1
|
Goyal S, Tibrewal S, Ratna R, Vanita V. Genetic and environmental factors contributing to anophthalmia and microphthalmia: Current understanding and future directions. World J Clin Pediatr 2025; 14:101982. [DOI: 10.5409/wjcp.v14.i2.101982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/18/2025] Open
Abstract
Anophthalmia is defined as a complete absence of one eye or both the eyes, while microphthalmia represents the presence of a small eye within the orbit. The estimated birth prevalence for anophthalmia is approximately 3 per 100000 live births, and for microphthalmia, it is around 14 per 100000 live births. However, combined evidence suggests that the prevalence of these malformations could be as high as 30 per 100000 individuals. Microphthalmia is reported to occur in 3.2% to 11.2% of blind children. Anophthalmia and microphthalmia (A/M) are part of a phenotypic spectrum alongside ocular coloboma, hypothesized to share a common genetic basis. Both A/M can occur in isolation or as part of a syndrome. Their complex etiology involves chromosomal aberrations, monogenic inheritance pattern, and the contribution of environmental factors such as gestational-acquired infections, maternal vitamin A deficiency (VAD), exposure to X-rays, solvent misuse, and thalidomide exposure. A/M exhibit significant clinical and genetic heterogeneity with over 90 genes identified so far. Familial cases of A/M have a complex genetic basis, including all Mendelian modes of inheritance, i.e., autosomal dominant, recessive, and X-linked. Most cases arise sporadically due to de novo mutations. Examining gene expression during eye development and the effects of various environmental variables will help us better understand the phenotypic heterogeneity found in A/M, leading to more effective diagnosis and management strategies. The present review focuses on key genetic factors, developmental abnormalities, and environmental modifiers linked with A/M. It also emphasizes at potential research areas including multiomic methods and disease modeling with induced pluripotent stem cell technologies, which aim to create innovative treatment options.
Collapse
Affiliation(s)
- Shiwali Goyal
- Department of Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Rockville, MD 20852, United States
| | - Shailja Tibrewal
- Department of Pediatric Ophthalmology, Dr. Shroff’s Charity Eye Hospital, New Delhi 110002, Delhi, India
- Department of Ocular Genetics (Center for Unknown and Rare Eye Diseases), Dr. Shroff’s Charity Eye Hospital, New Delhi 110002, Delhi, India
| | - Ria Ratna
- Department of Ocular Genetics (Center for Unknown and Rare Eye Diseases), Dr. Shroff’s Charity Eye Hospital, New Delhi 110002, Delhi, India
| | - Vanita Vanita
- Department of Human Genetics, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| |
Collapse
|
2
|
Tignard P, Pottin K, Geeverding A, Doulazmi M, Cabrera M, Fouquet C, Liffran M, Fouchard J, Rosello M, Albadri S, Del Bene F, Trembleau A, Breau MA. Basement membranes are crucial for proper olfactory placode shape, position and boundary with the brain, and for olfactory axon development. eLife 2024; 12:RP92004. [PMID: 39713923 DOI: 10.7554/elife.92004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
Despite recent progress, the complex roles played by the extracellular matrix in development and disease are still far from being fully understood. Here, we took advantage of the zebrafish sly mutation which affects Laminin γ1, a major component of basement membranes, to explore its role in the development of the olfactory system. Following a detailed characterisation of Laminin distribution in the developing olfactory circuit, we analysed basement membrane integrity, olfactory placode and brain morphogenesis, and olfactory axon development in sly mutants, using a combination of immunochemistry, electron microscopy and quantitative live imaging of cell movements and axon behaviours. Our results point to an original and dual contribution of Laminin γ1-dependent basement membranes in organising the border between the olfactory placode and the adjacent brain: they maintain placode shape and position in the face of major brain morphogenetic movements, they establish a robust physical barrier between the two tissues while at the same time allowing the local entry of the sensory axons into the brain and their navigation towards the olfactory bulb. This work thus identifies key roles of Laminin γ1-dependent basement membranes in neuronal tissue morphogenesis and axon development in vivo.
Collapse
Affiliation(s)
- Pénélope Tignard
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR8246), Inserm U1130, Institut de Biologie Paris-Seine (IBPS), Neuroscience Paris Seine (NPS), Paris, France
| | - Karen Pottin
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
| | - Audrey Geeverding
- Imaging Facility, Institut de Biologie Paris-Seine (IBPS), Paris, France
| | - Mohamed Doulazmi
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR8256), Institut de Biologie Paris-Seine (IBPS), Adaptation Biologique et Vieillissement, Paris, France
| | - Mélody Cabrera
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
| | - Coralie Fouquet
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
| | - Mathilde Liffran
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR8246), Inserm U1130, Institut de Biologie Paris-Seine (IBPS), Neuroscience Paris Seine (NPS), Paris, France
| | - Jonathan Fouchard
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
| | - Marion Rosello
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Shahad Albadri
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Filippo Del Bene
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Alain Trembleau
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR8246), Inserm U1130, Institut de Biologie Paris-Seine (IBPS), Neuroscience Paris Seine (NPS), Paris, France
| | - Marie Anne Breau
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| |
Collapse
|
3
|
Tignard P, Pottin K, Geeverding A, Doulazmi M, Cabrera M, Fouquet C, Liffran M, Fouchard J, Rosello M, Albadri S, Del Bene F, Trembleau A, Breau MA. Laminin γ1-dependent basement membranes are instrumental to ensure proper olfactory placode shape, position and boundary with the brain, as well as olfactory axon development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.29.547040. [PMID: 39253416 PMCID: PMC11383033 DOI: 10.1101/2023.06.29.547040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Despite recent progress, the complex roles played by the extracellular matrix in development and disease are still far from being fully understood. Here, we took advantage of the zebrafish sly mutation which affects Laminin γ1, a major component of basement membranes, to explore its role in the development of the olfactory system. Following a detailed characterisation of Laminin distribution in the developing olfactory circuit, we analysed basement membrane integrity, olfactory placode and brain morphogenesis, and olfactory axon development in sly mutants, using a combination of immunochemistry, electron microscopy and quantitative live imaging of cell movements and axon behaviours. Our results point to an original and dual contribution of Laminin γ1-dependent basement membranes in organising the border between the olfactory placode and the adjacent brain: they maintain placode shape and position in the face of major brain morphogenetic movements, they establish a robust physical barrier between the two tissues while at the same time allowing the local entry of the sensory axons into the brain and their navigation towards the olfactory bulb. This work thus identifies key roles of Laminin γ1-dependent basement membranes in neuronal tissue morphogenesis and axon development in vivo .
