and on their connection to other cells via ribbon synapses remain elusive. To investigate the effects of aging on rod bipolar cells and their ribbon synapses, we compared synaptic calcium currents, calcium dynamics, and exocytosis in zebrafish (Danio rerio) that were middle-aged (MA,18 months) or old-aged (OA, 36 months). The bipolar cell terminal in OA zebrafish exhibited a two-fold reduction in number of synaptic ribbons, an increased ribbon length, and a decrease in local Ca2+ signals at the tested ribbon location, with little change in the overall magnitude of the calcium current or exocytosis in response to brief pulses. Staining of the synaptic ribbons with antibodies specific for PKCa revealed shortening of the inner nuclear and plexiform layers (INL and IPL). These findings shed light on age-related changes in the retina that are related to synaptic ribbons and calcium signals.
Collapse
Affiliation(s)
- Abhishek P. Shrestha
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Nirujan Rameshkumar
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johane M. Boff
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Rhea Rajmanna
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | - Courtney E. Frederick
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA (D.Z.)
| | - David Zenisek
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA (D.Z.)
| | - Thirumalini Vaithianathan
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Ophthalmology, Hamilton Eye Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
21
|
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: 4.0] [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
|
22
|
Huang W, Wu T, Wu R, Peng J, Zhang Q, Shi X, Wu K. Fish to learn: insights into the effects of environmental chemicals on eye development and visual function in zebrafish. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023:10.1007/s11356-023-27629-3. [PMID: 37195602 DOI: 10.1007/s11356-023-27629-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/10/2023] [Indexed: 05/18/2023]
Abstract
Vision is the most essential sense system for the human being. Congenital visual impairment affects millions of people globally. It is increasingly realized that visual system development is an impressionable target of environmental chemicals. However, due to inaccessibility and ethical issues, the use of humans and other placental mammals is constrained, which limits our better understanding of environmental factors on ocular development and visual function in the embryonic stage. Therefore, as complementing laboratory rodents, zebrafish has been the most frequently employed to understand the effects of environmental chemicals on eye development and visual function. One of the major reasons for the increasing use of zebrafish is their polychromatic vision. Zebrafish retinas are morphologically and functionally analogous to those of mammalian, as well as evolutionary conservation among vertebrate eye. This review provides an update on harmful effects from exposure to environmental chemicals, involving metallic elements (ions), metal-derived nanoparticles, microplastics, nanoplastics, persistent organic pollutants, pesticides, and pharmaceutical pollutants on the eye development and visual function in zebrafish embryos. The collected data provide a comprehensive understanding of environmental factors on ocular development and visual function. This report highlights that zebrafish is promising as a model to identify hazardous toxicants toward eye development and is hopeful for developing preventative or postnatal therapies for human congenital visual impairment.
Collapse
Affiliation(s)
- Wenlong Huang
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Department of Preventive Medicine, Shantou University Medical College, Xinling Rd., No. 22, Shantou, 515041, Guangdong, China
- Department of Forensic Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Tianjie Wu
- Department of Anaesthesiology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen University, Shantou, 515041, Guangdong, China
| | - Ruotong Wu
- School of Life Science, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jiajun Peng
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Department of Preventive Medicine, Shantou University Medical College, Xinling Rd., No. 22, Shantou, 515041, Guangdong, China
| | - Qiong Zhang
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Department of Preventive Medicine, Shantou University Medical College, Xinling Rd., No. 22, Shantou, 515041, Guangdong, China
| | - Xiaoling Shi
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Department of Preventive Medicine, Shantou University Medical College, Xinling Rd., No. 22, Shantou, 515041, Guangdong, China
| | - Kusheng Wu
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Department of Preventive Medicine, Shantou University Medical College, Xinling Rd., No. 22, Shantou, 515041, Guangdong, China.
| |
Collapse
|
23
|
Zhang J, Jing M, Li P, Sun L, Pi X, Jiang N, Zhu KK, Li H, Li J, Wang M, Zhang J, Liu M, Mu H, Hu Y, Cui X. Knockout of DLIC1 leads to retinal cone degeneration via disturbing Rab8 transport in zebrafish. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166645. [PMID: 36682603 DOI: 10.1016/j.bbadis.2023.166645] [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: 11/14/2022] [Revised: 12/28/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023]
Abstract
Retinal photoreceptors execute phototransduction functions and require an efficient system for the transport of materials (e.g. proteins and lipids) from inner segments to outer segments. Cytoplasmic dynein 1 is a minus-end-directed microtubule motor and participates in cargo transport in the cytoplasm. However, the roles of dynein 1 motor in photoreceptor cargo transport and retinal development are still ambiguous. In our present study, the light intermediate chain protein DLIC1 (encoded by dync1li1), links activating adaptors to bind diverse cargos in the dynein 1 motor, was depleted using CRISPR-Cas9 technology in zebrafish. The dync1li1-/- zebrafish displayed progressive degeneration of retinal cone photoreceptors, especially blue cones. The retinal rods were not affected in dync1li1-/- zebrafish. Knockout of DLIC1 resulted in abnormal expression and localization of cone opsins in dync1li1-/- retinas. TUNEL staining suggested that apoptosis was induced after aberrant accumulation of cone opsins in photoreceptors of dync1li1-/- zebrafish. Instead of Rab11 transport, Rab8 transport was disturbed in dync1li1-/- retinas. Our data demonstrate that DLIC1 is required for function maintenance and survival of cone photoreceptors, and hint at an essential role of the cytoplasmic dynein 1 motor in photoreceptor cargo transport.
Collapse
Affiliation(s)
- Jing Zhang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Min Jing
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Ping Li
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Luqian Sun
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Xiahui Pi
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Ning Jiang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Ke-Ke Zhu
- Kaifeng Key Lab of Myopia and Cataract, Kaifeng Central Hospital, Kaifeng, China
| | - Hui Li
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Jing Li
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Mingli Wang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Jun Zhang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Mugen Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmei Mu
- Kaifeng Key Lab of Myopia and Cataract, Kaifeng Central Hospital, Kaifeng, China.
| | - Yanzhong Hu
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China; Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Xiukun Cui
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China.
| |
Collapse
|
24
|
Quint WH, van Buuren R, Kokke NCCJ, Meester-Smoor MA, Willemsen R, Broersma R, Iglesias AI, Lucassen M, Klaver CCW. Exposure to cyan or red light inhibits the axial growth of zebrafish eyes. Exp Eye Res 2023; 230:109437. [PMID: 36924981 DOI: 10.1016/j.exer.2023.109437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023]
Abstract
Myopia, or nearsightedness, is the most common type of refractive error and is characterized by a mismatch between the optical power and ocular axial length. Light, and more specifically the spectral composition of light, has been known to influence myopic axial growth. In this pilot study, we exposed zebrafish to illuminations that vary in spectral composition and screened for changes in axial length. The illumination spectra included narrow band ultra-violet A (UVA) (peak wavelength 369 nm), violet (425 nm), cyan (483 nm), green/yellow (557 nm), and red (633 nm) light, as well as broad band white light (2700 K and 6500 K), dim white light and broad spectrum (day) light. We found that rearing zebrafish in cyan or red light leads to a reduction of the ocular axial length. The results of this pilot study may contribute to new perspectives on the role of light and lighting as an intervention strategy for myopia control.
