1
|
Jobling AI, Findlay Q, Greferath U, Vessey KA, Gunnam S, Morrison V, Venables G, Guymer RH, Fletcher EL. Nanosecond laser induces proliferation and improved cellular health within the retinal pigment epithelium. Front Med (Lausanne) 2025; 12:1516900. [PMID: 40098930 PMCID: PMC11911352 DOI: 10.3389/fmed.2025.1516900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Background Age-related macular degeneration (AMD) is a leading cause of vision loss in those over 60 years of age. Although there are limited interventions that may prevent the development or progression of disease, more efficacious treatments are required. Short-pulsed laser treatment shows promise in delaying progression of early disease. This work details how nanosecond laser influences the retinal pigment epithelium (RPE), the principal cell type implicated in AMD. Methods C57BL/6J mice (3-month-old) underwent monocular nanosecond laser treatment to assess short-term RPE response, while 9-month-old C57BL/6J and ApoEnull mice were similarly treated and longer-term responses investigated after 3 months. Human tissue was also obtained after 2 nanosecond laser treatments (1 month apart). RPE proliferation was assessed using bromodeoxyuridine and RPE gene change explored using qPCR and RNAseq. Melanin and lipofuscin content were quantified using histological techniques. Results Nanosecond laser induced RPE proliferation in treated and fellow mouse eyes, with monolayer repair occurring within 3 days. This was replicated in human tissue, albeit over a longer duration (1-4 weeks). Wildtype animals showed no overt change in RPE gene expression after short or longer post-treatment durations, while laser treated ApoEnull animals showed increased Mertk and Pedf expression, and a reduced number of dysregulated aging genes in treated and fellow eyes after 3 months. Furthermore, melanin and lipofuscin content were restored to wildtype levels in laser-treated ApoEnull RPE, while melanolipofuscin granules were reduced within treated regions of human RPE. Conclusion This work shows nanosecond laser stimulates RPE proliferation and results in an improved cellular phenotype. These data provide a biological basis for the prophylactic use of nanosecond lasers in AMD.
Collapse
Affiliation(s)
- Andrew I. Jobling
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Quan Findlay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Ursula Greferath
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Kirstan A. Vessey
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Satya Gunnam
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Victoria Morrison
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Gene Venables
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Robyn H. Guymer
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Ophthalmology, Department of Surgery, The University of Melbourne, Parkville, VIC, Australia
| | - Erica L. Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
2
|
Leach LL, Gonzalez RG, Jayawardena SU, Gross JM. Interleukin-34 and debris clearance by mononuclear phagocytes drive retinal pigment epithelium regeneration in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632236. [PMID: 39868193 PMCID: PMC11761032 DOI: 10.1101/2025.01.10.632236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The retinal pigment epithelium (RPE) surrounds the posterior eye and maintains the health and function of the photoreceptors. Consequently, RPE dysfunction or damage has a devastating impact on vision. Due to complex etiologies, there are currently no cures for patients with RPE degenerative diseases, which remain some of the most prevalent causes of vision loss worldwide. Further, owing to a limited capacity for mammalian tissue repair, we know little about how the RPE regenerates. Here, we utilize zebrafish as a model to uncover novel mechanisms driving intrinsic RPE regeneration. We show that interleukin-34 signaling from damaged RPE is required for precisely timed recruitment of mononuclear phagocytes (MNPs) to the injury site. Additionally, we find that cellular debris clearance by MNPs is indispensable for regeneration, as microglia-deficient zebrafish fail to regenerate RPE and photoreceptor tissues. Together, our results establish specific pro-regenerative functions of MNPs after RPE damage.
Collapse
Affiliation(s)
- Lyndsay L. Leach
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
| | - Rebecca G. Gonzalez
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
| | - Sayuri U. Jayawardena
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
| | - Jeffrey M. Gross
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
| |
Collapse
|
3
|
Marchesi N, Capierri M, Pascale A, Barbieri A. Different Therapeutic Approaches for Dry and Wet AMD. Int J Mol Sci 2024; 25:13053. [PMID: 39684764 DOI: 10.3390/ijms252313053] [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: 10/25/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Age-related macular degeneration (AMD) is the most common cause of irreversible loss of central vision in elderly subjects, affecting men and women equally. It is a degenerative pathology that causes progressive damage to the macula, the central and most vital part of the retina. There are two forms of AMD depending on how the macula is damaged, dry AMD and wet or neovascular AMD. Dry AMD is the most common form; waste materials accumulate under the retina as old cells die, not being replaced. Wet AMD is less common, but can lead to vision loss much more quickly. Wet AMD is characterized by new abnormal blood vessels developing under the macula, where they do not normally grow. This frequently occurs in patients who already have dry AMD, as new blood vessels are developed to try to solve the problem. It is not known what causes AMD to develop; however, certain risk factors (i.e., age, smoking, genetic factors) can increase the risk of developing AMD. There are currently no treatments for dry AMD. There is evidence that not smoking, exercising regularly, eating nutritious food, and taking certain supplements can reduce the risk of acquiring AMD or slow its development. The main treatment for wet AMD is inhibitors of VEGF (vascular endothelial growth factor), a protein that stimulates the growth of new blood vessels. VEGF inhibitors can stop the growth of new blood vessels, preventing further damage to the macula and vision loss. In most patients, VEGF inhibitors can improve vision if macular degeneration is diagnosed early and treated accordingly. However, VEGF inhibitors cannot repair damage that has already occurred. Current AMD research is trying to find treatments for dry AMD and other options for wet AMD. This review provides a summary of the current evidence regarding the different treatments aimed at both forms of AMD with particular and greater attention to the dry form.
Collapse
Affiliation(s)
- Nicoletta Marchesi
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Martina Capierri
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Annalisa Barbieri
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
4
|
Saputra F, Kishida M, Hu SY. Nitrate and Nitrite Exposure Induces Visual Impairments in Adult Zebrafish. TOXICS 2024; 12:518. [PMID: 39058170 PMCID: PMC11281020 DOI: 10.3390/toxics12070518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Nitrate and nitrite have emerged as increasingly common environmental pollutants, posing significant risks to various forms of life within ecosystems. To understand their impact on the visual system of zebrafish, adult zebrafish were exposed to environmentally relevant concentrations of nitrate (10 mg/L) and nitrite (1 mg/L) for 7 days. Visual behaviors were examined using optomotor and avoidance response. The eyeballs of the zebrafish were collected for H&E staining, IHC, and qPCR. Exposure decreased visual behavior and the thickness of most retinal layers. Exposure decreased expression of pax6a, pax6b, gpx1a, and bcl2a. Exposure increased expression of esr1, esr1a, esr2b, cyp19a1b, sod1a, nos2a, casps3, and tp53, and increased retinal brain aromatase expression by IHC. Collectively, our findings demonstrate that nitrate and nitrite exposure negatively impacted the visual system of adult zebrafish, highlighting the potential hazards of these environmental pollutants on aquatic organisms.
Collapse
Affiliation(s)
- Febriyansyah Saputra
- Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan;
| | - Mitsuyo Kishida
- Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan;
| | - Shao-Yang Hu
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| |
Collapse
|
5
|
Bludau O, Weber A, Bosak V, Kuscha V, Dietrich K, Hans S, Brand M. Inflammation is a critical factor for successful regeneration of the adult zebrafish retina in response to diffuse light lesion. Front Cell Dev Biol 2024; 12:1332347. [PMID: 39071801 PMCID: PMC11272569 DOI: 10.3389/fcell.2024.1332347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 06/17/2024] [Indexed: 07/30/2024] Open
Abstract
Inflammation can lead to persistent and irreversible loss of retinal neurons and photoreceptors in mammalian vertebrates. In contrast, in the adult zebrafish brain, acute neural inflammation is both necessary and sufficient to stimulate regeneration of neurons. Here, we report on the critical, positive role of the immune system to support retina regeneration in adult zebrafish. After sterile ablation of photoreceptors by phototoxicity, we find rapid response of immune cells, especially monocytes/microglia and neutrophils, which returns to homeostatic levels within 14 days post lesion. Pharmacological or genetic impairment of the immune system results in a reduced Müller glia stem cell response, seen as decreased reactive proliferation, and a strikingly reduced number of regenerated cells from them, including photoreceptors. Conversely, injection of the immune stimulators flagellin, zymosan, or M-CSF into the vitreous of the eye, leads to a robust proliferation response and the upregulation of regeneration-associated marker genes in Müller glia. Our results suggest that neuroinflammation is a necessary and sufficient driver for retinal regeneration in the adult zebrafish retina.