Collapse
|
4
|
Li J, Ma J, Chen Y, Chen S, Luo L, Cheng H. Biologically Relevant Laminin-511 Moderates the Derivation and Proliferation of Human Lens Epithelial Stem/Progenitor-Like Cells. Invest Ophthalmol Vis Sci 2024; 65:12. [PMID: 39106056 PMCID: PMC11309036 DOI: 10.1167/iovs.65.10.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 04/06/2024] [Indexed: 08/07/2024] Open
Abstract
Purpose The role of specific extracellular matrix (ECM) molecules in lens cell development and regeneration is poorly understood, as appropriate cellular models are lacking. Here, a laminin-based lens cell in vitro induction system was developed to study the role of laminin in human lens epithelial stem/progenitor cell (LES/PC) development. Methods The human embryonic stem cell-based lens induction system followed a three-stage protocol. The expression profile of laminins during lens induction was screened, and laminin-511 (LN511) was tested as a candidate substitute. LN511 induction system cellular and molecular features, including induction efficiency, transcription factor expression related to different lens development stages, ECM alterations, and Hippo/YAP signaling, were evaluated. Results LAMA5, LAMB1, and LAMC1 were highly expressed around the time of LES/PC derivation. We chose LN511 (product of LAMA5, LAMB1, and LAMC1) and found that it considerably enhanced lens cell induction efficiency, compared to that in Matrigel-coated culture, as more and larger lentoid bodies were detected. Notably, LES/PC induction efficiency improved by promoting lens specification-related transcription factor expression and cell proliferation. Transcriptome analysis revealed that compared to those with Matrigel, ECM accumulation and cell adhesion were downregulated in the LN511 system. Hippo/YAP signaling was hypoactive during LES/P-like cell generation, and small molecule inhibitors of YAP/TAZ activity upregulated LES/PC marker expression and promoted the efficiency of LES/P-like cell derivation. Conclusions The laminin isoform LN511 is a reliable substitute for the LES/P-like cell induction system, and LN511-YAP acted as efficient modulators of LES/PC derivation; this contributes to knowledge of the role of the ECM in human lens development.
Collapse
Affiliation(s)
- Jinyan Li
- Department of Ophthalmology, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jingyu Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yijia Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shuyi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lixia Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hao Cheng
- Department of Ophthalmology, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Eintracht J, Owen N, Harding P, Moosajee M. Disruption of common ocular developmental pathways in patient-derived optic vesicle models of microphthalmia. Stem Cell Reports 2024; 19:839-858. [PMID: 38821055 PMCID: PMC11390689 DOI: 10.1016/j.stemcr.2024.05.001] [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: 09/13/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/02/2024] Open
Abstract
Genetic perturbations influencing early eye development can result in microphthalmia, anophthalmia, and coloboma (MAC). Over 100 genes are associated with MAC, but little is known about common disease mechanisms. In this study, we generated induced pluripotent stem cell (iPSC)-derived optic vesicles (OVs) from two unrelated microphthalmia patients and healthy controls. At day 20, 35, and 50, microphthalmia patient OV diameters were significantly smaller, recapitulating the "small eye" phenotype. RNA sequencing (RNA-seq) analysis revealed upregulation of apoptosis-initiating and extracellular matrix (ECM) genes at day 20 and 35. Western blot and immunohistochemistry revealed increased expression of lumican, nidogen, and collagen type IV, suggesting ECM overproduction. Increased apoptosis was observed in microphthalmia OVs with reduced phospho-histone 3 (pH3+) cells confirming decreased cell proliferation at day 35. Pharmacological inhibition of caspase-8 activity with Z-IETD-FMK decreased apoptosis in one patient model, highlighting a potential therapeutic approach. These data reveal shared pathophysiological mechanisms contributing to a microphthalmia phenotype.
Collapse
Affiliation(s)
| | | | | | - Mariya Moosajee
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; Moorfields Eye Hospital NHS Foundation Trust, London EC1V 9EL, UK; Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
6
|
Casey MA, Lusk S, Kwan KM. Eye Morphogenesis in Vertebrates. Annu Rev Vis Sci 2023; 9:221-243. [PMID: 37040791 DOI: 10.1146/annurev-vision-100720-111125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Proper eye structure is essential for visual function: Multiple essential eye tissues must take shape and assemble into a precise three-dimensional configuration. Accordingly, alterations to eye structure can lead to pathological conditions of visual impairment. Changes in eye shape can also be adaptive over evolutionary time. Eye structure is first established during development with the formation of the optic cup, which contains the neural retina, retinal pigment epithelium, and lens. This crucial yet deceptively simple hemispherical structure lays the foundation for all later elaborations of the eye. Building on descriptions of the embryonic eye that started with hand drawings and micrographs, the field is beginning to identify mechanisms driving dynamic changes in three-dimensional cell and tissue shape. A combination of molecular genetics, imaging, and pharmacological approaches is defining connections among transcription factors, signaling pathways, and the intracellular machinery governing the emergence of this crucial structure.
Collapse
Affiliation(s)
- Macaulie A Casey
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA; , ,
| | - Sarah Lusk
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA; , ,
| | - Kristen M Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA; , ,
| |
Collapse
|
7
|
Cardozo MJ, Sánchez-Bustamante E, Bovolenta P. Optic cup morphogenesis across species and related inborn human eye defects. Development 2023; 150:dev200399. [PMID: 36714981 PMCID: PMC10110496 DOI: 10.1242/dev.200399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The vertebrate eye is shaped as a cup, a conformation that optimizes vision and is acquired early in development through a process known as optic cup morphogenesis. Imaging living, transparent teleost embryos and mammalian stem cell-derived organoids has provided insights into the rearrangements that eye progenitors undergo to adopt such a shape. Molecular and pharmacological interference with these rearrangements has further identified the underlying molecular machineries and the physical forces involved in this morphogenetic process. In this Review, we summarize the resulting scenarios and proposed models that include common and species-specific events. We further discuss how these studies and those in environmentally adapted blind species may shed light on human inborn eye malformations that result from failures in optic cup morphogenesis, including microphthalmia, anophthalmia and coloboma.
Collapse
Affiliation(s)
- Marcos J. Cardozo
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
| | - Elena Sánchez-Bustamante
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
| |
Collapse
|
8
|
Laminin-111 mutant studies reveal a hierarchy within laminin-111 genes in their requirement for basal epithelial tissue folding. Dev Biol 2022; 492:172-186. [DOI: 10.1016/j.ydbio.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/21/2022]
|
9
|
Collective cell migration during optic cup formation features changing cell-matrix interactions linked to matrix topology. Curr Biol 2022; 32:4817-4831.e9. [PMID: 36208624 DOI: 10.1016/j.cub.2022.09.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/28/2022] [Accepted: 09/16/2022] [Indexed: 11/22/2022]
Abstract
Cell migration is crucial for organismal development and shapes organisms in health and disease. Although a lot of research has revealed the role of intracellular components and extracellular signaling in driving single and collective cell migration, the influence of physical properties of the tissue and the environment on migration phenomena in vivo remains less explored. In particular, the role of the extracellular matrix (ECM), which many cells move upon, is currently unclear. To overcome this gap, we use zebrafish optic cup formation, and by combining novel transgenic lines and image analysis pipelines, we study how ECM properties influence cell migration in vivo. We show that collectively migrating rim cells actively move over an immobile extracellular matrix. These cell movements require cryptic lamellipodia that are extended in the direction of migration. Quantitative analysis of matrix properties revealed that the topology of the matrix changes along the migration path. These changes in matrix topologies are accompanied by changes in the dynamics of cell-matrix interactions. Experiments and theoretical modeling suggest that matrix porosity could be linked to efficient migration. Indeed, interfering with matrix topology by increasing its porosity results in a loss of cryptic lamellipodia, less-directed cell-matrix interactions, and overall inefficient migration. Thus, matrix topology is linked to the dynamics of cell-matrix interactions and the efficiency of directed collective rim cell migration during vertebrate optic cup morphogenesis.
Collapse
|
10
|
Monnot P, Gangatharan G, Baraban M, Pottin K, Cabrera M, Bonnet I, Breau MA. Intertissue mechanical interactions shape the olfactory circuit in zebrafish. EMBO Rep 2022; 23:e52963. [PMID: 34889034 PMCID: PMC8811657 DOI: 10.15252/embr.202152963] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 02/05/2023] Open
Abstract
While the chemical signals guiding neuronal migration and axon elongation have been extensively studied, the influence of mechanical cues on these processes remains poorly studied in vivo. Here, we investigate how mechanical forces exerted by surrounding tissues steer neuronal movements and axon extension during the morphogenesis of the olfactory placode in zebrafish. We mainly focus on the mechanical contribution of the adjacent eye tissue, which develops underneath the placode through extensive evagination and invagination movements. Using quantitative analysis of cell movements and biomechanical manipulations, we show that the developing eye exerts lateral traction forces on the olfactory placode through extracellular matrix, mediating proper morphogenetic movements and axon extension within the placode. Our data shed new light on the key participation of intertissue mechanical interactions in the sculpting of neuronal circuits.