Collapse
Affiliation(s)
- Wim H Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Renee van Buuren
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Nina C C J Kokke
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Magda A Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Rémy Broersma
- Signify Research, Signify, Eindhoven, the Netherlands
| | - Adriana I Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands; Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.
| |
Collapse
|
25
|
Wu KY, Kulbay M, Toameh D, Xu AQ, Kalevar A, Tran SD. Retinitis Pigmentosa: Novel Therapeutic Targets and Drug Development. Pharmaceutics 2023; 15:685. [PMID: 36840007 PMCID: PMC9963330 DOI: 10.3390/pharmaceutics15020685] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023] Open
Abstract
Retinitis pigmentosa (RP) is a heterogeneous group of hereditary diseases characterized by progressive degeneration of retinal photoreceptors leading to progressive visual decline. It is the most common type of inherited retinal dystrophy and has a high burden on both patients and society. This condition causes gradual loss of vision, with its typical manifestations including nyctalopia, concentric visual field loss, and ultimately bilateral central vision loss. It is one of the leading causes of visual disability and blindness in people under 60 years old and affects over 1.5 million people worldwide. There is currently no curative treatment for people with RP, and only a small group of patients with confirmed RPE65 mutations are eligible to receive the only gene therapy on the market: voretigene neparvovec. The current therapeutic armamentarium is limited to retinoids, vitamin A supplements, protection from sunlight, visual aids, and medical and surgical interventions to treat ophthalmic comorbidities, which only aim to slow down the progression of the disease. Considering such a limited therapeutic landscape, there is an urgent need for developing new and individualized therapeutic modalities targeting retinal degeneration. Although the heterogeneity of gene mutations involved in RP makes its target treatment development difficult, recent fundamental studies showed promising progress in elucidation of the photoreceptor degeneration mechanism. The discovery of novel molecule therapeutics that can selectively target specific receptors or specific pathways will serve as a solid foundation for advanced drug development. This article is a review of recent progress in novel treatment of RP focusing on preclinical stage fundamental research on molecular targets, which will serve as a starting point for advanced drug development. We will review the alterations in the molecular pathways involved in the development of RP, mainly those regarding endoplasmic reticulum (ER) stress and apoptotic pathways, maintenance of the redox balance, and genomic stability. We will then discuss the therapeutic approaches under development, such as gene and cell therapy, as well as the recent literature identifying novel potential drug targets for RP.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Division of Ophthalmology, Department of Surgery, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Merve Kulbay
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Dana Toameh
- Faculty of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| | - An Qi Xu
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Ananda Kalevar
- Division of Ophthalmology, Department of Surgery, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
26
|
Wei S, Qiu L, Ru S, Yang Y, Wang J, Zhang X. Bisphenol S disrupts opsins gene expression and impairs the light-sensing function via antagonizing TH-TRβ signaling pathway in zebrafish larvae. Food Chem Toxicol 2023; 172:113588. [PMID: 36574878 DOI: 10.1016/j.fct.2022.113588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/15/2022] [Accepted: 12/23/2022] [Indexed: 12/25/2022]
Abstract
Bisphenol S (BPS) is extensively used in "bisphenol A-free" products such as baby bottles. Although the visual toxicity of BPS has been reported, the underlying mechanism was largely unknown. In the present study, zebrafish were exposed to 0, 4 and 400 nM BPS from 2 h post-fertilization (hpf) to 120 hpf to further explore the thyroid disruption mechanism underlying the BPS induced impairment of visual function. The results showed that BPS decreased T3 levels in larval eyes, induced retinal expression of thyroid hormone receptor β (TRβ), and thereby down-regulated the expression of TH-mediated opsin genes (opn1lw1, opn1lw2, opn1mw1, opn1mw2, opn1mw3, and opn1sw2) and impaired subsequent phototransduction pathways, leading to decreased visually mediated phototactic response and body color adaptation but stimulated visual motor response (VMR). Combining exposure of exogenous T3 or 1-850 (antagonist for TRβ) with BPS could partly compensate the inhibited expression of opsin genes (opn1mw2, opn1lw1, and opn1lw2) and alleviate the hyperactivity of larval VMR caused by BPS alone, suggesting that BPS disrupted the opsins expression and also light-sensing function via antagonizing TH-TRβ signaling pathway. This study underlined the importance of TH signaling in regulating the proper vision and proposed a novel mechanism for the visual toxicity of BPS.
Collapse
Affiliation(s)
- Shuhui Wei
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao, 266003, Shandong province, PR China
| | - Liguo Qiu
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao, 266003, Shandong province, PR China
| | - Shaoguo Ru
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao, 266003, Shandong province, PR China
| | - Yang Yang
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao, 266003, Shandong province, PR China
| | - Jun Wang
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao, 266003, Shandong province, PR China
| | - Xiaona Zhang
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao, 266003, Shandong province, PR China.
| |
Collapse
|
27
|
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
|
28
|
Bise T, Pfefferli C, Bonvin M, Taylor L, Lischer HEL, Bruggmann R, Jaźwińska A. The regeneration-responsive element careg monitors activation of Müller glia after MNU-induced damage of photoreceptors in the zebrafish retina. Front Mol Neurosci 2023; 16:1160707. [PMID: 37138703 PMCID: PMC10149768 DOI: 10.3389/fnmol.2023.1160707] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
In contrast to mammals, zebrafish can regenerate their damaged photoreceptors. This capacity depends on the intrinsic plasticity of Müller glia (MG). Here, we identified that the transgenic reporter careg, a marker of regenerating fin and heart, also participates in retina restoration in zebrafish. After methylnitrosourea (MNU) treatment, the retina became deteriorated and contained damaged cell types including rods, UV-sensitive cones and the outer plexiform layer. This phenotype was associated with the induction of careg expression in a subset of MG until the reconstruction of the photoreceptor synaptic layer. Single-cell RNA sequencing (scRNAseq) analysis of regenerating retinas revealed a population of immature rods, defined by high expression of rhodopsin and the ciliogenesis gene meig1, but low expression of phototransduction genes. Furthermore, cones displayed deregulation of metabolic and visual perception genes in response to retina injury. Comparison between careg:EGFP expressing and non-expressing MG demonstrated that these two subpopulations are characterized by distinct molecular signatures, suggesting their heterogenous responsiveness to the regenerative program. Dynamics of ribosomal protein S6 phosphorylation showed that TOR signaling became progressively switched from MG to progenitors. Inhibition of TOR with rapamycin reduced the cell cycle activity, but neither affected careg:EGFP expression in MG, nor prevented restoration of the retina structure. This indicates that MG reprogramming, and progenitor cell proliferation might be regulated by distinct mechanisms. In conclusion, the careg reporter detects activated MG, and provides a common marker of regeneration-competent cells in diverse zebrafish organs, including the retina.