Collapse
Affiliation(s)
- Oliver Bludau
- CRTD—Center for Regenerative Therapies, and PoL—Cluster of Excellence Physics of Life, Dresden, Germany
| | - Anke Weber
- CRTD—Center for Regenerative Therapies, and PoL—Cluster of Excellence Physics of Life, Dresden, Germany
| | - Viktoria Bosak
- CRTD—Center for Regenerative Therapies, and PoL—Cluster of Excellence Physics of Life, Dresden, Germany
| | - Veronika Kuscha
- CRTD—Center for Regenerative Therapies, and PoL—Cluster of Excellence Physics of Life, Dresden, Germany
| | - Kristin Dietrich
- CRTD—Center for Regenerative Therapies, and PoL—Cluster of Excellence Physics of Life, Dresden, Germany
| | - Stefan Hans
- CRTD—Center for Regenerative Therapies, and PoL—Cluster of Excellence Physics of Life, Dresden, Germany
| | - Michael Brand
- CRTD—Center for Regenerative Therapies, and PoL—Cluster of Excellence Physics of Life, Dresden, Germany
| |
Collapse
|
6
|
Lulli M, Tartaro R, Papucci L, Magnelli L, Kaur IP, Caporossi T, Rizzo S, Mannini A, Giansanti F, Schiavone N. Effects of a human amniotic membrane extract on ARPE-19 cells. Mol Biol Rep 2024; 51:746. [PMID: 38874663 PMCID: PMC11178654 DOI: 10.1007/s11033-024-09647-7] [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: 08/10/2023] [Accepted: 05/16/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Human Amniotic Membrane (hAM) is endowed with several biological activities and might be considered an optimal tool in surgical treatment for different ophthalmic pathologies. We pioneered the surgical use of hAM to treat retinal pathologies such as macular holes, tears, and retinal detachments, and to overcome photoreceptor damage in age-related macular degeneration. Although hAM contributed to improved outcomes, the mechanisms of its effects are not yet fully understood. The characterization and explanation of the effects of hAM would allow the adoption of this new natural product in different retinal pathologies, operative contexts, and hAM formulations. At this end, we studied the properties of a hAM extract (hAME) on the ARPE-19 cells. METHODS AND RESULTS A non-denaturing sonication-based technique was developed to obtain a suitable hAME. Viability, proliferation, apoptosis, oxidative stress, and epithelial-mesenchymal transition (EMT) were studied in hAME-treated ARPE-19 cells. The hAME was able to increase ARPE-19 cell viability even in the presence of oxidative stress (H2O2, TBHP). Moreover, hAME prevented the expression of EMT features, such as EMT-related proteins, fibrotic foci formation, and migration induced by different cytokines. CONCLUSIONS Our results demonstrate that the hAME retains most of the properties observed in the whole tissue by others. The hAME, other than providing a manageable research tool, could represent a cost-effective and abundant drug to treat retinal pathologies in the future.
Collapse
Affiliation(s)
- Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Ruggero Tartaro
- Department of NEUROFARBA, Ophthalmology, University of Florence, Careggi, Florence, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Indu Pal Kaur
- UGC-Centre of Advanced Study, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Tomaso Caporossi
- Vitreoretinal Surgery Unit, Isola Tiberina Gemelli Isola Hospital, Rome, Italy
- Catholic University Sacro Cuore, Rome, Italy
| | - Stanislao Rizzo
- Department of Ophthalmology, Catholic University of Sacred-Heart Foundation "Policlinico Universitario A. Gemelli" IRCCS, Rome, Italy
| | - Antonella Mannini
- Department of Experimental and Clinical Medicine - Internal Medicine Section, University of Florence, Florence, Italy.
| | - Fabrizio Giansanti
- Department of NEUROFARBA, Ophthalmology, University of Florence, Careggi, Florence, Italy
| | - Nicola Schiavone
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| |
Collapse
|
7
|
Yang Y, He B, Mu X, Qi S. Exposure to flutolanil at environmentally relevant concentrations can induce image and non-image-forming failure of zebrafish larvae through neuro and visual disruptions. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:134108. [PMID: 38521039 DOI: 10.1016/j.jhazmat.2024.134108] [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: 02/09/2024] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Numerous pesticides pose a threat to aquatic ecosystems, jeopardizing aquatic animal species and impacting human health. While the contamination of aquatic environment by flutolanil and its adverse effects on animal in the treatment of rich sheath blight have been reported, the neuro-visual effects of flutolanil at environmentally relevant concentrations remain unknown. In this study, we administered flutolanil to zebrafish embryos (0, 0.125, 0.50 and 2.0 mg/L) for 4 days to investigate its impact on the neuro and visual system. The results revealed that flutolanil induced abnormal behavior in larvae, affecting locomotor activity, stimuli response and phototactic response. Additionally, it led to defective brain and ocular development and differentiation. The disruption extended to the neurological system and visual phototransduction of larvae, evidenced by significant disturbances in genes and proteins related to neurodevelopment, neurotransmission, eye development, and visual function. Untargeted metabolomics analysis revealed that the GABAergic signaling pathway and increased levels of glutamine, glutamate, andγ-aminobutyric acid were implicated in the response to neuro and visual system injury induced by flutolanil, contributing to aberrant development, behavioral issues, and endocrine disruption. This study highlights the neuro-visual injury caused by flutolanil in aquatic environment, offering fresh insights into the mechanisms underlying image and non-image effects.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China
| | - Bin He
- Institute of Animal Husbandry and Veterinary, Wuhan Academy of Agricultural Sciences, Wuhan 430070, People's Republic of China
| | - Xiyan Mu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, People's Republic of China.
| | - Suzhen Qi
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, People's Republic of China.
| |
Collapse
|
8
|
Becker S, L'Ecuyer Z, Jones BW, Zouache MA, McDonnell FS, Vinberg F. Modeling complex age-related eye disease. Prog Retin Eye Res 2024; 100:101247. [PMID: 38365085 PMCID: PMC11268458 DOI: 10.1016/j.preteyeres.2024.101247] [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: 08/15/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Modeling complex eye diseases like age-related macular degeneration (AMD) and glaucoma poses significant challenges, since these conditions depend highly on age-related changes that occur over several decades, with many contributing factors remaining unknown. Although both diseases exhibit a relatively high heritability of >50%, a large proportion of individuals carrying AMD- or glaucoma-associated genetic risk variants will never develop these diseases. Furthermore, several environmental and lifestyle factors contribute to and modulate the pathogenesis and progression of AMD and glaucoma. Several strategies replicate the impact of genetic risk variants, pathobiological pathways and environmental and lifestyle factors in AMD and glaucoma in mice and other species. In this review we will primarily discuss the most commonly available mouse models, which have and will likely continue to improve our understanding of the pathobiology of age-related eye diseases. Uncertainties persist whether small animal models can truly recapitulate disease progression and vision loss in patients, raising doubts regarding their usefulness when testing novel gene or drug therapies. We will elaborate on concerns that relate to shorter lifespan, body size and allometries, lack of macula and a true lamina cribrosa, as well as absence and sequence disparities of certain genes and differences in their chromosomal location in mice. Since biological, rather than chronological, age likely predisposes an organism for both glaucoma and AMD, more rapidly aging organisms like small rodents may open up possibilities that will make research of these diseases more timely and financially feasible. On the other hand, due to the above-mentioned anatomical and physiological features, as well as pharmacokinetic and -dynamic differences small animal models are not ideal to study the natural progression of vision loss or the efficacy and safety of novel therapies. In this context, we will also discuss the advantages and pitfalls of alternative models that include larger species, such as non-human primates and rabbits, patient-derived retinal organoids, and human organ donor eyes.
Collapse
Affiliation(s)
- Silke Becker
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Zia L'Ecuyer
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Bryan W Jones
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Moussa A Zouache
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Frans Vinberg
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
9
|
Sharkova M, Aparicio G, Mouzaaber C, Zolessi FR, Hocking JC. Photoreceptor calyceal processes accompany the developing outer segment, adopting a stable length despite a dynamic core. J Cell Sci 2024; 137:jcs261721. [PMID: 38477343 PMCID: PMC11058337 DOI: 10.1242/jcs.261721] [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: 10/16/2023] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Vertebrate photoreceptors detect light through a large cilium-based outer segment, which is filled with photopigment-laden membranous discs. Surrounding the base of the outer segment are microvilli-like calyceal processes (CPs). Although CP disruption has been associated with altered outer segment morphology and photoreceptor degeneration, the role of the CPs remains elusive. Here, we used zebrafish as a model to characterize CPs. We quantified CP parameters and report a strong disparity in outer segment coverage between photoreceptor subtypes. CP length is stable across light and dark conditions, yet heat-shock inducible expression of tagged actin revealed rapid turnover of the CP actin core. Detailed imaging of the embryonic retina uncovered substantial remodeling of the developing photoreceptor apical surface, including a transition from dynamic tangential processes to vertically oriented CPs immediately prior to outer segment formation. Remarkably, we also found a direct connection between apical extensions of the Müller glia and retinal pigment epithelium, arranged as bundles around the ultraviolet sensitive cones. In summary, our data characterize the structure, development and surrounding environment of photoreceptor microvilli in the zebrafish retina.