Collapse
Affiliation(s)
- Pauline Monnot
- Centre National de la Recherche Scientifique (CNRS)Institut de Biologie Paris‐Seine (IBPS)Developmental Biology LaboratorySorbonne UniversitéParisFrance
- Institut CurieUniversité PSLSorbonne UniversitéCNRS UMR168Laboratoire Physico Chimie CurieParisFrance
- Laboratoire Jean PerrinParisFrance
| | - Girisaran Gangatharan
- Centre National de la Recherche Scientifique (CNRS)Institut de Biologie Paris‐Seine (IBPS)Developmental Biology LaboratorySorbonne UniversitéParisFrance
| | - Marion Baraban
- Centre National de la Recherche Scientifique (CNRS)Institut de Biologie Paris‐Seine (IBPS)Developmental Biology LaboratorySorbonne UniversitéParisFrance
- Laboratoire Jean PerrinParisFrance
| | - Karen Pottin
- Centre National de la Recherche Scientifique (CNRS)Institut de Biologie Paris‐Seine (IBPS)Developmental Biology LaboratorySorbonne UniversitéParisFrance
| | - Melody Cabrera
- Centre National de la Recherche Scientifique (CNRS)Institut de Biologie Paris‐Seine (IBPS)Developmental Biology LaboratorySorbonne UniversitéParisFrance
| | - Isabelle Bonnet
- Institut CurieUniversité PSLSorbonne UniversitéCNRS UMR168Laboratoire Physico Chimie CurieParisFrance
| | - Marie Anne Breau
- Centre National de la Recherche Scientifique (CNRS)Institut de Biologie Paris‐Seine (IBPS)Developmental Biology LaboratorySorbonne UniversitéParisFrance
- Laboratoire Jean PerrinParisFrance
- Institut National de la Santé et de la Recherche Médicale (INSERM)ParisFrance
| |
Collapse
|
11
|
Moreno-Mármol T, Ledesma-Terrón M, Tabanera N, Martin-Bermejo MJ, Cardozo MJ, Cavodeassi F, Bovolenta P. Stretching of the retinal pigment epithelium contributes to zebrafish optic cup morphogenesis. eLife 2021; 10:63396. [PMID: 34545806 PMCID: PMC8530511 DOI: 10.7554/elife.63396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
The vertebrate eye primordium consists of a pseudostratified neuroepithelium, the optic vesicle (OV), in which cells acquire neural retina or retinal pigment epithelium (RPE) fates. As these fates arise, the OV assumes a cup shape, influenced by mechanical forces generated within the neural retina. Whether the RPE passively adapts to retinal changes or actively contributes to OV morphogenesis remains unexplored. We generated a zebrafish Tg(E1-bhlhe40:GFP) line to track RPE morphogenesis and interrogate its participation in OV folding. We show that, in virtual absence of proliferation, RPE cells stretch and flatten, thereby matching the retinal curvature and promoting OV folding. Localized interference with the RPE cytoskeleton disrupts tissue stretching and OV folding. Thus, extreme RPE flattening and accelerated differentiation are efficient solutions adopted by fast-developing species to enable timely optic cup formation. This mechanism differs in amniotes, in which proliferation drives RPE expansion with a much-reduced need of cell flattening. Rounded eyeballs help to optimize vision – but how do they acquire their distinctive shape? In animals with backbones, including humans, the eye begins to form early in development. A single layer of embryonic tissue called the optic vesicle reorganizes itself into a two-layered structure: a thin outer layer of cells, known as the retinal pigmented epithelium (RPE for short), and a thicker inner layer called the neural retina. If this process fails, the animal may be born blind or visually impaired. How this flat two-layered structure becomes round is still being investigated. In fish, studies have shown that the inner cell layer – the neural retina – generates mechanical forces that cause the developing tissue to curve inwards to form a cup-like shape. But it was unclear whether the outer layer of cells (the RPE) also contributed to this process. Moreno-Marmol et al. were able to investigate this question by genetically modifying zebrafish to make all new RPE cells fluoresce. Following the early development of the zebrafish eye under a microscope revealed that RPE cells flattened themselves into long thin structures that stretched to cover the entire neural retina. This change was made possible by the cell’s internal skeleton reorganizing. In fact, preventing this reorganization stopped the RPE cells from flattening, and precluded the optic cup from acquiring its curved shape. The results thus confirmed a direct role for the RPE in generating curvature. The entire process did not require the RPE to produce new cells, allowing the curved shape to emerge in just a few hours. This is a major advantage for fast-developing species such as zebrafish. In species whose embryos develop more slowly, such as mice and humans, the RPE instead grows by producing additional cells – a process that takes many days. The development of the eye thus shows how various species use different evolutionary approaches to achieve a common goal.
Collapse
Affiliation(s)
- Tania Moreno-Mármol
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Mario Ledesma-Terrón
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain
| | - Noemi Tabanera
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Maria Jesús Martin-Bermejo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Marcos J Cardozo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Florencia Cavodeassi
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| |
Collapse
|
12
|
Casey MA, Lusk S, Kwan KM. Build me up optic cup: Intrinsic and extrinsic mechanisms of vertebrate eye morphogenesis. Dev Biol 2021; 476:128-136. [PMID: 33811855 PMCID: PMC8848517 DOI: 10.1016/j.ydbio.2021.03.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/16/2022]
Abstract
The basic structure of the eye, which is crucial for visual function, is established during the embryonic process of optic cup morphogenesis. Molecular pathways of specification and patterning are integrated with spatially distinct cell and tissue shape changes to generate the eye, with discrete domains and structural features: retina and retinal pigment epithelium enwrap the lens, and the optic fissure occupies the ventral surface of the eye and optic stalk. Interest in the underlying cell biology of eye morphogenesis has led to a growing body of work, combining molecular genetics and imaging to quantify cellular processes such as adhesion and actomyosin activity. These studies reveal that intrinsic machinery and spatiotemporally specific extrinsic inputs collaborate to control dynamics of cell movements and morphologies. Here we consider recent advances in our understanding of eye morphogenesis, with a focus on the mechanics of eye formation throughout vertebrate systems, including insights and potential opportunities using organoids, which may provide a tractable system to test hypotheses from embryonic models.
Collapse
Affiliation(s)
- Macaulie A Casey
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Sarah Lusk
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Kristen M Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
13
|
Lusk S, Casey MA, Kwan KM. 4-Dimensional Imaging of Zebrafish Optic Cup Morphogenesis. J Vis Exp 2021:10.3791/62155. [PMID: 34125104 PMCID: PMC8848516 DOI: 10.3791/62155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Visual system function requires the establishment of precise tissue and organ structures. In the vertebrate eye, structural defects are a common cause of visual impairment, yet mechanisms of eye morphogenesis are still poorly understood. The basic organization of the embryonic eye is conserved throughout vertebrates, thus live imaging of zebrafish embryos has become a powerful approach to directly observe eye development at real time under normal and pathological conditions. Dynamic cell processes including movements, morphologies, interactions, division, and death can be visualized in the embryo. We have developed methods for uniform labeling of subcellular structures and timelapse confocal microscopy of early eye development in zebrafish. This protocol outlines the method of generating capped mRNA for injection into the 1-cell zebrafish embryo, mounting embryos at optic vesicle stage (~12 hours post fertilization, hpf), and performing multi-dimensional timelapse imaging of optic cup morphogenesis on a laser scanning confocal microscope, such that multiple datasets are acquired sequentially in the same imaging session. Such an approach yields data that can be used for a variety of purposes, including cell tracking, volume measurements, three-dimensional (3D) rendering, and visualization. Our approaches allow us to pinpoint the cellular and molecular mechanisms driving optic cup development, in both wild type and genetic mutant conditions. These methods can be employed directly by other groups or adapted to visualize many additional aspects of zebrafish eye development.