Collapse
Affiliation(s)
- Thomas Bise
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Marylène Bonvin
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Lea Taylor
- Interfaculty Bioinformatics Unit, University of Bern, Bern, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Heidi E. L. Lischer
- Interfaculty Bioinformatics Unit, University of Bern, Bern, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit, University of Bern, Bern, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- *Correspondence: Anna Jaźwińska,
| |
Collapse
|
29
|
Lee Y, Yang J. Frozen Block Tissue Staining for Eye Structure of Zebrafish Embryo. Methods Mol Biol 2023; 2678:191-198. [PMID: 37326715 DOI: 10.1007/978-1-0716-3255-0_15] [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] [Indexed: 06/17/2023]
Abstract
Histology is a technique used to study the morphology of cell structures by cutting samples into thin sections. Histological cross-section and staining are the techniques needed to visualize the morphology of cell tissues. A suitable tissue staining experiment was created to observe changes in the retinal layer in zebrafish embryos. Zebrafish have a human-like visual system, retina, and eye structures. Due to the small size of zebrafish and undeveloped bones in the embryonic stage, the resistance through cross-section is inevitably small. Here, we present optimized protocol changes in eye tissue of zebrafish using frozen blocks.
Collapse
Affiliation(s)
- Yujin Lee
- Department of Ophthalmology, Inje University Busan Paik Hospital, Busan, South Korea
| | - Jaewook Yang
- Department of Ophthalmology, Inje University Busan Paik Hospital, Busan, South Korea.
| |
Collapse
|
30
|
Tulp1 deficiency causes early-onset retinal degeneration through affecting ciliogenesis and activating ferroptosis in zebrafish. Cell Death Dis 2022; 13:962. [PMID: 36396940 PMCID: PMC9672332 DOI: 10.1038/s41419-022-05372-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/22/2022] [Accepted: 10/24/2022] [Indexed: 11/19/2022]
Abstract
Mutations in TUB-like protein 1 (TULP1) are associated with severe early-onset retinal degeneration in humans. However, the pathogenesis remains largely unknown. There are two homologous genes of TULP1 in zebrafish, namely tulp1a and tulp1b. Here, we generated the single knockout (tulp1a-/- and tulp1b-/-) and double knockout (tulp1-dKO) models in zebrafish. Knockout of tulp1a resulted in the mislocalization of UV cone opsins and the degeneration of UV cones specifically, while knockout of tulp1b resulted in mislocalization of rod opsins and rod-cone degeneration. In the tulp1-dKO zebrafish, mislocalization of opsins was present in all types of photoreceptors, and severe degeneration was observed at a very early age, mimicking the clinical manifestations of TULP1 patients. Photoreceptor cilium length was significantly reduced in the tulp1-dKO retinas. RNA-seq analysis showed that the expression of tektin2 (tekt2), a ciliary and flagellar microtubule structural component, was downregulated in the tulp1-dKO zebrafish. Dual-luciferase reporter assay suggested that Tulp1a and Tulp1b transcriptionally activate the promoter of tekt2. In addition, ferroptosis might be activated in the tulp1-dKO zebrafish, as suggested by the up-regulation of genes related to the ferroptosis pathway, the shrinkage of mitochondria, reduction or disappearance of mitochondria cristae, and the iron and lipid droplet deposition in the retina of tulp1-dKO zebrafish. In conclusion, our study establishes an appropriate zebrafish model for TULP1-associated retinal degeneration and proposes that loss of TULP1 causes defects in cilia structure and opsin trafficking through the downregulation of tekt2, which further increases the death of photoreceptors via ferroptosis. These findings offer insight into the pathogenesis and clinical treatment of early-onset retinal degeneration.
Collapse
|
31
|
Gölz L, Baumann L, Pannetier P, Braunbeck T, Knapen D, Vergauwen L. AOP Report: Thyroperoxidase Inhibition Leading to Altered Visual Function in Fish Via Altered Retinal Layer Structure. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2022; 41:2632-2648. [PMID: 35942927 DOI: 10.1002/etc.5452] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Thyroid hormones (THs) are involved in the regulation of many important physiological and developmental processes, including vertebrate eye development. Thyroid hormone system-disrupting chemicals (THSDCs) may have severe consequences, because proper functioning of the visual system is a key factor for survival in wildlife. However, the sequence of events leading from TH system disruption (THSD) to altered eye development in fish has not yet been fully described. The development of this adverse outcome pathway (AOP) was based on an intensive literature review of studies that focused on THSD and impacts on eye development, mainly in fish. In total, approximately 120 studies (up to the end of 2021) were used in the development of this AOP linking inhibition of the key enzyme for TH synthesis, thyroperoxidase (TPO), to effects on retinal layer structure and visual function in fish (AOP-Wiki, AOP 363). In a weight-of-evidence evaluation, the confidence levels were overall moderate, with ample studies showing the link between reduced TH levels and altered retinal layer structure. However, some uncertainties about the underlying mechanism(s) remain. Although the current weight-of-evidence evaluation is based on fish, the AOP is plausibly applicable to other vertebrate classes. Through the re-use of several building blocks, this AOP is connected to the AOPs leading from TPO and deiodinase inhibition to impaired swim bladder inflation in fish (AOPs 155-159), together forming an AOP network describing THSD in fish. This AOP network addresses the lack of thyroid-related endpoints in existing fish test guidelines for the evaluation of THSDCs. Environ Toxicol Chem 2022;41:2632-2648. © 2022 SETAC.
Collapse
Affiliation(s)
- Lisa Gölz
- Aquatic Ecology and Toxicology Research Group, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Lisa Baumann
- Aquatic Ecology and Toxicology Research Group, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Pauline Pannetier
- Aquatic Ecology and Toxicology Research Group, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology Research Group, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Dries Knapen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Lucia Vergauwen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
32
|
Shrestha AP, Saravanakumar A, Konadu B, Madireddy S, Gibert Y, Vaithianathan T. Embryonic Hyperglycemia Delays the Development of Retinal Synapses in a Zebrafish Model. Int J Mol Sci 2022; 23:ijms23179693. [PMID: 36077087 PMCID: PMC9456524 DOI: 10.3390/ijms23179693] [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: 07/08/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022] Open
Abstract
Embryonic hyperglycemia negatively impacts retinal development, leading to abnormal visual behavior, altered timing of retinal progenitor differentiation, decreased numbers of retinal ganglion cells and Müller glia, and vascular leakage. Because synaptic disorganization is a prominent feature of many neurological diseases, the goal of the current work was to study the potential impact of hyperglycemia on retinal ribbon synapses during embryonic development. Our approach utilized reverse transcription quantitative PCR (RT-qPCR) and immunofluorescence labeling to compare the transcription of synaptic proteins and their localization in hyperglycemic zebrafish embryos, respectively. Our data revealed that the maturity of synaptic ribbons was compromised in hyperglycemic zebrafish larvae, where altered ribeye expression coincided with the delay in establishing retinal ribbon synapses and an increase in the immature synaptic ribbons. Our results suggested that embryonic hyperglycemia disrupts retinal synapses by altering the development of the synaptic ribbon, which can lead to visual defects. Future studies using zebrafish models of hyperglycemia will allow us to study the underlying mechanisms of retinal synapse development.