Collapse
Affiliation(s)
- Maria Sharkova
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gonzalo Aparicio
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, 11400, Uruguay
- Institut Pasteur Montevideo, Uruguay
| | - Constantin Mouzaaber
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Flavio R. Zolessi
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, 11400, Uruguay
- Institut Pasteur Montevideo, Uruguay
| | - Jennifer C. Hocking
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Division of Anatomy, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
10
|
Konar GJ, Flickinger Z, Sharma S, Vallone KT, Lyon CE, Doshier C, Lingan A, Lyon W, Patton JG. Damage-Induced Senescent Immune Cells Regulate Regeneration of the Zebrafish Retina. AGING BIOLOGY 2024; 2:e20240021. [PMID: 39156966 PMCID: PMC11328971 DOI: 10.59368/agingbio.20240021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Zebrafish spontaneously regenerate their retinas in response to damage through the action of Müller glia (MG). Even though MG are conserved in higher vertebrates, the capacity to regenerate retinal damage is lost. Recent work has focused on the regulation of inflammation during tissue regeneration, with temporal roles for macrophages and microglia. Senescent cells that have withdrawn from the cell cycle have mostly been implicated in aging but are still metabolically active, releasing a variety of signaling molecules as part of the senescence-associated secretory phenotype. Here, we discover that in response to retinal damage, a subset of cells expressing markers of microglia/macrophages also express markers of senescence. These cells display a temporal pattern of appearance and clearance during retina regeneration. Premature removal of senescent cells by senolytic treatment led to a decrease in proliferation and incomplete repair of the ganglion cell layer after N-methyl-D-aspartate damage. Our results demonstrate a role for modulation of senescent cell responses to balance inflammation, regeneration, plasticity, and repair as opposed to fibrosis and scarring.
Collapse
Affiliation(s)
- Gregory J. Konar
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Zachary Flickinger
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Shivani Sharma
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Kyle T. Vallone
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Charles E. Lyon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Claire Doshier
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Audrey Lingan
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - William Lyon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - James G. Patton
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
11
|
Nguyen VP, Zhe J, Hu J, Ahmed U, Paulus YM. Molecular and cellular imaging of the eye. BIOMEDICAL OPTICS EXPRESS 2024; 15:360-386. [PMID: 38223186 PMCID: PMC10783915 DOI: 10.1364/boe.502350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/25/2023] [Accepted: 12/02/2023] [Indexed: 01/16/2024]
Abstract
The application of molecular and cellular imaging in ophthalmology has numerous benefits. It can enable the early detection and diagnosis of ocular diseases, facilitating timely intervention and improved patient outcomes. Molecular imaging techniques can help identify disease biomarkers, monitor disease progression, and evaluate treatment responses. Furthermore, these techniques allow researchers to gain insights into the pathogenesis of ocular diseases and develop novel therapeutic strategies. Molecular and cellular imaging can also allow basic research to elucidate the normal physiological processes occurring within the eye, such as cell signaling, tissue remodeling, and immune responses. By providing detailed visualization at the molecular and cellular level, these imaging techniques contribute to a comprehensive understanding of ocular biology. Current clinically available imaging often relies on confocal microscopy, multi-photon microscopy, PET (positron emission tomography) or SPECT (single-photon emission computed tomography) techniques, optical coherence tomography (OCT), and fluorescence imaging. Preclinical research focuses on the identification of novel molecular targets for various diseases. The aim is to discover specific biomarkers or molecular pathways associated with diseases, allowing for targeted imaging and precise disease characterization. In parallel, efforts are being made to develop sophisticated and multifunctional contrast agents that can selectively bind to these identified molecular targets. These contrast agents can enhance the imaging signal and improve the sensitivity and specificity of molecular imaging by carrying various imaging labels, including radionuclides for PET or SPECT, fluorescent dyes for optical imaging, or nanoparticles for multimodal imaging. Furthermore, advancements in technology and instrumentation are being pursued to enable multimodality molecular imaging. Integrating different imaging modalities, such as PET/MRI (magnetic resonance imaging) or PET/CT (computed tomography), allows for the complementary strengths of each modality to be combined, providing comprehensive molecular and anatomical information in a single examination. Recently, photoacoustic microscopy (PAM) has been explored as a novel imaging technology for visualization of different retinal diseases. PAM is a non-invasive, non-ionizing radiation, and hybrid imaging modality that combines the optical excitation of contrast agents with ultrasound detection. It offers a unique approach to imaging by providing both anatomical and functional information. Its ability to utilize molecularly targeted contrast agents holds great promise for molecular imaging applications in ophthalmology. In this review, we will summarize the application of multimodality molecular imaging for tracking chorioretinal angiogenesis along with the migration of stem cells after subretinal transplantation in vivo.
Collapse
Affiliation(s)
- Van Phuc Nguyen
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Josh Zhe
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Justin Hu
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Umayr Ahmed
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Yannis M. Paulus
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
12
|
Abidi SNF, Hsu FTY, Smith-Bolton RK. Regenerative growth is constrained by brain tumor to ensure proper patterning in Drosophila. PLoS Genet 2023; 19:e1011103. [PMID: 38127821 PMCID: PMC10769103 DOI: 10.1371/journal.pgen.1011103] [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] [Received: 05/03/2023] [Revised: 01/05/2024] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Some animals respond to injury by inducing new growth to regenerate the lost structures. This regenerative growth must be carefully controlled and constrained to prevent aberrant growth and to allow correct organization of the regenerating tissue. However, the factors that restrict regenerative growth have not been identified. Using a genetic ablation system in the Drosophila wing imaginal disc, we have identified one mechanism that constrains regenerative growth, impairment of which also leads to erroneous patterning of the final appendage. Regenerating discs with reduced levels of the RNA-regulator Brain tumor (Brat) exhibit enhanced regeneration, but produce adult wings with disrupted margins that are missing extensive tracts of sensory bristles. In these mutants, aberrantly high expression of the pro-growth factor Myc and its downstream targets likely contributes to this loss of cell-fate specification. Thus, Brat constrains the expression of pro-regeneration genes and ensures that the regenerating tissue forms the proper final structure.
Collapse
Affiliation(s)
- Syeda Nayab Fatima Abidi
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Felicity Ting-Yu Hsu
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Rachel K. Smith-Bolton
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Carle R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| |
Collapse
|
13
|
Liu Y. Zebrafish as a Model Organism for Studying Pathologic Mechanisms of Neurodegenerative Diseases and other Neural Disorders. Cell Mol Neurobiol 2023; 43:2603-2620. [PMID: 37004595 PMCID: PMC11410131 DOI: 10.1007/s10571-023-01340-w] [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: 02/17/2023] [Accepted: 03/19/2023] [Indexed: 04/04/2023]
Abstract
Zebrafish are widely considered an excellent vertebrate model for studying the pathogenesis of human diseases because of their transparency of embryonic development, easy breeding, high similarity with human genes, and easy gene manipulation. Previous studies have shown that zebrafish as a model organism provides an ideal operating platform for clarifying the pathological and molecular mechanisms of neurodegenerative diseases and related human diseases. This review mainly summarizes the achievements and prospects of zebrafish used as model organisms in the research of neurodegenerative diseases and other human diseases related to the nervous system in recent years. In the future study of human disease mechanisms, the application of the zebrafish model will continue to provide a valuable operating platform and technical support for investigating and finding better prevention and treatment of these diseases, which has broad application prospects and practical significance. Zebrafish models used in neurodegenerative diseases and other diseases related to the nervous system.
Collapse
Affiliation(s)
- Yanying Liu
- Department of Basic Medicine, School of Nursing and Health, Qingdao Huanghai University, Qingdao, 266427, China.
| |
Collapse
|
14
|
Ning K, Bhuckory MB, Lo CH, Sendayen BE, Kowal TJ, Chen M, Bansal R, Chang KC, Vollrath D, Berbari NF, Mahajan VB, Hu Y, Sun Y. Cilia-associated wound repair mediated by IFT88 in retinal pigment epithelium. Sci Rep 2023; 13:8205. [PMID: 37211572 PMCID: PMC10200793 DOI: 10.1038/s41598-023-35099-3] [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: 12/06/2022] [Accepted: 05/12/2023] [Indexed: 05/23/2023] Open
Abstract
Primary cilia are conserved organelles that integrate extracellular cues into intracellular signals and are critical for diverse processes, including cellular development and repair responses. Deficits in ciliary function cause multisystemic human diseases known as ciliopathies. In the eye, atrophy of the retinal pigment epithelium (RPE) is a common feature of many ciliopathies. However, the roles of RPE cilia in vivo remain poorly understood. In this study, we first found that mouse RPE cells only transiently form primary cilia. We then examined the RPE in the mouse model of Bardet-Biedl Syndrome 4 (BBS4), a ciliopathy associated with retinal degeneration in humans, and found that ciliation in BBS4 mutant RPE cells is disrupted early during development. Next, using a laser-induced injury model in vivo, we found that primary cilia in RPE reassemble in response to laser injury during RPE wound healing and then rapidly disassemble after the repair is completed. Finally, we demonstrated that RPE-specific depletion of primary cilia in a conditional mouse model of cilia loss promoted wound healing and enhanced cell proliferation. In summary, our data suggest that RPE cilia contribute to both retinal development and repair and provide insights into potential therapeutic targets for more common RPE degenerative diseases.