Collapse
Affiliation(s)
- Sarah Lusk
- Department of Human Genetics, University of Utah
| | | | | |
Collapse
|
14
|
Soans KG, Norden C. Shining a light on extracellular matrix dynamics in vivo. Semin Cell Dev Biol 2021; 120:85-93. [PMID: 34030949 DOI: 10.1016/j.semcdb.2021.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 11/19/2022]
Abstract
The extracellular matrix is involved in facilitating morphogenesis during development in many contexts. Its role as a stable structure that supports, constrains and acts a substrate for migrating cells in developing tissues is well known and explored. However, recent studies that image fluorescently tagged matrix proteins in developing embryos and tissues, show more dynamic characteristics of matrices in diverse developmental contexts. In this review, we discuss new insights revealed by live-imaging of matrix proteins that help with the understanding of the dynamics of matrix deposition, degradation, turnover and rearrangement. Further, we discuss the mechanisms by which matrix dynamics can influence morphogenesis during development. We present our view on how the field can move in the future and what live-imaging approaches in diverse model organisms can contribute to this exciting area of developmental biology.
Collapse
Affiliation(s)
- Karen G Soans
- Instituto Gulbenkian de Ciênca, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal; Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany; Excellence Physics of Life, TU Dresden, Germany.
| | - Caren Norden
- Instituto Gulbenkian de Ciênca, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal; Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany.
| |
Collapse
|
15
|
Zebrafish Models of Autosomal Recessive Ataxias. Cells 2021; 10:cells10040836. [PMID: 33917666 PMCID: PMC8068028 DOI: 10.3390/cells10040836] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Autosomal recessive ataxias are much less well studied than autosomal dominant ataxias and there are no clearly defined systems to classify them. Autosomal recessive ataxias, which are characterized by neuronal and multisystemic features, have significant overlapping symptoms with other complex multisystemic recessive disorders. The generation of animal models of neurodegenerative disorders increases our knowledge of their cellular and molecular mechanisms and helps in the search for new therapies. Among animal models, the zebrafish, which shares 70% of its genome with humans, offer the advantages of being small in size and demonstrating rapid development, making them optimal for high throughput drug and genetic screening. Furthermore, embryo and larval transparency allows to visualize cellular processes and central nervous system development in vivo. In this review, we discuss the contributions of zebrafish models to the study of autosomal recessive ataxias characteristic phenotypes, behavior, and gene function, in addition to commenting on possible treatments found in these models. Most of the zebrafish models generated to date recapitulate the main features of recessive ataxias.
Collapse
|
16
|
Sun WR, Ramirez S, Spiller KE, Zhao Y, Fuhrmann S. Nf2 fine-tunes proliferation and tissue alignment during closure of the optic fissure in the embryonic mouse eye. Hum Mol Genet 2020; 29:3373-3387. [PMID: 33075808 DOI: 10.1093/hmg/ddaa228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 11/14/2022] Open
Abstract
Uveal coloboma represents one of the most common congenital ocular malformations accounting for up to 10% of childhood blindness (~1 in 5000 live birth). Coloboma originates from defective fusion of the optic fissure (OF), a transient gap that forms during eye morphogenesis by asymmetric, ventral invagination. Genetic heterogeneity combined with the activity of developmentally regulated genes suggests multiple mechanisms regulating OF closure. The tumor suppressor and FERM domain protein Neurofibromin 2 (NF2) controls diverse processes in cancer, development and regeneration, via Hippo pathway and cytoskeleton regulation. In humans, NF2 mutations can cause ocular abnormalities, including coloboma, however, its actual role in OF closure is unknown. Using conditional inactivation in the embryonic mouse eye, our data indicate that loss of Nf2 function results in a novel underlying cause for coloboma. In particular, mutant eyes show substantially increased retinal pigmented epithelium (RPE) proliferation in the fissure region with concomitant acquisition of RPE cell fate. Cells lining the OF margin can maintain RPE fate ectopically and fail to transition from neuroepithelial to cuboidal shape. In the dorsal RPE of the optic cup, Nf2 inactivation leads to a robust increase in cell number, with local disorganization of the cytoskeleton components F-actin and pMLC2. We propose that RPE hyperproliferation is the primary cause for the observed defects causing insufficient alignment of the OF margins in Nf2 mutants and failure to fuse properly, resulting in persistent coloboma. Our findings indicate that limiting proliferation particularly in the RPE layer is a critical mechanism during OF closure.
Collapse
Affiliation(s)
- Wesley R Sun
- Department of Ophthalmology and Visual Sciences, VEI, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sara Ramirez
- Department of Ophthalmology and Visual Sciences, VEI, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Kelly E Spiller
- Department of Ophthalmology and Visual Sciences, VEI, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yan Zhao
- Department of Ophthalmology and Visual Sciences, VEI, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sabine Fuhrmann
- Department of Ophthalmology and Visual Sciences, VEI, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
17
|
Yoon KH, Fox SC, Dicipulo R, Lehmann OJ, Waskiewicz AJ. Ocular coloboma: Genetic variants reveal a dynamic model of eye development. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:590-610. [PMID: 32852110 DOI: 10.1002/ajmg.c.31831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022]
Abstract
Ocular coloboma is a congenital disorder of the eye where a gap exists in the inferior retina, lens, iris, or optic nerve tissue. With a prevalence of 2-19 per 100,000 live births, coloboma, and microphthalmia, an associated ocular disorder, represent up to 10% of childhood blindness. It manifests due to the failure of choroid fissure closure during eye development, and it is a part of a spectrum of ocular disorders that include microphthalmia and anophthalmia. Use of genetic approaches from classical pedigree analyses to next generation sequencing has identified more than 40 loci that are associated with the causality of ocular coloboma. As we have expanded studies to include singleton cases, hereditability has been very challenging to prove. As such, researchers over the past 20 years, have unraveled the complex interrelationship amongst these 40 genes using vertebrate model organisms. Such research has greatly increased our understanding of eye development. These genes function to regulate initial specification of the eye field, migration of retinal precursors, patterning of the retina, neural crest cell biology, and activity of head mesoderm. This review will discuss the discovery of loci using patient data, their investigations in animal models, and the recent advances stemming from animal models that shed new light in patient diagnosis.