Collapse
Affiliation(s)
- Abhishek P. Shrestha
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ambalavanan Saravanakumar
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Program in Biology, Rhodes College, Memphis, TN 38112, USA
| | - Bridget Konadu
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Saivikram Madireddy
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yann Gibert
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Thirumalini Vaithianathan
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Correspondence: ; Tel.: +1-901-448-2786
| |
Collapse
|
33
|
Liu CF, Ou-Yang Y, Huang CY, Jao SW, Kuo YK, Chen HC, Cheng SC, Wang NK, Chuang LH, Chen YH, Chen WY. Zebrafish (Danio rerio) Is an Economical and Efficient Animal Model for Screening Potential Anti-cataract Compounds. Transl Vis Sci Technol 2022; 11:21. [PMID: 35994266 PMCID: PMC9419454 DOI: 10.1167/tvst.11.8.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/28/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose To develop a zebrafish cataract model for screening potential anti-cataract compounds. Methods Living zebrafish were anesthetized and exposed to ultraviolet-C (UV-C) irradiation at a dosage of 3250 mJ/cm2/d until they developed severe cataracts. These cataracts were graded based on photographs analyzed with ImageQuant TL version 7.0. Fish with severe cataracts were used to evaluate a range of compounds for cataract treatment, including the previously demonstrated hit compound lanosterol. For the initial evaluation, fish were divided into four groups: no treatment, balanced salt solution, β-cyclodextrin (β-CD), and lanosterol dissolved in β-CD. The treatments were performed for 10 days, and the clarity of lenses was evaluated. To assess the persistence of treatment, fish were treated with β-CD and lanosterol dissolved in β-CD for seven consecutive days followed by monitoring for three days without treatment. Results The average time for zebrafish to develop severe cataracts using the present UV-C irradiation protocol was 7.8 days (range 4-15 days). Both study designs required only another 10 days to determine the effect of hit compounds. The total experimental period could be completed within one month, and the entire experiment was economical. Conclusions We could assay a large number of hit compounds at a reasonable cost and within a short time using this newly developed zebrafish cataract model. These assays may allow development of an efficient platform for screening potential anti-cataract compounds. Translational Relevance The results may facilitate the development of ani-cataract medication for humans after further experiments and investigations.
Collapse
Affiliation(s)
- Chun-Fu Liu
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Department of Medicine, College of Medicine Chang Gung University, Taoyuan, Taiwan
- Program in Molecular Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen Ou-Yang
- Department of Chemistry, Tamkang University, Tamsui, New Taipei City, Taiwan
| | - Ching-Ying Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Wei Jao
- Department of Chemistry, Tamkang University, Tamsui, New Taipei City, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Kai Kuo
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Department of Medicine, College of Medicine Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chi Chen
- Department of Medicine, College of Medicine Chang Gung University, Taoyuan, Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taiwan
- Center for Tissue Engineering, Chang Memorial Hospital, Linkou, Taiwan
| | - Shu-Chun Cheng
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Nan-Kai Wang
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, New York, NY, USA
| | - Lan-Hsin Chuang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Department of Medicine, College of Medicine Chang Gung University, Taoyuan, Taiwan
| | - Yau-Hung Chen
- Department of Chemistry, Tamkang University, Tamsui, New Taipei City, Taiwan
| | - Wei-Yi Chen
- Program in Molecular Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
34
|
Catalani E, Cherubini A, Del Quondam S, Cervia D. Regenerative Strategies for Retinal Neurons: Novel Insights in Non-Mammalian Model Organisms. Int J Mol Sci 2022; 23:ijms23158180. [PMID: 35897754 PMCID: PMC9331597 DOI: 10.3390/ijms23158180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
A detailed knowledge of the status of the retina in neurodegenerative conditions is a crucial point for the development of therapeutics in retinal pathologies and to translate eye research to CNS disease. In this context, manipulating signaling pathways that lead to neuronal regeneration offers an excellent opportunity to substitute damaged cells and, thus, restore the tissue functionality. Alternative systems and methods are increasingly being considered to replace/reduce in vivo approaches in the study of retina pathophysiology. Herein, we present recent data obtained from the zebrafish (Danio rerio) and the fruit fly Drosophila melanogaster that bring promising advantages into studying and modeling, at a preclinical level, neurodegeneration and regenerative approaches in retinal diseases. Indeed, the regenerative ability of vertebrate model zebrafish is particularly appealing. In addition, the fruit fly is ideal for regenerative studies due to its high degree of conservation with vertebrates and the broad spectrum of genetic variants achievable. Furthermore, a large part of the drosophila brain is dedicated to sight, thus offering the possibility of studying common mechanisms of the visual system and the brain at once. The knowledge acquired from these alternative models may help to investigate specific well-conserved factors of interest in human neuroregeneration after injuries or during pathologies.
Collapse
|
35
|
Connor B, Titialii-Torres K, Rockenhaus AE, Passamonte S, Morris AC, Lee YS. Biliverdin regulates NR2E3 and zebrafish retinal photoreceptor development. Sci Rep 2022; 12:7310. [PMID: 35508617 PMCID: PMC9068610 DOI: 10.1038/s41598-022-11502-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
NR2E3 is an orphan nuclear receptor whose loss-of-function causes abnormal retinal photoreceptor development and degeneration. However, despite that many nuclear receptors are regulated by binding of small molecule ligands, biological small molecule ligands regulating NR2E3 have not been identified. Identification of an endogenous NR2E3 ligand might reveal a previously unrecognized component contributing to retinal development and maintenance. Here we report that biliverdin, a conserved green pigment from heme catabolism, regulates NR2E3 and is necessary for zebrafish retinal photoreceptor development. Biliverdin from retinal extracts specifically bound to NR2E3’s ligand-binding domain and induced NR2E3-dependent reporter gene expression. Inhibition of biliverdin synthesis decreased photoreceptor cell populations in zebrafish larvae, and this phenotype was alleviated by exogenously supplied biliverdin. Thus, biliverdin is an endogenous small molecule ligand for NR2E3 and a component necessary for the proper development of photoreceptor cells. This result suggests a possible role of heme metabolism in the regulation of retinal photoreceptor cell development.