Collapse
Affiliation(s)
- Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Mohajeet B Bhuckory
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Chien-Hui Lo
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Brent E Sendayen
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
- Palo Alto Veterans Administration, Palo Alto, CA, USA
| | - Tia J Kowal
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Ming Chen
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Ruchi Bansal
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Kun-Che Chang
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Douglas Vollrath
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
- Department of Genetics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Nicolas F Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Vinit B Mahajan
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA.
- Palo Alto Veterans Administration, Palo Alto, CA, USA.
| |
Collapse
|
15
|
Chen DD, Liu B, Wang Y, Jiang M, Shang G, Xue M, Jia X, Lang Y, Zhou G, Zhang F, Peng X, Hu Y. The downregulation of HSP90-controlled CRALBP expression is associated with age-related vision attenuation. FASEB J 2023; 37:e22832. [PMID: 36826429 DOI: 10.1096/fj.202201608rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/01/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023]
Abstract
The dysfunction of CRALBP, a key regulator of the visual cycle, is associated with retinitis punctata albescens characterized by night vision loss and retinal degeneration. In this paper, we find that the expression of CRALBP is regulated by heat shock protein 90 (HSP90). Inhibition of HSP90α or HSP90β expression by using the CRISPR-Cas9 technology downregulates CRALBP's mRNA and protein expression in ARPE-19 cells by triggering the degradation of transcription factor SP1 in the ubiquitin-proteasome pathway. SP1 can bind to CRALBP's promoter, and inhibition of SP1 by its inhibitor plicamycin or siRNA downregulates CRALBP's mRNA expression. In the zebrafish, inhibition of HSP90 by the intraperitoneal injection of IPI504 reduces the thickness of the retinal outer nuclear layer and Rlbp1b mRNA expression. Interestingly, the expression of HSP90, SP1, and CRALBP is correlatedly downregulated in the senescent ARPE-19 and Pig primary RPE cells in vitro and in the aged zebrafish and mouse retinal tissues in vivo. The aged mice exhibit the low night adaption activity. Taken together, these data indicate that the HSP90-SP1 is a novel regulatory axis of CRALBP transcriptional expression in RPE cells. The age-mediated downregulation of the HSP90-SP1-CRALBP axis is a potential etiology for the night vision reduction in senior people.
Collapse
Affiliation(s)
- Dan-Dan Chen
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Baixue Liu
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yuxuan Wang
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Mingjun Jiang
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Guohui Shang
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Mengjiao Xue
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xiaolin Jia
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - YouFei Lang
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Guiling Zhou
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Fengyan Zhang
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xuyan Peng
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yanzhong Hu
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.,The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, China
| |
Collapse
|
16
|
Lu F, Leach LL, Gross JM. A CRISPR-Cas9-mediated F0 screen to identify pro-regenerative genes in the zebrafish retinal pigment epithelium. Sci Rep 2023; 13:3142. [PMID: 36823429 PMCID: PMC9950062 DOI: 10.1038/s41598-023-29046-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/30/2023] [Indexed: 02/25/2023] Open
Abstract
Ocular diseases resulting in death of the retinal pigment epithelium (RPE) lead to vision loss and blindness. There are currently no FDA-approved strategies to restore damaged RPE cells. Stimulating intrinsic regenerative responses within damaged tissues has gained traction as a possible mechanism for tissue repair. Zebrafish possess remarkable regenerative abilities, including within the RPE; however, our understanding of the underlying mechanisms remains limited. Here, we conducted an F0 in vivo CRISPR-Cas9-mediated screen of 27 candidate RPE regeneration genes. The screen involved injection of a ribonucleoprotein complex containing three highly mutagenic guide RNAs per target gene followed by PCR-based genotyping to identify large intragenic deletions and MATLAB-based automated quantification of RPE regeneration. Through this F0 screening pipeline, eight positive and seven negative regulators of RPE regeneration were identified. Further characterization of one candidate, cldn7b, revealed novel roles in regulating macrophage/microglia infiltration after RPE injury and in clearing RPE/pigment debris during late-phase RPE regeneration. Taken together, these data support the utility of targeted F0 screens for validating pro-regenerative factors and reveal novel factors that could regulate regenerative responses within the zebrafish RPE.
Collapse
Affiliation(s)
- Fangfang Lu
- grid.21925.3d0000 0004 1936 9000Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA ,grid.452708.c0000 0004 1803 0208Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Lyndsay L. Leach
- grid.21925.3d0000 0004 1936 9000Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA ,grid.89336.370000 0004 1936 9924Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712 USA
| | - Jeffrey M. Gross
- grid.21925.3d0000 0004 1936 9000Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA ,grid.89336.370000 0004 1936 9924Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712 USA
| |
Collapse
|
17
|
Konar G, Flickinger Z, Sharma S, Vallone K, Lyon C, Doshier C, Lyon W, Patton JG. Damage-induced senescent immune cells regulate regeneration of the zebrafish retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.16.524296. [PMID: 36711649 PMCID: PMC9882244 DOI: 10.1101/2023.01.16.524296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Zebrafish spontaneously regenerate their retina in response to damage through the action of Müller glia. Even though Müller glia (MG) are conserved in higher vertebrates, the capacity to regenerate retinal damage is lost. Recent work has focused on the regulation of inflammation during tissue regeneration with precise temporal roles for macrophages and microglia. Senescent cells that have withdrawn from the cell cycle have mostly been implicated in aging, but are still metabolically active, releasing proinflammatory signaling molecules as part of the Senescence Associated Secretory Phenotype (SASP). Here, we discover that in response to retinal damage, a subset of cells expressing markers of microglia/macrophages also express markers of senescence. These cells display a temporal pattern of appearance and clearance during retina regeneration. Premature removal of senescent cells by senolytic treatment led to a decrease in proliferation and incomplete repair of the ganglion cell layer after NMDA damage. Our results demonstrate a role for modulation of senescent cell responses to balance inflammation, regeneration, plasticity, and repair as opposed to fibrosis and scarring.
Collapse
Affiliation(s)
| | | | - Shivani Sharma
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - Kyle Vallone
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - Charles Lyon
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - Claire Doshier
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - William Lyon
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - James G. Patton
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| |
Collapse
|
18
|
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: 1.5] [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
|
19
|
Frey RA, Barrett LM, Parkin L, Blakeley B, Ålund M, Byford G, Euhus A, Tsarnas C, Boughman JW, Stenkamp DL. Eye flukes (Diplostomum spp) damage retinal tissue and may cause a regenerative response in wild threespine stickleback fish. Exp Eye Res 2022; 225:109298. [PMID: 36288754 DOI: 10.1016/j.exer.2022.109298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/11/2022] [Accepted: 10/18/2022] [Indexed: 01/29/2023]
Abstract
Fish rely upon vision as a dominant sensory system for foraging, predator avoidance, and mate selection. Damage to the visual system, in particular to the neural retina of the eye, has been demonstrated to result in a regenerative response in captive fish that serve as model organisms (e.g. zebrafish), and this response restores some visual function. The purpose of the present study is to determine whether damage to the visual system that occurs in wild populations of fish also results in a regenerative response, offering a potentially ecologically relevant model of retinal regeneration. Adult threespine stickleback were collected from several water bodies of Iceland, and cryosectioned eye tissues were processed for hematoxylin and eosin staining or for indirect immunofluorescence using cell-specific markers. In many of the samples, eye flukes (metacercariae of Diplostomum spp) were present, frequently between the neural retina and retinal pigmented epithelium (RPE). Damage to the retina and to the RPE was evident in eyes containing flukes, and RPE fragments were observed within fluke bodies, suggesting they had consumed this eye tissue. Expression of a cell proliferation marker was also observed in both retina and RPE, consistent with a proliferative response to the damage. Interestingly, some regions of infected retina displayed "laminar fusions," in which neuronal cell bodies were misplaced within the major synaptic layer of the retina. These laminar fusions are also frequently found in regenerated zebrafish retina following non-parasitic (experimental) forms of retinal damage. The stickleback retina may therefore respond to fluke-mediated damage by engaging in retinal regeneration.