Collapse
Affiliation(s)
- Kevin H Yoon
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,Women & Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Sabrina C Fox
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,Women & Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Renée Dicipulo
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,Women & Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Ordan J Lehmann
- Women & Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada.,Department of Ophthalmology, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew J Waskiewicz
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,Women & Children's Health Research Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
18
|
Fabian L, Dowling JJ. Zebrafish Models of LAMA2-Related Congenital Muscular Dystrophy (MDC1A). Front Mol Neurosci 2020; 13:122. [PMID: 32742259 PMCID: PMC7364686 DOI: 10.3389/fnmol.2020.00122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/11/2020] [Indexed: 01/28/2023] Open
Abstract
LAMA2-related congenital muscular dystrophy (CMD; LAMA2-MD), also referred to as merosin deficient CMD (MDC1A), is a severe neonatal onset muscle disease caused by recessive mutations in the LAMA2 gene. LAMA2 encodes laminin α2, a subunit of the extracellular matrix (ECM) oligomer laminin 211. There are currently no treatments for MDC1A, and there is an incomplete understanding of disease pathogenesis. Zebrafish, due to their high degree of genetic conservation with humans, large clutch sizes, rapid development, and optical clarity, have emerged as an excellent model system for studying rare Mendelian diseases. They are particularly suitable as a model for muscular dystrophy because they contain at least one orthologue to all major human MD genes, have muscle that is similar to human muscle in structure and function, and manifest obvious and easily measured MD related phenotypes. In this review article, we present the existing zebrafish models of MDC1A, and discuss their contribution to the understanding of MDC1A pathomechanisms and therapy development.
Collapse
Affiliation(s)
- Lacramioara Fabian
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada.,Division of Neurology, Hospital for Sick Children, Toronto, ON, Canada.,Departments of Pediatrics and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Allan K, DiCicco R, Ramos M, Asosingh K, Yuan A. Preparing a Single Cell Suspension from Zebrafish Retinal Tissue for Flow Cytometric Cell Sorting of Müller Glia. Cytometry A 2020; 97:638-646. [PMID: 31769194 PMCID: PMC7246168 DOI: 10.1002/cyto.a.23936] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/23/2022]
Abstract
Preparation of a single cell suspension from solid tissue is vital for a successful flow cytometry experiment. We report a detailed and reproducible method to produce a quality cell suspension from the zebrafish retina. Zebrafish retinas, especially their Müller glia cells, are of particular interest for their inherent regenerative capacity, making them a useful model for regenerative medicine and cell therapy research. Here, we detail a papain-based dissociation that is gentle enough to keep cells intact, but strong enough to disrupt cell-cell and cell-matrix interactions to yield a cell suspension that produces clean and reliable flow cytometric cell sorting results. This procedure consistently results in over 90% viability and three populations of cells based on GFP expression. The dissociation procedure described herein has been optimized for the collection of Müller glia from Tg(apoe:gfp) zebrafish retinas; however, the overall process may be applicable to other cell types in the fish retina, additional flow cytometric techniques, or preparing cell suspensions from similar tissues. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Kristin Allan
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
- Department of Ophthalmic Research, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
- Cole Eye Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Rose DiCicco
- Department of Ophthalmic Research, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
- Cole Eye Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Michael Ramos
- Department of Ophthalmic Research, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
- Cole Eye Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
- Flow Cytometry Core, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Alex Yuan
- Department of Ophthalmic Research, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
- Cole Eye Institute, The Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
20
|
Bryan CD, Casey MA, Pfeiffer RL, Jones BW, Kwan KM. Optic cup morphogenesis requires neural crest-mediated basement membrane assembly. Development 2020; 147:dev181420. [PMID: 31988185 PMCID: PMC7044464 DOI: 10.1242/dev.181420] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 01/13/2020] [Indexed: 12/21/2022]
Abstract
Organogenesis requires precise interactions between a developing tissue and its environment. In vertebrates, the developing eye is surrounded by a complex extracellular matrix as well as multiple mesenchymal cell populations. Disruptions to either the matrix or periocular mesenchyme can cause defects in early eye development, yet in many cases the underlying mechanism is unknown. Here, using multidimensional imaging and computational analyses in zebrafish, we establish that cell movements in the developing optic cup require neural crest. Ultrastructural analysis reveals that basement membrane formation around the developing eye is also dependent on neural crest, but only specifically around the retinal pigment epithelium. Neural crest cells produce the extracellular matrix protein nidogen: impairing nidogen function disrupts eye development, and, strikingly, expression of nidogen in the absence of neural crest partially restores optic cup morphogenesis. These results demonstrate that eye formation is regulated in part by extrinsic control of extracellular matrix assembly.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Chase D Bryan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Macaulie A Casey
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Rebecca L Pfeiffer
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Bryan W Jones
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Kristen M Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
21
|
George A, Cogliati T, Brooks BP. Genetics of syndromic ocular coloboma: CHARGE and COACH syndromes. Exp Eye Res 2020; 193:107940. [PMID: 32032630 DOI: 10.1016/j.exer.2020.107940] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023]
Abstract
Optic fissure closure defects result in uveal coloboma, a potentially blinding condition affecting between 0.5 and 2.6 per 10,000 births that may cause up to 10% of childhood blindness. Uveal coloboma is on a phenotypic continuum with microphthalmia (small eye) and anophthalmia (primordial/no ocular tissue), the so-called MAC spectrum. This review gives a brief overview of the developmental biology behind coloboma and its clinical presentation/spectrum. Special attention will be given to two prominent, syndromic forms of coloboma, namely, CHARGE (Coloboma, Heart defect, Atresia choanae, Retarded growth and development, Genital hypoplasia, and Ear anomalies/deafness) and COACH (Cerebellar vermis hypoplasia, Oligophrenia, Ataxia, Coloboma, and Hepatic fibrosis) syndromes. Approaches employed to identify genes involved in optic fissure closure in animal models and recent advances in live imaging of zebrafish eye development are also discussed.
Collapse
Affiliation(s)
- Aman George
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health. Bethesda, Maryland, 20892, USA
| | - Tiziana Cogliati
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health. Bethesda, Maryland, 20892, USA
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health. Bethesda, Maryland, 20892, USA.
| |
Collapse
|
22
|
Carney KR, Bryan CD, Gordon HB, Kwan KM. LongAxis: A MATLAB-based program for 3D quantitative analysis of epithelial cell shape and orientation. Dev Biol 2019; 458:1-11. [PMID: 31589834 DOI: 10.1016/j.ydbio.2019.09.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/13/2019] [Accepted: 09/27/2019] [Indexed: 10/25/2022]
Abstract
Epithelial morphogenesis, a fundamental aspect of development, generates 3-dimensional tissue structures crucial for organ function. Underlying morphogenetic mechanisms are, in many cases, poorly understood, but mutations that perturb organ development can affect epithelial cell shape and orientation - difficult features to quantify in three dimensions. The basic structure of the eye is established via epithelial morphogenesis: in the embryonic optic cup, the retinal progenitor epithelium enwraps the lens. We previously found that loss of the extracellular matrix protein laminin-alpha1 (lama1) led to mislocalization of apical polarity markers and apparent misorientation of retinal progenitors. We sought to visualize and quantify this phenotype, and determine whether loss of the apical polarity determinant pard3 might rescue the phenotype. To this end, we developed LongAxis, a MATLAB-based program optimized for the retinal progenitor neuroepithelium. LongAxis facilitates 3-dimensional cell segmentation, visualization, and quantification of cell orientation and morphology. Using LongAxis, we find that retinal progenitors in the lama1-/- optic cup are misoriented and slightly less elongated. In the lama1;MZpard3 double mutant, cells are still misoriented, but larger. Therefore, loss of pard3 does not rescue loss of lama1, and in fact uncovers a novel cell size phenotype. LongAxis enables population-level visualization and quantification of retinal progenitor cell orientation and morphology. These results underscore the importance of visualizing and quantifying cell orientation and shape in three dimensions within the retina.