Collapse
Affiliation(s)
- Blaine Connor
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | | | - Abigail E Rockenhaus
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Samuel Passamonte
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Ann C Morris
- Department of Biology, University of Kentucky, Lexington, KY, 40506, USA
| | - Young-Sam Lee
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40536, USA.
| |
Collapse
|
36
|
Maleski ALA, Rosa JGS, Bernardo JTG, Astray RM, Walker CIB, Lopes-Ferreira M, Lima C. Recapitulation of Retinal Damage in Zebrafish Larvae Infected with Zika Virus. Cells 2022; 11:1457. [PMID: 35563763 PMCID: PMC9100881 DOI: 10.3390/cells11091457] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/24/2022] [Accepted: 04/07/2022] [Indexed: 02/06/2023] Open
Abstract
Zebrafish are increasingly being utilized as a model to investigate infectious diseases and to advance the understanding of pathogen-host interactions. Here, we take advantage of the zebrafish to recapitulate congenital ZIKV infection and, for the first time, demonstrate that it can be used to model infection and reinfection and monitor anti-viral and inflammatory immune responses, as well as brain growth and eye abnormalities during embryonic development. By injecting a Brazilian strain of ZIKV into the yolk sac of one-cell stage embryos, we confirmed that, after 72 h, ZIKV successfully infected larvae, and the physical condition of the virus-infected hosts included gross morphological changes in surviving embryos (84%), with a reduction in larval head size and retinal damage characterized by increased thickness of the lens and inner nuclear layer. Changes in locomotor activity and the inability to perceive visual stimuli are a result of changes in retinal morphology caused by ZIKV. Furthermore, we demonstrated the ability of ZIKV to replicate in zebrafish larvae and infect new healthy larvae, impairing their visual and neurological functions. These data reinforce the deleterious activity of ZIKV in the brain and visual structures and establish the zebrafish as a model to study the molecular mechanisms involved in the pathology of the virus.
Collapse
Affiliation(s)
- Adolfo Luis Almeida Maleski
- Immunoregulation Unit of the Laboratory of Applied Toxinology (CeTICs/FAPESP), Butantan Institute, São Paulo 05503-900, Brazil; (A.L.A.M.); (J.G.S.R.); (J.T.G.B.); (M.L.-F.)
- Laboratory of Neuropharmacological Studies (LABEN), Post-Graduation Program of Pharmaceutical Science, Federal University of Sergipe, São Paulo 05503-009, Brazil;
| | - Joao Gabriel Santos Rosa
- Immunoregulation Unit of the Laboratory of Applied Toxinology (CeTICs/FAPESP), Butantan Institute, São Paulo 05503-900, Brazil; (A.L.A.M.); (J.G.S.R.); (J.T.G.B.); (M.L.-F.)
| | - Jefferson Thiago Gonçalves Bernardo
- Immunoregulation Unit of the Laboratory of Applied Toxinology (CeTICs/FAPESP), Butantan Institute, São Paulo 05503-900, Brazil; (A.L.A.M.); (J.G.S.R.); (J.T.G.B.); (M.L.-F.)
| | | | - Cristiani Isabel Banderó Walker
- Laboratory of Neuropharmacological Studies (LABEN), Post-Graduation Program of Pharmaceutical Science, Federal University of Sergipe, São Paulo 05503-009, Brazil;
| | - Monica Lopes-Ferreira
- Immunoregulation Unit of the Laboratory of Applied Toxinology (CeTICs/FAPESP), Butantan Institute, São Paulo 05503-900, Brazil; (A.L.A.M.); (J.G.S.R.); (J.T.G.B.); (M.L.-F.)
| | - Carla Lima
- Immunoregulation Unit of the Laboratory of Applied Toxinology (CeTICs/FAPESP), Butantan Institute, São Paulo 05503-900, Brazil; (A.L.A.M.); (J.G.S.R.); (J.T.G.B.); (M.L.-F.)
| |
Collapse
|
37
|
Quint WH, Tadema KCD, Crins JHC, Kokke NCCJ, Meester-Smoor MA, Willemsen R, Klaver CCW, Iglesias AI. Zebrafish: An In Vivo Screening Model to Study Ocular Phenotypes. Transl Vis Sci Technol 2022; 11:17. [PMID: 35285860 PMCID: PMC8934544 DOI: 10.1167/tvst.11.3.17] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To establish a set of assays that allow the in vivo screening of candidate genes for ocular diseases in zebrafish, with an emphasis on refractive error. Methods Our pipeline includes the most relevant ocular screening measurements to assess (1) ocular biometry using spectral domain optical coherence tomography, (2) refractive status using an eccentric photorefractor, (3) intraocular pressure by tonometry, and (4) optokinetic response to study visual capability in zebrafish. To validate our pipeline and to demonstrate the potential of zebrafish as a valid animal model, we chose two well-characterized genes with an ocular phenotype (PRSS56 and FBN1) and generated two mutant zebrafish lines (prss56 and fbn1). Mutant fish were assessed at 2, 4, and 6 months after fertilization. Results With the proposed phenotyping pipeline, we showed that ocular biometry, refractive status, intraocular pressure, and visual function can be studied in zebrafish. In the prss56 mutant, the pipeline revealed a dramatic decrease in axial length, mainly owing to a decreased vitreous chamber depth, whereas in the fbn1 mutant, ectopia lentis was the most distinctive ocular phenotype observed. Tonometry in both mutant lines showed an increase in intraocular pressure. Conclusions The proposed pipeline was applied successfully in zebrafish and can be used for future genetic screenings of candidate genes. While validating our pipeline, we found a close resemblance between the ocular manifestations in the zebrafish mutants and patients harboring mutations in PRSS56 and FBN1. Our results support the validity of our pipeline and highlight the potential of zebrafish as an animal model for in vivo screening of candidate genes for ocular diseases.
Collapse
Affiliation(s)
- Wim H Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Kirke C D Tadema
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Johan H C Crins
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Nina C C J Kokke
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Magda A Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands.,Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Adriana I Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
38
|
van der Sande E, Haarman AEG, Quint WH, Tadema KCD, Meester-Smoor MA, Kamermans M, De Zeeuw CI, Klaver CCW, Winkelman BHJ, Iglesias AI. The Role of GJD2(Cx36) in Refractive Error Development. Invest Ophthalmol Vis Sci 2022; 63:5. [PMID: 35262731 PMCID: PMC8934558 DOI: 10.1167/iovs.63.3.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Refractive errors are common eye disorders characterized by a mismatch between the focal power of the eye and its axial length. An increased axial length is a common cause of the refractive error myopia (nearsightedness). The substantial increase in myopia prevalence over the last decades has raised public health concerns because myopia can lead to severe ocular complications later in life. Genomewide association studies (GWAS) have made considerable contributions to the understanding of the genetic architecture of refractive errors. Among the hundreds of genetic variants identified, common variants near the gap junction delta-2 (GJD2) gene have consistently been reported as one of the top hits. GJD2 encodes the connexin 36 (Cx36) protein, which forms gap junction channels and is highly expressed in the neural retina. In this review, we provide current evidence that links GJD2(Cx36) to the development of myopia. We summarize the gap junctional communication in the eye and the specific role of GJD2(Cx36) in retinal processing of visual signals. Finally, we discuss the pathways involving dopamine and gap junction phosphorylation and coupling as potential mechanisms that may explain the role of GJD2(Cx36) in refractive error development.