Collapse
Affiliation(s)
- Ruth A Frey
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Lindsey M Barrett
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Lauren Parkin
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Brittany Blakeley
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Murielle Ålund
- Department of Integrative Biology, Michigan State University, Lansing, MI, 48824, USA
| | - Gregory Byford
- Department of Integrative Biology, Michigan State University, Lansing, MI, 48824, USA
| | - Abigail Euhus
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Christine Tsarnas
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Janette W Boughman
- Department of Integrative Biology, Michigan State University, Lansing, MI, 48824, USA
| | - Deborah L Stenkamp
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA.
| |
Collapse
|
20
|
Grigoryan EN. Cell Sources for Retinal Regeneration: Implication for Data Translation in Biomedicine of the Eye. Cells 2022; 11:cells11233755. [PMID: 36497013 PMCID: PMC9738527 DOI: 10.3390/cells11233755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
The main degenerative diseases of the retina include macular degeneration, proliferative vitreoretinopathy, retinitis pigmentosa, and glaucoma. Novel approaches for treating retinal diseases are based on cell replacement therapy using a variety of exogenous stem cells. An alternative and complementary approach is the potential use of retinal regeneration cell sources (RRCSs) containing retinal pigment epithelium, ciliary body, Müller glia, and retinal ciliary region. RRCSs in lower vertebrates in vivo and in mammals mostly in vitro are able to proliferate and exhibit gene expression and epigenetic characteristics typical for neural/retinal cell progenitors. Here, we review research on the factors controlling the RRCSs' properties, such as the cell microenvironment, growth factors, cytokines, hormones, etc., that determine the regenerative responses and alterations underlying the RRCS-associated pathologies. We also discuss how the current data on molecular features and regulatory mechanisms of RRCSs could be translated in retinal biomedicine with a special focus on (1) attempts to obtain retinal neurons de novo both in vivo and in vitro to replace damaged retinal cells; and (2) investigations of the key molecular networks stimulating regenerative responses and preventing RRCS-related pathologies.
Collapse
Affiliation(s)
- Eleonora N Grigoryan
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
21
|
Choi YK. An Altered Neurovascular System in Aging-Related Eye Diseases. Int J Mol Sci 2022; 23:ijms232214104. [PMID: 36430581 PMCID: PMC9694120 DOI: 10.3390/ijms232214104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/13/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022] Open
Abstract
The eye has a complex and metabolically active neurovascular system. Repeated light injuries induce aging and trigger age-dependent eye diseases. Damage to blood vessels is related to the disruption of the blood-retinal barrier (BRB), altered cellular communication, disrupted mitochondrial functions, and exacerbated aggregated protein accumulation. Vascular complications, such as insufficient blood supply and BRB disruption, have been suggested to play a role in glaucoma, age-related macular degeneration (AMD), and Alzheimer's disease (AD), resulting in neuronal cell death. Neuronal loss can induce vision loss. In this review, we discuss the importance of the neurovascular system in the eye, especially in aging-related diseases such as glaucoma, AMD, and AD. Beneficial molecular pathways to prevent or slow down retinal pathologic processes will also be discussed.
Collapse
Affiliation(s)
- Yoon Kyung Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
22
|
Sharma P, Ramachandran R. Retina regeneration: lessons from vertebrates. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac012. [PMID: 38596712 PMCID: PMC10913848 DOI: 10.1093/oons/kvac012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/24/2022] [Accepted: 06/25/2022] [Indexed: 04/11/2024]
Abstract
Unlike mammals, vertebrates such as fishes and frogs exhibit remarkable tissue regeneration including the central nervous system. Retina being part of the central nervous system has attracted the interest of several research groups to explore its regenerative ability in different vertebrate models including mice. Fishes and frogs completely restore the size, shape and tissue structure of an injured retina. Several studies have unraveled molecular mechanisms underlying retina regeneration. In teleosts, soon after injury, the Müller glial cells of the retina reprogram to form a proliferating population of Müller glia-derived progenitor cells capable of differentiating into various neural cell types and Müller glia. In amphibians, the transdifferentiation of retinal pigment epithelium and differentiation of ciliary marginal zone cells contribute to retina regeneration. In chicks and mice, supplementation with external growth factors or genetic modifications cause a partial regenerative response in the damaged retina. The initiation of retina regeneration is achieved through sequential orchestration of gene expression through controlled modulations in the genetic and epigenetic landscape of the progenitor cells. Several developmental biology pathways are turned on during the Müller glia reprogramming, retinal pigment epithelium transdifferentiation and ciliary marginal zone differentiation. Further, several tumorigenic pathways and gene expression events also contribute to the complete regeneration cascade of events. In this review, we address the various retinal injury paradigms and subsequent gene expression events governed in different vertebrate species. Further, we compared how vertebrates such as teleost fishes and amphibians can achieve excellent regenerative responses in the retina compared with their mammalian counterparts.
Collapse
Affiliation(s)
- Poonam Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| | - Rajesh Ramachandran
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| |
Collapse
|
23
|
Martins RR, Zamzam M, Tracey-White D, Moosajee M, Thummel R, Henriques CM, MacDonald RB. Müller Glia maintain their regenerative potential despite degeneration in the aged zebrafish retina. Aging Cell 2022; 21:e13597. [PMID: 35315590 PMCID: PMC9009236 DOI: 10.1111/acel.13597] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
Ageing is a significant risk factor for degeneration of the retina. Müller glia cells (MG) are key for neuronal regeneration, so harnessing the regenerative capacity of MG in the retina offers great promise for the treatment of age-associated blinding conditions. Yet, the impact of ageing on MG regenerative capacity is unclear. Here, we show that the zebrafish retina undergoes telomerase-independent, age-related neurodegeneration but that this is insufficient to stimulate MG proliferation and regeneration. Instead, age-related neurodegeneration is accompanied by MG morphological aberrations and loss of vision. Mechanistically, yes-associated protein (Yap), part of the Hippo signalling, has been shown to be critical for the regenerative response in the damaged retina, and we show that Yap expression levels decline with ageing. Despite this, morphologically and molecularly altered aged MG retain the capacity to regenerate neurons after acute light damage, therefore, highlighting key differences in the MG response to high-intensity acute damage versus chronic neuronal loss in the zebrafish retina.
Collapse
Affiliation(s)
- Raquel R Martins
- The Bateson Centre, Healthy Lifespan Institute, MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing and Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, UK
| | - Mazen Zamzam
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | - Mariya Moosajee
- Institute of Ophthalmology, University College London, London, UK.,Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.,The Francis Crick Institute, London, UK
| | - Ryan Thummel
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Catarina M Henriques
- The Bateson Centre, Healthy Lifespan Institute, MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing and Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, UK
| | - Ryan B MacDonald
- Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
24
|
Lu F, Leach LL, Gross JM. mTOR activity is essential for retinal pigment epithelium regeneration in zebrafish. PLoS Genet 2022; 18:e1009628. [PMID: 35271573 PMCID: PMC8939802 DOI: 10.1371/journal.pgen.1009628] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 03/22/2022] [Accepted: 02/23/2022] [Indexed: 12/30/2022] Open
Abstract
The retinal pigment epithelium (RPE) plays numerous critical roles in maintaining vision and this is underscored by the prevalence of degenerative blinding diseases like age-related macular degeneration (AMD), in which visual impairment is caused by progressive loss of RPE cells. In contrast to mammals, zebrafish possess the ability to intrinsically regenerate a functional RPE layer after severe injury. The molecular underpinnings of this regenerative process remain largely unknown yet hold tremendous potential for developing treatment strategies to stimulate endogenous regeneration in the human eye. In this study, we demonstrate that the mTOR pathway is activated in RPE cells post-genetic ablation. Pharmacological and genetic inhibition of mTOR activity impaired RPE regeneration, while mTOR activation enhanced RPE recovery post-injury, demonstrating that mTOR activity is essential for RPE regeneration in zebrafish. RNA-seq of RPE isolated from mTOR-inhibited larvae identified a number of genes and pathways dependent on mTOR activity at early and late stages of regeneration; amongst these were components of the immune system, which is emerging as a key regulator of regenerative responses across various tissue and model systems. Our results identify crosstalk between macrophages/microglia and the RPE, wherein mTOR activity is required for recruitment of macrophages/microglia to the RPE injury site. Macrophages/microglia then reinforce mTOR activity in regenerating RPE cells. Interestingly, the function of macrophages/microglia in maintaining mTOR activity in the RPE appeared to be inflammation-independent. Taken together, these data identify mTOR activity as a key regulator of RPE regeneration and link the mTOR pathway to immune responses in facilitating RPE regeneration.