Collapse
Affiliation(s)
- Keith R Carney
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Chase D Bryan
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Hannah B Gordon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Kristen M Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
23
|
Cavodeassi F, Wilson SW. Looking to the future of zebrafish as a model to understand the genetic basis of eye disease. Hum Genet 2019; 138:993-1000. [PMID: 31422478 PMCID: PMC6710215 DOI: 10.1007/s00439-019-02055-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
Abstract
In this brief commentary, we provide some of our thoughts and opinions on the current and future use of zebrafish to model human eye disease, dissect pathological progression and advance in our understanding of the genetic bases of microphthalmia, andophthalmia and coloboma (MAC) in humans. We provide some background on eye formation in fish and conservation and divergence across vertebrates in this process, discuss different approaches for manipulating gene function and speculate on future research areas where we think research using fish may prove to be particularly effective.
Collapse
Affiliation(s)
- Florencia Cavodeassi
- Institute of Medical and Biomedical Education, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, UK.
| | - Stephen W Wilson
- Department of Cell and Developmental Biology, Biosciences, UCL, Gower St, London, WC1E 6BT, UK
| |
Collapse
|
24
|
Carrara N, Weaver M, Piedade WP, Vöcking O, Famulski JK. Temporal characterization of optic fissure basement membrane composition suggests nidogen may be an initial target of remodeling. Dev Biol 2019; 452:43-54. [PMID: 31034836 DOI: 10.1016/j.ydbio.2019.04.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/08/2019] [Accepted: 04/23/2019] [Indexed: 01/26/2023]
Abstract
Fusion of the optic fissure is necessary to complete retinal morphogenesis and ensure proper function of the optic stalk. Failure of this event leads to congenital coloboma, one of the leading causes of pediatric blindness. Mechanistically it is widely accepted that the basement membrane (BM) surrounding the maturing retina needs to be remodeled within the fissure in order to facilitate subsequent epithelial sheet fusion. However, the mechanism driving BM remodeling has yet to be elucidated. As a first step to understanding this critical molecular event we comprehensively characterized the core composition of optic fissure BMs in the zebrafish embryos. Zebrafish optic fissure BMs were found to express laminin a1, a4, b1a, c1 and c3, nidogen 1a, 1b and 2a, collagen IV a1 and a2 as well as perlecan. Furthermore, we observed that laminin, perlecan and collagen IV expression persists in the fissure during fusion, up to 56 hpf, while nidogen expression is downregulated upon initiation of fusion, at 36 hpf. Using immunohistochemistry we also show that nidogen is removed from the BM prior to that of laminin, indicating that remodeling of the BM is an ordered event. Lastly, we characterized retinal morphogenesis in the absence of nidogen function and documented retinal malformation similar to what is observed in laminin mutants. Taken together, we propose a model of BM remodeling where nidogen acts as a linchpin during initiation of optic fissure fusion.
Collapse
Affiliation(s)
| | - Megan Weaver
- Department of Biology, University of Kentucky, USA
| | | | | | - J K Famulski
- Department of Biology, University of Kentucky, USA.
| |
Collapse
|
25
|
Tang M, Luo Z, Wu Y, Zhuang J, Li K, Hu D, Rong H, Xian B, Ge J. BAM15 attenuates transportation-induced apoptosis in iPS-differentiated retinal tissue. Stem Cell Res Ther 2019; 10:64. [PMID: 30795805 PMCID: PMC6387563 DOI: 10.1186/s13287-019-1151-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 02/08/2023] Open
Abstract
Background BAM15 is a novel mitochondrial protonophore uncoupler capable of protecting mammals from acute renal ischemic-reperfusion injury and cold-induced microtubule damage. The purpose of our study was to investigate the effect of BAM15 on apoptosis during 5-day transportation of human-induced pluripotent stem (hiPS)-differentiated retinal tissue. Methods Retinal tissues of 30 days and 60 days were transported with or without BAM15 for 5 days in the laboratory or by real express. Immunofluorescence staining of apoptosis marker cleaved caspase3, proliferation marker Ki67, and neural axon marker NEFL was performed. And expression of apoptotic-related factors p53, NFkappaB, and TNF-a was detected by real-time PCR. Also, location of ganglion cells, photoreceptor cells, amacrine cells, and precursors of neuronal cell types in retinal tissue was stained by immunofluorescence after transportation. Furthermore, cell viability was assessed by CCK8 assay. Results Results showed transportation remarkably intensified expression of apoptotic factor cleaved caspase3, p53, NFkappaB, and TNF-a, which could be reduced by supplement of BAM15. In addition, neurons were severely injured after transportation, with axons manifesting disrupted and tortuous by staining NEFL. And the addition of BAM15 in transportation was able to protect neuronal structure and increase cell viability without affecting subtypes cells location of retinal tissue. Conclusions BAM15 might be used as a protective reagent on apoptosis during transporting retinal tissues, holding great potential in research and clinical applications. Electronic supplementary material The online version of this article (10.1186/s13287-019-1151-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mingjun Tang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Ziming Luo
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Yihui Wu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Kaijing Li
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Dongpeng Hu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Huifeng Rong
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Bikun Xian
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China.
| |
Collapse
|
26
|
The extracellular matrix-myosin pathway in mechanotransduction: from molecule to tissue. Emerg Top Life Sci 2018; 2:727-737. [PMID: 33530663 PMCID: PMC7289002 DOI: 10.1042/etls20180043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/26/2018] [Accepted: 09/28/2018] [Indexed: 12/23/2022]
Abstract
Mechanotransduction via the extracellular matrix (ECM)–myosin pathway is involved in determining cell morphology during development and in coupling external transient mechanical stimuli to the reorganization of the cytoskeleton. Here, we present a review on the molecular mechanisms involved in this pathway and how they influence cellular development and organization. We investigate key proteins involved in the ECM–myosin pathway and discuss how specific binding events and conformational changes under force are related to mechanical signaling. We connect these molecular mechanisms with observed morphological changes at the cellular and organism level. Finally, we propose a model encompassing the biomechanical signals along the ECM–myosin pathway and how it could be involved in cell adhesion, cell migration, and tissue architecture.
Collapse
|
27
|
Gordon HB, Lusk S, Carney KR, Wirick EO, Murray BF, Kwan KM. Hedgehog signaling regulates cell motility and optic fissure and stalk formation during vertebrate eye morphogenesis. Development 2018; 145:dev.165068. [PMID: 30333214 PMCID: PMC6262791 DOI: 10.1242/dev.165068] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 10/08/2018] [Indexed: 12/18/2022]
Abstract
Establishment of precise three-dimensional tissue structure is vital for organ function. In the visual system, optic fissure and stalk morphogenesis is a crucial yet poorly understood process, disruptions of which can lead to coloboma, a birth defect causing visual impairment. Here, we use four-dimensional imaging, cell tracking, and molecular genetics in zebrafish to define the cell movements underlying normal optic fissure and stalk formation. We determine how these events are disrupted in a coloboma model in which the Hedgehog (Hh) receptor ptch2 is lost, resulting in overactive Hh signaling. In the ptch2 mutant, cells exhibit defective motile behaviors and morphology. Cells that should contribute to the fissure do not arrive at their correct position, and instead contribute to an ectopically large optic stalk. Our results suggest that overactive Hh signaling, through overexpression of downstream transcriptional targets, impairs cell motility underlying optic fissure and stalk formation, via non-cell-autonomous and cell-autonomous mechanisms. More broadly, our cell motility and morphology analyses provide a new framework for studying other coloboma-causing mutations that disrupt optic fissure or stalk formation. Summary: Multidimensional imaging of ptch2 mutant zebrafish uncovers a role for the Hh signaling pathway in regulating the cell and tissue dynamics underlying early eye morphogenesis.