Collapse
Affiliation(s)
- Emilie van der Sande
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
| | - Annechien E. G. Haarman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wim H. Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kirke C. D. Tadema
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Magda A. Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maarten Kamermans
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
- Department of Biomedical Physics and Biomedical Photonics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Chris I. De Zeeuw
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline C. W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Beerend H. J. Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adriana I. Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
39
|
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: 21] [Impact Index Per Article: 10.5] [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
|
40
|
Liu Y, Wang Y, Li N, Jiang S. Avobenzone and nanoplastics affect the development of zebrafish nervous system and retinal system and inhibit their locomotor behavior. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 806:150681. [PMID: 34599957 DOI: 10.1016/j.scitotenv.2021.150681] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/08/2021] [Accepted: 09/26/2021] [Indexed: 06/13/2023]
Abstract
The use of cosmetics is growing with each passing day, arousing widespread attention to their ingredients. Avobenzone (AVO) and nanoplastics (NPs) are typical ingredients in cosmetics, which coexist in the aquatic environment and have a combined effect on aquatic organisms. In this study, the accumulation of AVO and NPs in zebrafish larvae and effects on gene expression and enzymatic activity related to nervous functions, and locomotor behavior were investigated. AVO and NPs accumulated continuously in zebrafish, and the combined exposure enhanced AVO accumulation. After recovery, the accumulated concentrations of AVO and NPs in zebrafish remained unchanged, suggesting that AVO and NPs could not be eliminated in 72 h. The genes regulated nervous system development were affected mainly by AVO exposure, while the genes regulated retinal system development were affected by NPs exposure. Single and combined exposures of AVO and NPs affected the activities of acetylcholinesterase and antioxidant enzymes in zebrafish, and superoxide dismutase activity could not return to normal level after 72 h of recovery period. The locomotor activity of zebrafish larvae was significantly inhibited by AVO and NPs, which might be related to the alterations in functions of nervous system development and retinal system development as well as the interference of neurotransmitter system and antioxidant system.
Collapse
Affiliation(s)
- Yuxuan Liu
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| | - Yonghua Wang
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China.
| | - Na Li
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| | - Shengnan Jiang
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| |
Collapse
|
41
|
Mahon N, Slater K, O'Brien J, Alvarez Y, Reynolds A, Kennedy B. Discovery and Development of the Quininib Series of Ocular Drugs. J Ocul Pharmacol Ther 2022; 38:33-42. [PMID: 35089801 DOI: 10.1089/jop.2021.0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The quininib series is a novel collection of small-molecule drugs with antiangiogenic, antivascular permeability, anti-inflammatory, and antiproliferative activity. Quininib was initially identified as a drug hit during a random chemical library screen for determinants of developmental ocular angiogenesis in zebrafish. To enhance drug efficacy, novel quininib analogs were designed by applying medicinal chemistry approaches. The resulting quininib drug series has efficacy in in vitro and ex vivo models of angiogenesis utilizing human cell lines and tissues. In vivo, quininib drugs reduce pathological angiogenesis and retinal vascular permeability in rodent models. Quininib acts as a cysteinyl leukotriene (CysLT) receptor antagonist, revealing new roles of these G-protein-coupled receptors in developmental angiogenesis of the eye and unexpectedly in uveal melanoma (UM). The quininib series highlighted the potential of CysLT receptors as therapeutic targets for retinal vasculopathies (e.g., neovascular age-related macular degeneration, diabetic retinopathy, and diabetic macular edema) and ocular cancers (e.g., UM).
Collapse
Affiliation(s)
- Niamh Mahon
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Kayleigh Slater
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Justine O'Brien
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Yolanda Alvarez
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Alison Reynolds
- UCD Conway Institute, University College Dublin, Dublin, Ireland.,UCD School of Veterinary Medicine, Veterinary Sciences Center, University College Dublin, Dublin, Ireland
| | - Breandán Kennedy
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
42
|
Damage to Olfactory Organs of Adult Zebrafish Induced by Diesel Particulate Matter. Int J Mol Sci 2021; 23:ijms23010407. [PMID: 35008830 PMCID: PMC8745429 DOI: 10.3390/ijms23010407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/19/2021] [Accepted: 12/27/2021] [Indexed: 01/19/2023] Open
Abstract
Particulate matter (PM) is an environmental hazard that is associated with various human health risks. The olfactory system is directly exposed to PM; therefore, the influence of PM exposure on olfactory function must be investigated. In this study, we propose a zebrafish olfactory model to evaluate the effects of exposure to diesel particulate matter (DPM), which was labeled Korean diesel particulate matter (KDP20). KDP20 comprises heavy metals and polycyclic aromatic hydrocarbons (PAHs). KDP20 exposed olfactory organs exhibited reduced cilia and damaged epithelium. Olfactory dysfunction was confirmed using an odor-mediated behavior test. Furthermore, the olfactory damage was analyzed using Alcian blue and anti-calretinin staining. KDP20 exposed olfactory organs exhibited histological damages, such as increased goblet cells, decreased cell density, and calretinin level. Quantitative real-time polymerase chain reaction (qRT-PCR) revealed that PAHs exposure related genes (AHR2 and CYP1A) were upregulated. Reactive oxidation stress (ROS) (CAT) and inflammation (IL-1B) related genes were upregulated. Furthermore, olfactory sensory neuron (OSN) related genes (OMP and S100) were downregulated. In conclusion, KDP20 exposure induced dysfunction of the olfactory system. Additionally, the zebrafish olfactory system exhibited a regenerative capacity with recovery conditions. Thus, this model may be used in future investigating PM-related diseases.
Collapse
|
43
|
An Overview of Zebrafish Modeling Methods in Drug Discovery and Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1387:145-169. [PMID: 34961915 DOI: 10.1007/5584_2021_684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Animal studies are recognized as a significant step forward in the bridging between drug discovery and clinical applications. Animal models, due to their relative genetic, molecular, physiological, and even anatomical similarities to humans, can provide a suitable platform for unraveling the mechanisms underlying human diseases and discovering new therapeutic approaches as well. Recently, zebrafish has attracted attention as a valuable experimental and pharmacological model in drug discovery and development studies due to its prominent characteristics such as the high degree of genetic similarity with humans, genetic manipulability, and prominent clinical features. Since advancing a theory to a valid and reliable observation requires the manipulation of animals, it is, therefore, essential to use efficient modeling methods appropriate to the different aspects of experimental conditions. In this context, applying several various approaches such as using chemicals, pathogens, and genetic manipulation approaches allows zebrafish development into a preferable model that mimics some human disease pathophysiology. Thus, such modeling approaches not only can provide a framework for a comprehensive understanding of the human disease mechanisms that have a counterpart in zebrafish but also can pave the way for discovering new drugs that are accompanied by higher amelioration effects on different human diseases.