Collapse
Affiliation(s)
- Fangfang Lu
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lyndsay L. Leach
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jeffrey M. Gross
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
25
|
Leach LL, Fisher GB, Gross JM. Nitroreductase/Metronidazole-Mediated Ablation and a MATLAB Platform (RpEGEN) for Studying Regeneration of the Zebrafish Retinal Pigment Epithelium. J Vis Exp 2022:10.3791/63658. [PMID: 35311832 PMCID: PMC9036407 DOI: 10.3791/63658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The retinal pigment epithelium (RPE) resides at the back of the eye and performs functions essential for maintaining the health and integrity of adjacent retinal and vascular tissues. At present, the limited reparative capacity of mammalian RPE, which is restricted to small injuries, has hindered progress to understanding in vivo RPE regenerative processes. Here, a detailed methodology is provided to facilitate the study of in vivo RPE repair utilizing the zebrafish, a vertebrate model capable of robust tissue regeneration. This protocol describes a transgenic nitroreductase/metronidazole (NTR/MTZ)-mediated injury paradigm (rpe65a:nfsB-eGFP), which results in ablation of the central two-thirds of the RPE after 24 h treatment with MTZ, with subsequent tissue recovery. Focus is placed on RPE ablations in larval zebrafish and methods for testing the effects of pharmacological compounds on RPE regeneration are also outlined. Generation and validation of RpEGEN, a MATLAB script created to automate quantification of RPE regeneration based on pigmentation, is also discussed. Beyond active RPE repair mechanisms, this protocol can be expanded to studies of RPE degeneration and injury responses as well as the effects of RPE damage on adjacent retinal and vascular tissues, among other cellular and molecular processes. This zebrafish system holds significant promise in identifying genes, networks, and processes that drive RPE regeneration and RPE disease-related mechanisms, with the long-term goal of applying this knowledge to mammalian systems and, ultimately, toward therapeutic development.
Collapse
Affiliation(s)
- Lyndsay L. Leach
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine
| | - G. Burch Fisher
- Earth Research Institute, University of California, Santa Barbara
| | - Jeffrey M. Gross
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine,Department of Developmental Biology, University of Pittsburgh School of Medicine
| |
Collapse
|
26
|
Liu F, Qin Y, Huang Y, Gao P, Li J, Yu S, Jia D, Chen X, Lv Y, Tu J, Sun K, Han Y, Reilly J, Shu X, Lu Q, Tang Z, Xu C, Luo D, Liu M. Rod genesis driven by mafba in an nrl knockout zebrafish model with altered photoreceptor composition and progressive retinal degeneration. PLoS Genet 2022; 18:e1009841. [PMID: 35245286 PMCID: PMC8926279 DOI: 10.1371/journal.pgen.1009841] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 03/16/2022] [Accepted: 02/17/2022] [Indexed: 12/25/2022] Open
Abstract
Neural retina leucine zipper (NRL) is an essential gene for the fate determination and differentiation of the precursor cells into rod photoreceptors in mammals. Mutations in NRL are associated with the autosomal recessive enhanced S-cone syndrome and autosomal dominant retinitis pigmentosa. However, the exact role of Nrl in regulating the development and maintenance of photoreceptors in the zebrafish (Danio rerio), a popular animal model used for retinal degeneration and regeneration studies, has not been fully determined. In this study, we generated an nrl knockout zebrafish model via the CRISPR-Cas9 technology and observed a surprising phenotype characterized by a reduced number, but not the total loss, of rods and over-growth of green cones. We discovered two waves of rod genesis, nrl-dependent and -independent at the embryonic and post-embryonic stages, respectively, in zebrafish by monitoring the rod development. Through bulk and single-cell RNA sequencing, we characterized the gene expression profiles of the whole retina and each retinal cell type from the wild type and nrl knockout zebrafish. The over-growth of green cones and mis-expression of green-cone-specific genes in rods in nrl mutants suggested that there are rod/green-cone bipotent precursors, whose fate choice between rod versus green-cone is controlled by nrl. Besides, we identified the mafba gene as a novel regulator of the nrl-independent rod development, based on the cell-type-specific expression patterns and the retinal phenotype of nrl/mafba double-knockout zebrafish. Gene collinearity analysis revealed the evolutionary origin of mafba and suggested that the function of mafba in rod development is specific to modern fishes. Furthermore, the altered photoreceptor composition and abnormal gene expression in nrl mutants caused progressive retinal degeneration and subsequent regeneration. Accordingly, this study revealed a novel function of the mafba gene in rod development and established a working model for the developmental and regulatory mechanisms regarding the rod and green-cone photoreceptors in zebrafish. Vision is mediated by two types of light-sensing cells named rod and cone photoreceptors in animal eyes. Abnormal generation, dysfunction or death of photoreceptor cells all cause irreversible vision problems. NRL is an essential gene for the generation and function of rod cells in mice and humans. Surprisingly, we found that in the zebrafish, a popular animal model for human diseases and therapeutic testing, there are two types of rod cells, and eliminating the function of nrl gene affects the rod cell formation at the embryonic stage but not at the juvenile and adult stages. The rod cell formation at the post-embryonic is driven by the mafba gene, which has not been reported to play a role in rod cells. In addition to the reduced number of rod cells, deletion of nrl also results in the emergence of rod/green-cone hybrid cells and an increased number of green cones. The ensuing cellular and molecular alterations collectively lead to retinal degeneration. These findings expand our understanding of photoreceptor development and maintenance and highlight the underlying conserved and species-specific regulatory mechanisms.
Collapse
Affiliation(s)
- Fei Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, P.R. China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yayun Qin
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
- Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yuwen Huang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Pan Gao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jingzhen Li
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Shanshan Yu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Danna Jia
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiang Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yuexia Lv
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jiayi Tu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Kui Sun
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yunqiao Han
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - James Reilly
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, United Kingdom
| | - Xinhua Shu
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, United Kingdom
| | - Qunwei Lu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhaohui Tang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
- * E-mail: (CX); (DL); (ML)
| | - Daji Luo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, P.R. China
- University of Chinese Academy of Sciences, Beijing, China
- * E-mail: (CX); (DL); (ML)
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P.R. China
- * E-mail: (CX); (DL); (ML)
| |
Collapse
|
27
|
Chowdhury K, Lin S, Lai SL. Comparative Study in Zebrafish and Medaka Unravels the Mechanisms of Tissue Regeneration. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.783818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tissue regeneration has been in the spotlight of research for its fascinating nature and potential applications in human diseases. The trait of regenerative capacity occurs diversely across species and tissue contexts, while it seems to decline over evolution. Organisms with variable regenerative capacity are usually distinct in phylogeny, anatomy, and physiology. This phenomenon hinders the feasibility of studying tissue regeneration by directly comparing regenerative with non-regenerative animals, such as zebrafish (Danio rerio) and mice (Mus musculus). Medaka (Oryzias latipes) is a fish model with a complete reference genome and shares a common ancestor with zebrafish approximately 110–200 million years ago (compared to 650 million years with mice). Medaka shares similar features with zebrafish, including size, diet, organ system, gross anatomy, and living environment. However, while zebrafish regenerate almost every organ upon experimental injury, medaka shows uneven regenerative capacity. Their common and distinct biological features make them a unique platform for reciprocal analyses to understand the mechanisms of tissue regeneration. Here we summarize current knowledge about tissue regeneration in these fish models in terms of injured tissues, repairing mechanisms, available materials, and established technologies. We further highlight the concept of inter-species and inter-organ comparisons, which may reveal mechanistic insights and hint at therapeutic strategies for human diseases.
Collapse
|
28
|
Sherpa RD, Hui SP. An insight on established retinal injury mechanisms and prevalent retinal stem cell activation pathways in vertebrate models. Animal Model Exp Med 2021; 4:189-203. [PMID: 34557646 PMCID: PMC8446703 DOI: 10.1002/ame2.12177] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/09/2021] [Indexed: 12/22/2022] Open
Abstract
Implementing different tools and injury mechanisms in multiple animal models of retina regeneration, researchers have discovered the existence of retinal stem/progenitor cells. Although they appear to be distributed uniformly across the vertebrate lineage, the reparative potential of the retina is mainly restricted to lower vertebrates. Regenerative repair post-injury requires the creation of a proliferative niche, vital for proper stem cell activation, propagation, and lineage differentiation. This seems to be lacking in mammals. Hence, in this review, we first discuss the many forms of retinal injuries that have been generated using animal models. Next, we discuss how they are utilized to stimulate regeneration and mimic eye disease pathologies. The key to driving stem cell activation in mammals relies on the information we can gather from these models. Lastly, we present a brief update about the genes, growth factors, and signaling pathways that have been brought to light using these models.