Collapse
Affiliation(s)
- Hannah B Gordon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Sarah Lusk
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Keith R Carney
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Emily O Wirick
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Kristen M Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
28
|
Visetsouk MR, Falat EJ, Garde RJ, Wendlick JL, Gutzman JH. Basal epithelial tissue folding is mediated by differential regulation of microtubules. Development 2018; 145:dev.167031. [PMID: 30333212 PMCID: PMC6262788 DOI: 10.1242/dev.167031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/09/2018] [Indexed: 01/02/2023]
Abstract
The folding of epithelial tissues is crucial for development of three-dimensional structure and function. Understanding this process can assist in determining the etiology of developmental disease and engineering of tissues for the future of regenerative medicine. Folding of epithelial tissues towards the apical surface has long been studied, but the molecular mechanisms that mediate epithelial folding towards the basal surface are just emerging. Here, we utilize zebrafish neuroepithelium to identify mechanisms that mediate basal tissue folding to form the highly conserved embryonic midbrain-hindbrain boundary. Live imaging revealed Wnt5b as a mediator of anisotropic epithelial cell shape, both apically and basally. In addition, we uncovered a Wnt5b-mediated mechanism for specific regulation of basal anisotropic cell shape that is microtubule dependent and likely to involve JNK signaling. We propose a model in which a single morphogen can differentially regulate apical versus basal cell shape during tissue morphogenesis. Summary: Examination of cell shape changes during zebrafish neuroepithelium tissue folding reveals that Wnt5b specifically regulates basal anisotropic cell shape via a microtubule-dependent mechanism, likely involving JNK signaling.
Collapse
Affiliation(s)
- Mike R Visetsouk
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201, USA
| | - Elizabeth J Falat
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201, USA
| | - Ryan J Garde
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201, USA
| | - Jennifer L Wendlick
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201, USA
| | - Jennifer H Gutzman
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201, USA
| |
Collapse
|
29
|
Moreno-Marmol T, Cavodeassi F, Bovolenta P. Setting Eyes on the Retinal Pigment Epithelium. Front Cell Dev Biol 2018; 6:145. [PMID: 30406103 PMCID: PMC6207792 DOI: 10.3389/fcell.2018.00145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/08/2018] [Indexed: 01/08/2023] Open
Abstract
The neural component of the zebrafish eye derives from a small group of cells known as the eye/retinal field. These cells, positioned in the anterior neural plate, rearrange extensively and generate the optic vesicles (OVs). Each vesicle subsequently folds over itself to form the double-layered optic cup, from which the mature eye derives. During this transition, cells of the OV are progressively specified toward three different fates: the retinal pigment epithelium (RPE), the neural retina, and the optic stalk. Recent studies have shown that folding of the zebrafish OV into a cup is in part driven by basal constriction of the cells of the future neural retina. During folding, however, RPE cells undergo an even more dramatic shape conversion that seems to entail the acquisition of unique properties. How these changes occur and whether they contribute to optic cup formation is still poorly understood. Here we will review present knowledge on RPE morphogenesis and discuss potential mechanisms that may explain such transformation using examples taken from embryonic Drosophila tissues that undergo similar shape changes. We will also put forward a hypothesis for optic cup folding that considers an active contribution from the RPE.
Collapse
Affiliation(s)
- Tania Moreno-Marmol
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Florencia Cavodeassi
- Institute of Medical and Biomedical Education, University of London, London, United Kingdom
| | - Paola Bovolenta
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
30
|
Hehr CL, Halabi R, McFarlane S. Polarity and morphogenesis of the eye epithelium requires the adhesion junction associated adaptor protein Traf4. Cell Adh Migr 2018; 12:489-502. [PMID: 29961393 DOI: 10.1080/19336918.2018.1477900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
During development, neuroepithelial progenitors acquire apico-basal polarity and adhere to one another via apically located tight and adherens junction complexes. This polarized neuroepithelium must continue to integrate cells arising through cell divisions and intercalation, and allow for cell movements, at the same time as undergoing morphogenesis. Cell proliferation, migration and intercalation all occur in the morphing embryonic eye. To understand how eye development might depend on dynamic epithelial adhesion, we investigated the function of a known regulator of junctional plasticity, Tumour necrosis factor receptor-associated factor 4 (Traf4). traf4a mRNA is expressed in the developing eye vesicle over the period of optic cup morphogenesis, and Traf4a loss leads to disrupted evagination and elongation of the eye vesicles, and aberrant organization and apico-basal polarity of the eye epithelium. We propose a model whereby Traf4a regulates apical junction plasticity in nascent eye epithelium, allowing for its polarization and morphogenesis. Symbols and Abbreviations: AB: apico-basal; aPKC: atypical protein kinase-C; CRISPR: clustered regularly-interspaced short palindromic repeats; GFP: green fluorescent protein; hpf: hours post-fertilization; MO: antisense morpholino oligonucleotide; pHH3: phospho histone H3; ss: somite stage; Traf4: Tumour necrosis factor receptor-associated factor 4; ZO-1: zona occludens-1.
Collapse
Affiliation(s)
- Carrie Lynn Hehr
- a Department of Cell Biology and Anatomy , University of Calgary, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute , Calgary , AB , Canada
| | - Rami Halabi
- a Department of Cell Biology and Anatomy , University of Calgary, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute , Calgary , AB , Canada
| | - Sarah McFarlane
- a Department of Cell Biology and Anatomy , University of Calgary, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute , Calgary , AB , Canada
| |
Collapse
|
31
|
Dorgau B, Felemban M, Sharpe A, Bauer R, Hallam D, Steel DH, Lindsay S, Mellough C, Lako M. Laminin γ3 plays an important role in retinal lamination, photoreceptor organisation and ganglion cell differentiation. Cell Death Dis 2018; 9:615. [PMID: 29795281 PMCID: PMC5966411 DOI: 10.1038/s41419-018-0648-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/12/2018] [Accepted: 04/26/2018] [Indexed: 01/02/2023]
Abstract
Laminins are heterotrimeric glycoproteins of the extracellular matrix. Eleven different laminin chains have been identified in vertebrates. They are ubiquitously expressed in the human body, with a distinct tissue distribution. Laminin expression in neural retina and their functional role during human retinogenesis is still unknown. This study investigated the laminin expression in human developing and adult retina, showing laminin α1, α5, β1, β2 and γ1 to be predominantly expressed in Bruch's membrane and the inner limiting membrane. Laminin-332 and laminin γ3 expression were mainly observed in the neural retina during retinal histogenesis. These expression patterns were largely conserved in pluripotent stem cell-derived retinal organoids. Blocking of laminin γ3 function in retinal organoids resulted in the disruption of laminar organisation and synapse formation, the loss of photoreceptor organisation and retinal ganglion cells. Our data demonstrate a unique temporal and spatial expression for laminins and reveal a novel role for laminin γ3 during human retinogenesis.