Collapse
|
44
|
Chen XF, Chen ZF, Lin ZC, Liao XL, Zou T, Qi Z, Cai Z. Toxic effects of triclocarban on larval zebrafish: A focus on visual dysfunction. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 241:106013. [PMID: 34731642 DOI: 10.1016/j.aquatox.2021.106013] [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: 06/18/2021] [Revised: 10/07/2021] [Accepted: 10/24/2021] [Indexed: 06/13/2023]
Abstract
Triclocarban (TCC) is considered an endocrine disruptor and shows antagonist activity on thyroid receptors. In view of the report that thyroid hormone signaling mediates retinal cone photoreceptor specification, we hypothesize that TCC could impair visual function, which is vital to wildlife. In order to verify our hypothesis, we assessed alteration in the retinal structure (retinal layer thickness and cell density), visually-mediated behavior, cone and rod opsin gene expression, and photoreceptor immunostaining in zebrafish larvae exposed to TCC at environmentally realistic concentrations (0.16 ± 0.005 µg/L, L-group) and one-fifth of the median lethal concentrations (25.4 ± 1.02 µg/L, H-group). Significant decrease in eye size, ganglion cell density, optokinetic response, and phototactic response can be observed in the L-group, while the thickness of outer nuclear layer, where the cell bodies of cone and rod cells are located, was significantly reduced with the down-regulation of critical opsin gene (opn1sw2, opn1mw1, opn1mw3, opn1lw1, opn1lw2, and rho) expression and rhodopsin immunofluorescence in the H-group. It should be noted that TCC could affect the sensitivity of zebrafish larvae to red and green light according to the results of behavioral and opsin gene expression analysis. These findings provide the first evidence to support our hypothesis that the visual system, a novel toxicological target, is affected by TCC. Consequently, we urgently call for a more in-depth exploration of TCC-induced ocular toxicity to aquatic organisms and even to humans.
Collapse
Affiliation(s)
- Xiao-Fan Chen
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhi-Feng Chen
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, South China Normal University, Guangzhou 510006, China.
| | - Zhi-Cheng Lin
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiao-Liang Liao
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Ting Zou
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Zenghua Qi
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Zongwei Cai
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China; State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| |
Collapse
|
45
|
Landowski M, Bowes Rickman C. Targeting Lipid Metabolism for the Treatment of Age-Related Macular Degeneration: Insights from Preclinical Mouse Models. J Ocul Pharmacol Ther 2021; 38:3-32. [PMID: 34788573 PMCID: PMC8817708 DOI: 10.1089/jop.2021.0067] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Age-related macular degeneration (AMD) is a major leading cause of irreversible visual impairment in the world with limited therapeutic interventions. Histological, biochemical, genetic, and epidemiological studies strongly implicate dysregulated lipid metabolism in the retinal pigmented epithelium (RPE) in AMD pathobiology. However, effective therapies targeting lipid metabolism still need to be identified and developed for this blinding disease. To test lipid metabolism-targeting therapies, preclinical AMD mouse models are needed to establish therapeutic efficacy and the role of lipid metabolism in the development of AMD-like pathology. In this review, we provide a comprehensive overview of current AMD mouse models available to researchers that could be used to provide preclinical evidence supporting therapies targeting lipid metabolism for AMD. Based on previous studies of AMD mouse models, we discuss strategies to modulate lipid metabolism as well as examples of studies evaluating lipid-targeting therapeutics to restore lipid processing in the RPE. The use of AMD mouse models may lead to worthy lipid-targeting candidate therapies for clinical trials to prevent the blindness caused by AMD.
Collapse
Affiliation(s)
- Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA.,McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
46
|
Ganzen L, Ko MJ, Zhang M, Xie R, Chen Y, Zhang L, James R, Mumm J, van Rijn RM, Zhong W, Pang CP, Zhang M, Tsujikawa M, Leung YF. Drug screening with zebrafish visual behavior identifies carvedilol as a potential treatment for an autosomal dominant form of retinitis pigmentosa. Sci Rep 2021; 11:11432. [PMID: 34075074 PMCID: PMC8169685 DOI: 10.1038/s41598-021-89482-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 04/23/2021] [Indexed: 02/04/2023] Open
Abstract
Retinitis Pigmentosa (RP) is a mostly incurable inherited retinal degeneration affecting approximately 1 in 4000 individuals globally. The goal of this work was to identify drugs that can help patients suffering from the disease. To accomplish this, we screened drugs on a zebrafish autosomal dominant RP model. This model expresses a truncated human rhodopsin transgene (Q344X) causing significant rod degeneration by 7 days post-fertilization (dpf). Consequently, the larvae displayed a deficit in visual motor response (VMR) under scotopic condition. The diminished VMR was leveraged to screen an ENZO SCREEN-WELL REDOX library since oxidative stress is postulated to play a role in RP progression. Our screening identified a beta-blocker, carvedilol, that ameliorated the deficient VMR of the RP larvae and increased their rod number. Carvedilol may directly on rods as it affected the adrenergic pathway in the photoreceptor-like human Y79 cell line. Since carvedilol is an FDA-approved drug, our findings suggest that carvedilol can potentially be repurposed to treat autosomal dominant RP patients.
Collapse
Affiliation(s)
- Logan Ganzen
- grid.169077.e0000 0004 1937 2197Department of Biological Sciences, Purdue University, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue University Life Sciences Program, Purdue University, West Lafayette, IN 47907 USA
| | - Mee Jung Ko
- grid.169077.e0000 0004 1937 2197Purdue University Life Sciences Program, Purdue University, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907 USA
| | - Mengrui Zhang
- grid.213876.90000 0004 1936 738XDepartment of Statistics, University of Georgia, Athens, GA 30602 USA
| | - Rui Xie
- grid.170430.10000 0001 2159 2859Department of Statistics and Data Science, University of Central Florida, Orlando, FL 32816 USA
| | - Yongkai Chen
- grid.213876.90000 0004 1936 738XDepartment of Statistics, University of Georgia, Athens, GA 30602 USA
| | - Liyun Zhang
- grid.21107.350000 0001 2171 9311Wilmer Eye Institute, John Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Rebecca James
- grid.169077.e0000 0004 1937 2197Department of Biological Sciences, Purdue University, West Lafayette, IN 47907 USA
| | - Jeff Mumm
- grid.21107.350000 0001 2171 9311Wilmer Eye Institute, John Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Richard M. van Rijn
- grid.169077.e0000 0004 1937 2197Purdue University Life Sciences Program, Purdue University, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue Institute for Integrative Neuroscience, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue Institute for Drug Discovery, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907 USA
| | - Wenxuan Zhong
- grid.213876.90000 0004 1936 738XDepartment of Statistics, University of Georgia, Athens, GA 30602 USA
| | - Chi Pui Pang
- grid.10784.3a0000 0004 1937 0482Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, Hong Kong, China ,grid.263451.70000 0000 9927 110XJoint Shantou International Eye Center, Shantou University and the Chinese University of Hong Kong, Shantou, China
| | - Mingzhi Zhang
- grid.263451.70000 0000 9927 110XJoint Shantou International Eye Center, Shantou University and the Chinese University of Hong Kong, Shantou, China
| | - Motokazu Tsujikawa
- grid.136593.b0000 0004 0373 3971Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan ,grid.136593.b0000 0004 0373 3971Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuk Fai Leung
- grid.169077.e0000 0004 1937 2197Department of Biological Sciences, Purdue University, West Lafayette, IN 47907 USA ,grid.257413.60000 0001 2287 3919Department of Biochemistry and Molecular Biology, Indiana University School of Medicine Lafayette, 625 Harrison Street, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue Institute for Integrative Neuroscience, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue Institute for Drug Discovery, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907 USA
| |
Collapse
|
47
|
Chalin D, Bureau C, Parmeggiani A, Rochal S, Kissa K, Golushko I. Modeling and live imaging of mechanical instabilities in the zebrafish aorta during hematopoiesis. Sci Rep 2021; 11:9316. [PMID: 33927284 PMCID: PMC8085226 DOI: 10.1038/s41598-021-88667-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/01/2021] [Indexed: 01/27/2023] Open
Abstract
All blood cells originate from hematopoietic stem/progenitor cells (HSPCs). HSPCs are formed from endothelial cells (ECs) of the dorsal aorta (DA), via endothelial-to-hematopoietic transition (EHT). The zebrafish is a primary model organism to study the process in vivo. While the role of mechanical stress in controlling gene expression promoting cell differentiation is actively investigated, mechanisms driving shape changes of the DA and individual ECs remain poorly understood. We address this problem by developing a new DA micromechanical model and applying it to experimental data on zebrafish morphogenesis. The model considers the DA as an isotropic tubular membrane subjected to hydrostatic blood pressure and axial stress. The DA evolution is described as a movement in the dimensionless controlling parameters space: normalized hydrostatic pressure and axial stress. We argue that HSPC production is accompanied by two mechanical instabilities arising in the system due to the plane stress in the DA walls and show how a complex interplay between mechanical forces in the system drives the emerging morphological changes.