Collapse
Affiliation(s)
| | - Subhra Prakash Hui
- S. N. Pradhan Centre for NeurosciencesUniversity of CalcuttaKolkataIndia
| |
Collapse
|
29
|
The immune response is a critical regulator of zebrafish retinal pigment epithelium regeneration. Proc Natl Acad Sci U S A 2021; 118:2017198118. [PMID: 34006636 DOI: 10.1073/pnas.2017198118] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Loss of the retinal pigment epithelium (RPE) because of dysfunction or disease can lead to blindness in humans. Harnessing the intrinsic ability of the RPE to self-repair is an attractive therapeutic strategy; however, mammalian RPE is limited in its regenerative capacity. Zebrafish possess tremendous intrinsic regenerative potential in ocular tissues, including the RPE, but little is known about the mechanisms driving RPE regeneration. Here, utilizing transgenic and mutant zebrafish lines, pharmacological manipulations, transcriptomics, and imaging analyses, we identified elements of the immune response as critical mediators of intrinsic RPE regeneration. After genetic ablation, the RPE express immune-related genes, including leukocyte recruitment factors such as interleukin 34 We demonstrate that macrophage/microglia cells are responsive to RPE damage and that their function is required for the timely progression of the regenerative response. These data identify the molecular and cellular underpinnings of RPE regeneration and hold significant potential for translational approaches aimed toward promoting a pro-regenerative environment in mammalian RPE.
Collapse
|
30
|
George SM, Lu F, Rao M, Leach LL, Gross JM. The retinal pigment epithelium: Development, injury responses, and regenerative potential in mammalian and non-mammalian systems. Prog Retin Eye Res 2021; 85:100969. [PMID: 33901682 DOI: 10.1016/j.preteyeres.2021.100969] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/23/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022]
Abstract
Diseases that result in retinal pigment epithelium (RPE) degeneration, such as age-related macular degeneration (AMD), are among the leading causes of blindness worldwide. Atrophic (dry) AMD is the most prevalent form of AMD and there are currently no effective therapies to prevent RPE cell death or restore RPE cells lost from AMD. An intriguing approach to treat AMD and other RPE degenerative diseases is to develop therapies focused on stimulating endogenous RPE regeneration. For this to become feasible, a deeper understanding of the mechanisms underlying RPE development, injury responses and regenerative potential is needed. In mammals, RPE regeneration is extremely limited; small lesions can be repaired by the expansion of adjacent RPE cells, but large lesions cannot be repaired as remaining RPE cells are unable to functionally replace lost RPE tissue. In some injury paradigms, RPE cells proliferate but do not regenerate a morphologically normal monolayer, while in others, proliferation is pathogenic and results in further disruption to the retina. This is in contrast to non-mammalian vertebrates, which possess tremendous RPE regenerative potential. Here, we discuss what is known about RPE formation during development in mammalian and non-mammalian vertebrates, we detail the processes by which RPE cells respond to injury, and we describe examples of RPE-to-retina and RPE-to-RPE regeneration in non-mammalian vertebrates. Finally, we outline barriers to RPE-dependent regeneration in mammals that could potentially be overcome to stimulate a regenerative response from the RPE.
Collapse
Affiliation(s)
- Stephanie M George
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Fangfang Lu
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Mishal Rao
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Lyndsay L Leach
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Jeffrey M Gross
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
31
|
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: 5.0] [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
|
32
|
Noel NCL, MacDonald IM, Allison WT. Zebrafish Models of Photoreceptor Dysfunction and Degeneration. Biomolecules 2021; 11:78. [PMID: 33435268 PMCID: PMC7828047 DOI: 10.3390/biom11010078] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
Zebrafish are an instrumental system for the generation of photoreceptor degeneration models, which can be utilized to determine underlying causes of photoreceptor dysfunction and death, and for the analysis of potential therapeutic compounds, as well as the characterization of regenerative responses. We review the wealth of information from existing zebrafish models of photoreceptor disease, specifically as they relate to currently accepted taxonomic classes of human rod and cone disease. We also highlight that rich, detailed information can be derived from studying photoreceptor development, structure, and function, including behavioural assessments and in vivo imaging of zebrafish. Zebrafish models are available for a diversity of photoreceptor diseases, including cone dystrophies, which are challenging to recapitulate in nocturnal mammalian systems. Newly discovered models of photoreceptor disease and drusenoid deposit formation may not only provide important insights into pathogenesis of disease, but also potential therapeutic approaches. Zebrafish have already shown their use in providing pre-clinical data prior to testing genetic therapies in clinical trials, such as antisense oligonucleotide therapy for Usher syndrome.
Collapse
Affiliation(s)
- Nicole C. L. Noel
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada; (I.M.M.); (W.T.A.)
| | - Ian M. MacDonald
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada; (I.M.M.); (W.T.A.)
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - W. Ted Allison
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada; (I.M.M.); (W.T.A.)
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8, Canada
| |
Collapse
|
33
|
Cassar S, Dunn C, Ramos MF. Zebrafish as an Animal Model for Ocular Toxicity Testing: A Review of Ocular Anatomy and Functional Assays. Toxicol Pathol 2020; 49:438-454. [PMID: 33063651 DOI: 10.1177/0192623320964748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Xenobiotics make their way into organisms from diverse sources including diet, medication, and pollution. Our understanding of ocular toxicities from xenobiotics in humans, livestock, and wildlife is growing thanks to laboratory animal models. Anatomy and physiology are conserved among vertebrate eyes, and studies with common mammalian preclinical species (rodent, dog) can predict human ocular toxicity. However, since the eye is susceptible to toxicities that may not involve a histological correlate, and these species rely heavily on smell and hearing to navigate their world, discovering visual deficits can be challenging with traditional animal models. Alternative models capable of identifying functional impacts on vision and requiring minimal amounts of chemical are valuable assets to toxicology. Human and zebrafish eyes are anatomically and functionally similar, and it has been reported that several common human ocular toxicants cause comparable toxicity in zebrafish. Vision develops rapidly in zebrafish; the tiny larvae rely on visual cues as early as 4 days, and behavioral responses to those cues can be monitored in high-throughput fashion. This article describes the comparative anatomy of the zebrafish eye, the notable differences from the mammalian eye, and presents practical applications of this underutilized model for assessment of ocular toxicity.
Collapse
Affiliation(s)
- Steven Cassar
- Preclinical Safety, 419726AbbVie, Inc, North Chicago, IL, USA
| | - Christina Dunn
- Preclinical Safety, 419726AbbVie, Inc, North Chicago, IL, USA
| | | |
Collapse
|
34
|
Grigoryan EN. Potential Endogenous Cell Sources for Retinal Regeneration in Vertebrates and Humans: Progenitor Traits and Specialization. Biomedicines 2020; 8:E208. [PMID: 32664635 PMCID: PMC7400588 DOI: 10.3390/biomedicines8070208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/04/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022] Open
Abstract
Retinal diseases often cause the loss of photoreceptor cells and, consequently, impairment of vision. To date, several cell populations are known as potential endogenous retinal regeneration cell sources (RRCSs): the eye ciliary zone, the retinal pigment epithelium, the iris, and Müller glia. Factors that can activate the regenerative responses of RRCSs are currently under investigation. The present review considers accumulated data on the relationship between the progenitor properties of RRCSs and the features determining their differentiation. Specialized RRCSs (all except the ciliary zone in low vertebrates), despite their differences, appear to be partially "prepared" to exhibit their plasticity and be reprogrammed into retinal neurons due to the specific gene expression and epigenetic landscape. The "developmental" characteristics of RRCS gene expression are predefined by the pathway by which these cell populations form during eye morphogenesis; the epigenetic features responsible for chromatin organization in RRCSs are under intracellular regulation. Such genetic and epigenetic readiness is manifested in vivo in lower vertebrates and in vitro in higher ones under conditions permissive for cell phenotype transformation. Current studies on gene expression in RRCSs and changes in their epigenetic landscape help find experimental approaches to replacing dead cells through recruiting cells from endogenous resources in vertebrates and humans.
Collapse
Affiliation(s)
- Eleonora N Grigoryan
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
35
|
Rastoin O, Pagès G, Dufies M. Experimental Models in Neovascular Age Related Macular Degeneration. Int J Mol Sci 2020; 21:ijms21134627. [PMID: 32610682 PMCID: PMC7370120 DOI: 10.3390/ijms21134627] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/25/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Neovascular age-related macular degeneration (vAMD), characterized by the neo-vascularization of the retro-foveolar choroid, leads to blindness within few years. This disease depends on angiogenesis mediated by the vascular endothelial growth factor A (VEGF) and to inflammation. The only available treatments consist of monthly intravitreal injections of antibodies directed against VEGF or VEGF/VEGFB/PlGF decoy receptors. Despite their relative efficacy, these drugs only delay progression to blindness and 30% of the patients are insensitive to these treatments. Hence, new therapeutic strategies are urgently needed. Experimental models of vAMD are essential to screen different innovative therapeutics. The currently used in vitro and in vivo models in ophthalmic translational research and their relevance are discussed in this review.
Collapse
Affiliation(s)
- Olivia Rastoin
- Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, University Cote d’Azur (UCA), 06000 Nice, France; (O.R.); (G.P.)
| | - Gilles Pagès
- Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, University Cote d’Azur (UCA), 06000 Nice, France; (O.R.); (G.P.)