Collapse
Affiliation(s)
- Birthe Dorgau
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Majed Felemban
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alexander Sharpe
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Roman Bauer
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dean Hallam
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David H Steel
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Susan Lindsay
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Carla Mellough
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, University of Western Australia, Nedlands WA, Australia
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
32
|
Sun H, Wan N, Wang X, Chang L, Cheng D. Genotype-Phenotype Analysis, Neuropsychological Assessment, and Growth Hormone Response in a Patient with 18p Deletion Syndrome. Cytogenet Genome Res 2018; 154:71-78. [PMID: 29544220 DOI: 10.1159/000487371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2017] [Indexed: 02/01/2023] Open
Abstract
18p deletion syndrome is a rare chromosomal disease caused by deletion of the short arm of chromosome 18. By using cytogenetic and SNP array analysis, we identified a girl with 18p deletion syndrome exhibiting craniofacial anomalies, intellectual disability, and short stature. G-banding analysis of metaphase cells revealed an abnormal karyotype 46,XX,del(18)(p10). Further, SNP array detected a 15.3-Mb deletion at 18p11.21p11.32 (chr18:12842-15375878) including 61 OMIM genes. Genotype-phenotype correlation analysis showed that clinical manifestations of the patient were correlated with LAMA1, TWSG1, and GNAL deletions. Her neuropsychological assessment test demonstrated delay in most cognitive functions including impaired mathematics, linguistic skills, visual motor perception, respond speed, and executive function. Meanwhile, her integrated visual and auditory continuous performance test (IVA-CPT) indicated a severe comprehensive attention deficit. At age 7 and 1/12 years, her height was 110.8 cm (-2.5 SD height for age). Growth hormone (GH) treatment was initiated. After 27 months treatment, her height was increased to 129.6 cm (-1.0 SD height for age) at 9 and 4/12 years, indicating an effective response to GH treatment.
Collapse
Affiliation(s)
- Huihui Sun
- Department of Pediatrics, Beijing Jishuitan Hospital, Beijing, PR China
| | | | | | | | | |
Collapse
|
33
|
Dynamic Tissue Rearrangements during Vertebrate Eye Morphogenesis: Insights from Fish Models. J Dev Biol 2018; 6:jdb6010004. [PMID: 29615553 PMCID: PMC5875564 DOI: 10.3390/jdb6010004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/22/2018] [Accepted: 02/27/2018] [Indexed: 12/16/2022] Open
Abstract
Over the last thirty years, fish models, such as the zebrafish and medaka, have become essential to pursue developmental studies and model human disease. Community efforts have led to the generation of wide collections of mutants, a complete sequence of their genomes, and the development of sophisticated genetic tools, enabling the manipulation of gene activity and labelling and tracking of specific groups of cells during embryonic development. When combined with the accessibility and optical clarity of fish embryos, these approaches have made of them an unbeatable model to monitor developmental processes in vivo and in real time. Over the last few years, live-imaging studies in fish have provided fascinating insights into tissue morphogenesis and organogenesis. This review will illustrate the advantages of fish models to pursue morphogenetic studies by highlighting the findings that, in the last decade, have transformed our understanding of eye morphogenesis.
Collapse
|
34
|
Martinez-Morales JR, Cavodeassi F, Bovolenta P. Coordinated Morphogenetic Mechanisms Shape the Vertebrate Eye. Front Neurosci 2017; 11:721. [PMID: 29326547 PMCID: PMC5742352 DOI: 10.3389/fnins.2017.00721] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/11/2017] [Indexed: 11/22/2022] Open
Abstract
The molecular bases of vertebrate eye formation have been extensively investigated during the past 20 years. This has resulted in the definition of the backbone of the gene regulatory networks controlling the different steps of eye development and has further highlighted a substantial conservation of these networks among vertebrates. Yet, the precise morphogenetic events allowing the formation of the optic cup from a small group of cells within the anterior neural plate are still poorly understood. It is also unclear if the morphogenetic events leading to eyes of very similar shape are indeed comparable among all vertebrates or if there are any species-specific peculiarities. Improved imaging techniques have enabled to follow how the eye forms in living embryos of a few vertebrate models, whereas the development of organoid cultures has provided fascinating tools to recapitulate tissue morphogenesis of other less accessible species. Here, we will discuss what these advances have taught us about eye morphogenesis, underscoring possible similarities and differences among vertebrates. We will also discuss the contribution of cell shape changes to this process and how morphogenetic and patterning mechanisms integrate to assemble the final architecture of the eye.
Collapse
Affiliation(s)
| | - Florencia Cavodeassi
- Centro de Biología Molecular Severo Ochoa, (CSIC/UAM), Madrid, Spain.,CIBERER, ISCIII, Madrid, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, (CSIC/UAM), Madrid, Spain.,CIBERER, ISCIII, Madrid, Spain
| |
Collapse
|
35
|
Cvekl A, Zhang X. Signaling and Gene Regulatory Networks in Mammalian Lens Development. Trends Genet 2017; 33:677-702. [PMID: 28867048 DOI: 10.1016/j.tig.2017.08.001] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 11/16/2022]
Abstract
Ocular lens development represents an advantageous system in which to study regulatory mechanisms governing cell fate decisions, extracellular signaling, cell and tissue organization, and the underlying gene regulatory networks. Spatiotemporally regulated domains of BMP, FGF, and other signaling molecules in late gastrula-early neurula stage embryos generate the border region between the neural plate and non-neural ectoderm from which multiple cell types, including lens progenitor cells, emerge and undergo initial tissue formation. Extracellular signaling and DNA-binding transcription factors govern lens and optic cup morphogenesis. Pax6, c-Maf, Hsf4, Prox1, Sox1, and a few additional factors regulate the expression of the lens structural proteins, the crystallins. Extensive crosstalk between a diverse array of signaling pathways controls the complexity and order of lens morphogenetic processes and lens transparency.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
36
|
Sidhaye J, Norden C. Concerted action of neuroepithelial basal shrinkage and active epithelial migration ensures efficient optic cup morphogenesis. eLife 2017; 6:22689. [PMID: 28372636 PMCID: PMC5380436 DOI: 10.7554/elife.22689] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 03/14/2017] [Indexed: 12/27/2022] Open
Abstract
Organ formation is a multi-scale event that involves changes at the intracellular, cellular and tissue level. Organogenesis often starts with the formation of characteristically shaped organ precursors. However, the cellular mechanisms driving organ precursor formation are often not clear. Here, using zebrafish, we investigate the epithelial rearrangements responsible for the development of the hemispherical retinal neuroepithelium (RNE), a part of the optic cup. We show that in addition to basal shrinkage of RNE cells, active migration of connected epithelial cells into the RNE is a crucial player in its formation. This cellular movement is driven by progressive cell-matrix contacts and actively translocates prospective RNE cells to their correct location before they adopt neuroepithelial fate. Failure of this migration during neuroepithelium formation leads to ectopic determination of RNE cells and consequently impairs optic cup formation. Overall, this study illustrates how spatiotemporal coordination between morphogenic movements and fate determination critically influences organogenesis.
Collapse
Affiliation(s)
- Jaydeep Sidhaye
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Dresden International Graduate School for Biomedicine and Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
37
|
Blanco-Sánchez B, Clément A, Phillips JB, Westerfield M. Zebrafish models of human eye and inner ear diseases. Methods Cell Biol 2016; 138:415-467. [PMID: 28129854 DOI: 10.1016/bs.mcb.2016.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Eye and inner ear diseases are the most common sensory impairments that greatly impact quality of life. Zebrafish have been intensively employed to understand the fundamental mechanisms underlying eye and inner ear development. The zebrafish visual and vestibulo-acoustic systems are very similar to these in humans, and although not yet mature, they are functional by 5days post-fertilization (dpf). In this chapter, we show how the zebrafish has significantly contributed to the field of biomedical research and how researchers, by establishing disease models and meticulously characterizing their phenotypes, have taken the first steps toward therapies. We review here models for (1) eye diseases, (2) ear diseases, and (3) syndromes affecting eye and/or ear. The use of new genome editing technologies and high-throughput screening systems should increase considerably the speed at which knowledge from zebrafish disease models is acquired, opening avenues for better diagnostics, treatments, and therapies.
Collapse
Affiliation(s)
| | - A Clément
- University of Oregon, Eugene, OR, United States
| | | | | |
Collapse
|