Collapse
Affiliation(s)
- Dmitrii Chalin
- Research and Education Center "Materials", Don State Technical University, 1 Gagarin Square, Rostov-on-Don, 344000, Russia
| | - Charlotte Bureau
- LPHI, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Andrea Parmeggiani
- LPHI, University of Montpellier, CNRS, INSERM, Montpellier, France.,Laboratoire Charles Coulomb, University of Montpellier, CNRS, Montpellier, France
| | - Sergei Rochal
- Faculty of Physics, Southern Federal University, Zorge 5, Rostov-on-Don, 344090, Russian Federation
| | - Karima Kissa
- LPHI, University of Montpellier, CNRS, INSERM, Montpellier, France.
| | - Ivan Golushko
- Research and Education Center "Materials", Don State Technical University, 1 Gagarin Square, Rostov-on-Don, 344000, Russia.
| |
Collapse
|
48
|
Fitzgerald JA, Könemann S, Krümpelmann L, Županič A, Vom Berg C. Approaches to Test the Neurotoxicity of Environmental Contaminants in the Zebrafish Model: From Behavior to Molecular Mechanisms. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2021; 40:989-1006. [PMID: 33270929 DOI: 10.1002/etc.4951] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/15/2020] [Accepted: 11/29/2020] [Indexed: 06/12/2023]
Abstract
The occurrence of neuroactive chemicals in the aquatic environment is on the rise and poses a potential threat to aquatic biota of currently unpredictable outcome. In particular, subtle changes caused by these chemicals to an organism's sensation or behavior are difficult to tackle with current test systems that focus on rodents or with in vitro test systems that omit whole-animal responses. In recent years, the zebrafish (Danio rerio) has become a popular model organism for toxicological studies and testing strategies, such as the standardized use of zebrafish early life stages in the Organisation for Economic Co-operation and Development's guideline 236. In terms of neurotoxicity, the zebrafish provides a powerful model to investigate changes to the nervous system from several different angles, offering the ability to tackle the mechanisms of action of chemicals in detail. The mechanistic understanding gained through the analysis of this model species provides a good basic knowledge of how neuroactive chemicals might interact with a teleost nervous system. Such information can help infer potential effects occurring to other species exposed to neuroactive chemicals in their aquatic environment and predicting potential risks of a chemical for the aquatic ecosystem. In the present article, we highlight approaches ranging from behavioral to structural, functional, and molecular analysis of the larval zebrafish nervous system, providing a holistic view of potential neurotoxic outcomes. Environ Toxicol Chem 2021;40:989-1006. © 2020 SETAC.
Collapse
Affiliation(s)
- Jennifer A Fitzgerald
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Sarah Könemann
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
- EPF Lausanne, School of Architecture, Civil and Environmental Engineering, Lausanne, Switzerland
| | - Laura Krümpelmann
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Anže Županič
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
- National Institute of Biology, Ljubljana, Slovenia
| | - Colette Vom Berg
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| |
Collapse
|
49
|
Neurodegeneration, Neuroprotection and Regeneration in the Zebrafish Retina. Cells 2021; 10:cells10030633. [PMID: 33809186 PMCID: PMC8000332 DOI: 10.3390/cells10030633] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/10/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Neurodegenerative retinal diseases, such as glaucoma and diabetic retinopathy, involve a gradual loss of neurons in the retina as the disease progresses. Central nervous system neurons are not able to regenerate in mammals, therefore, an often sought after course of treatment for neuronal loss follows a neuroprotective or regenerative strategy. Neuroprotection is the process of preserving the structure and function of the neurons that have survived a harmful insult; while regenerative approaches aim to replace or rewire the neurons and synaptic connections that were lost, or induce regrowth of damaged axons or dendrites. In order to test the neuroprotective effectiveness or the regenerative capacity of a particular agent, a robust experimental model of retinal neuronal damage is essential. Zebrafish are being used more often in this type of study because their eye structure and development is well-conserved between zebrafish and mammals. Zebrafish are robust genetic tools and are relatively inexpensive to maintain. The large array of functional and behavioral tests available in zebrafish makes them an attractive model for neuroprotection studies. Some common insults used to model retinal disease and study neuroprotection in zebrafish include intense light, chemical toxicity and mechanical damage. This review covers the existing retinal neuroprotection and regeneration literature in the zebrafish and highlights their potential for future studies.
Collapse
|
50
|
Development of a zebrafish screening model for diabetic retinopathy induced by hyperglycemia: Reproducibility verification in animal model. Biomed Pharmacother 2021; 135:111201. [PMID: 33421732 DOI: 10.1016/j.biopha.2020.111201] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/15/2020] [Accepted: 12/26/2020] [Indexed: 02/08/2023] Open
Abstract
This study aimed at creating a zebrafish screening model for diabetic retinopathy, and evaluated the effects of aflibercept, which is being used to treated diabetic retinopathy. A morphological change occurred at 160 mM of glucose. The survival and hatching rate decreased in a dose-dependent manner. In the 130 mM glucose group, the retinal vessel diameter was more than double that in the normal group. The zebrafish embryo morphology changed in 200 μg/mL and 400 μg/mL at aflibercept. The survival and hatching rate decrease at 400 μg/mL. Aflibercept 100 μg/mL was a nontoxic and effective dose for the zebrafish diabetic retinopathy model. The expression of diabetic retinopathy inflammatory markers was increased in hyperglycemia. But the inflammation was improved by aflibercept in the zebrafish eye. In a zebrafish diabetic retinopathy model, the diameters of retinal vessels were reduced after treatment with aflibercept, and molecular biological and histopathological efficacy was confirmed. This model can serve for screening of new drug candidates for treatment of in diabetic retinopathy.
Collapse
|