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Maeva Dufies
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco
- Correspondence:
| |
Collapse
|
36
|
Storm T, Wilson I, Campbell R, Bolinches-Amorós A, Russell AJ, Davies SG, Barnard AR, MacLaren RE. A Semiautomated, Phenotypic, In Vitro Scratch Assay for Assessing Retinal Pigment Epithelial Cell Wound Healing. J Ocul Pharmacol Ther 2020; 36:257-266. [PMID: 32027217 DOI: 10.1089/jop.2019.0116] [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: 11/12/2022] Open
Abstract
Purpose: Age-related macular degeneration leads to retinal pigment epithelium (RPE) cell death and loss of central vision. In vivo studies have shown that the RPE layer has an innate, but limited, ability to repopulate atrophic areas. We aimed to establish a semiautomated, in vitro, wound healing assay workflow for targeted screening of compounds able to influence RPE wound healing. Methods: The ARPE-19 phenotype was evaluated using bright-field microscopy, immunocytochemistry, and quantitative real-time polymerase chain reaction. ARPE-19 monolayers were simultaneously scratched in a 96-well format and treated with Hoechst-33342 and an array of compounds. Initial wound dimensions and wound healing were subsequently evaluated using the EVOS FL Auto 2.0 imaging platform combined with automated image analyses. Results: Long-term cultured ARPE-19 cells displayed a more in vivo RPE-like phenotype compared with recently seeded or short-term cultured cells. No statistical difference of initial scratch width was observed between short-term and long-term cultured cells, but more wells were excluded from analyses in total in the latter case due to scratch width, scratch smoothness, and imaging errors. Furthermore, the previous time spent in continuous culture had an effect on the observation of an altered wound healing response to different treatment conditions. Conclusions: We have established a semiautomated, 96-well format, in vitro wound healing assay with a reproducible workflow. This would enable screening of a significant number of compounds and greatly advances the potential of identifying novel therapeutics that may enhance the innate ability of RPE cells to repopulate atrophic areas.
Collapse
Affiliation(s)
- Tina Storm
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Iain Wilson
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ross Campbell
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Arantxa Bolinches-Amorós
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Angela J Russell
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom.,Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Stephen G Davies
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Alun R Barnard
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom.,Oxford Eye Hospital, Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom.,Oxford Eye Hospital, Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
37
|
Ma L, Ng M, van der Weele CM, Yoshizawa M, Jeffery WR. Dual roles of the retinal pigment epithelium and lens in cavefish eye degeneration. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2020; 334:438-449. [PMID: 31930686 DOI: 10.1002/jez.b.22923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/04/2019] [Accepted: 12/21/2019] [Indexed: 01/03/2023]
Abstract
Astyanax mexicanus consists of two forms, a sighted surface dwelling form (surface fish) and a blind cave-dwelling form (cavefish). Embryonic eyes are initially formed in cavefish but they are subsequently arrested in growth and degenerate during larval development. Previous lens transplantation studies have shown that the lens plays a central role in cavefish eye loss. However, several lines of evidence suggest that additional factors, such as the retinal pigment epithelium (RPE), which is morphologically altered in cavefish, could also be involved in the eye regression process. To explore the role of the RPE in cavefish eye degeneration, we generated an albino eyed (AE) strain by artificial selection for hybrid individuals with large eyes and a depigmented RPE. The AE strain exhibited an RPE lacking pigment granules and showed reduced expression of the RPE specific enzyme retinol isomerase, allowing eye development to be studied by lens ablation in an RPE background resembling cavefish. We found that lens ablation in the AE strain had stronger negative effects on eye growth than in surface fish, suggesting that an intact RPE is required for normal eye development. We also found that the AE strain develops a cartilaginous sclera lacking boney ossicles, a trait similar to cavefish. Extrapolation of the results to cavefish suggests that the RPE and lens have dual roles in eye degeneration, and that deficiencies in the RPE may be associated with evolutionary changes in scleral ossification.
Collapse
Affiliation(s)
- Li Ma
- Department of Biology, University of Maryland, College Park, Maryland
| | - Mandy Ng
- Department of Biology, University of Maryland, College Park, Maryland
| | | | - Masato Yoshizawa
- Department of Biology, University of Maryland, College Park, Maryland
| | - William R Jeffery
- Department of Biology, University of Maryland, College Park, Maryland
| |
Collapse
|
38
|
Patterson M, Swift SK. Residual Diploidy in Polyploid Tissues: A Cellular State with Enhanced Proliferative Capacity for Tissue Regeneration? Stem Cells Dev 2019; 28:1527-1539. [PMID: 31608782 PMCID: PMC11001963 DOI: 10.1089/scd.2019.0193] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/09/2019] [Indexed: 01/07/2023] Open
Abstract
A major objective of modern biomedical research aims to promote tissue self-regeneration after injury, obviating the need for whole organ transplantation and avoiding mortality due to organ failure. Identifying the population of cells capable of regeneration, alongside understanding the molecular mechanisms that activate that population to re-enter the cell cycle, are two important steps to advancing the field of endogenous tissue regeneration toward the clinic. In recent years, an emerging trend has been observed, whereby polyploidy of relevant parenchymal cells, arising from alternative cell cycles as part of a normal developmental process, is linked to restricted proliferative capacity of those cells. An accompanying hypothesis, therefore, is that a residual subpopulation of diploid parenchymal cells retains proliferative competence and is the major driver for any detected postnatal cell turnover. In this perspective review, we examine the emerging literature on residual diploid parenchymal cells and the possible link of this population to endogenous tissue regeneration, in the context of both heart and liver. We speculate on additional cell types that may play a similar role in their respective tissues and discuss outstanding questions for the field.
Collapse
Affiliation(s)
- Michaela Patterson
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Samantha K. Swift
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
39
|
Xia QQ, Zhang LM, Zhou YY, Wu YL, Li J. All-trans-retinoic acid generation is an antidotal clearance pathway for all-trans-retinal in the retina. J Zhejiang Univ Sci B 2019; 20:960-971. [PMID: 31749343 DOI: 10.1631/jzus.b1900271] [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: 11/11/2022]
Abstract
The present study was designed to analyze the metabolites of all-trans-retinal (atRal) and compare the cytotoxicity of atRal versus its derivative all-trans-retinoic acid (atRA) in human retinal pigment epithelial (RPE) cells. We confirmed that atRA was produced in normal pig neural retina and RPE. The amount of all-trans-retinol (atROL) converted from atRal was about 2.7 times that of atRal-derived atRA after incubating RPE cells with 10 μmol/L atRal for 24 h, whereas atRA in medium supernatant is more plentiful (91 vs. 29 pmol/mL), suggesting that atRA conversion facilitates elimination of excess atRal in the retina. Moreover, we found that mRNA expression of retinoic acid-specific hydroxylase CYP26b1 was dose-dependently up-regulated by atRal exposure in RPE cells, indicating that atRA inactivation may be also initiated in atRal-accumulated RPE cells. Our data show that atRA-caused viability inhibition was evidently reduced compared with the equal concentration of its precursor atRal. Excess accumulation of atRal provoked intracellular reactive oxygen species (ROS) overproduction, heme oxygenase-1 (HO-1) expression, and increased cleaved poly(ADP-ribose) polymerase 1 (PARP1) expression in RPE cells. In contrast, comparable dosage of atRA-induced oxidative stress was much weaker, and it could not activate apoptosis in RPE cells. These results suggest that atRA generation is an antidotal metabolism pathway for atRal in the retina. Moreover, we found that in the eyes of ABCA4-/-RDH8-/- mice, a mouse model with atRal accumulation in the retina, the atRA content was almost the same as that in the wild type. It is possible that atRal accumulation simultaneously and equally promotes atRA synthesis and clearance in eyes of ABCA4-/-RDH8-/- mice, thus inhibiting the further increase of atRA in the retina. Our present study provides further insights into atRal clearance in the retina.
Collapse
Affiliation(s)
- Qing-Qing Xia
- Central Laboratory, Department of Laboratory Medicine, Huangyan Hospital of Wenzhou Medical University, Taizhou First People's Hospital, Taizhou 318020, China
| | - Ling-Min Zhang
- Central Laboratory, Department of Laboratory Medicine, Huangyan Hospital of Wenzhou Medical University, Taizhou First People's Hospital, Taizhou 318020, China
| | - Ying-Ying Zhou
- Central Laboratory, Department of Laboratory Medicine, Huangyan Hospital of Wenzhou Medical University, Taizhou First People's Hospital, Taizhou 318020, China
| | - Ya-Lin Wu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jie Li
- Central Laboratory, Department of Laboratory Medicine, Huangyan Hospital of Wenzhou Medical University, Taizhou First People's Hospital, Taizhou 318020, China
| |
Collapse
|