Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Nat Commun ; 14(1): 8477, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38123561

ABSTRACT

Following acute retinal damage, zebrafish possess the ability to regenerate all neuronal subtypes through Müller glia (MG) reprogramming and asymmetric cell division that produces a multipotent Müller glia-derived neuronal progenitor cell (MGPC). This raises three key questions. First, do MG reprogram to a developmental retinal progenitor cell (RPC) state? Second, to what extent does regeneration recapitulate retinal development? And finally, does loss of different retinal cell subtypes induce unique MG regeneration responses? We examined these questions by performing single-nuclear and single-cell RNA-Seq and ATAC-Seq in both developing and regenerating retinas. Here we show that injury induces MG to reprogram to a state similar to late-stage RPCs. However, there are major transcriptional differences between MGPCs and RPCs, as well as major transcriptional differences between activated MG and MGPCs when different retinal cell subtypes are damaged. Validation of candidate genes confirmed that loss of different subtypes induces differences in transcription factor gene expression and regeneration outcomes.


Subject(s)
Gene Regulatory Networks , Zebrafish , Animals , Zebrafish/genetics , Retina/metabolism , Neurogenesis/genetics , Neuroglia/metabolism , Cell Proliferation/physiology , Ependymoglial Cells/metabolism
2.
Res Sq ; 2023 Sep 18.
Article in English | MEDLINE | ID: mdl-37790324

ABSTRACT

Following acute retinal damage, zebrafish possess the ability to regenerate all neuronal subtypes. This regeneration requires Müller glia (MG) to reprogram and divide asymmetrically to produce a multipotent Müller glia-derived neuronal progenitor cell (MGPC). This raises three key questions. First, does loss of different retinal cell subtypes induce unique MG regeneration responses? Second, do MG reprogram to a developmental retinal progenitor cell state? And finally, to what extent does regeneration recapitulate retinal development? We examined these questions by performing single-nuclear and single-cell RNA-Seq and ATAC-Seq in both developing and regenerating retinas. While MG reprogram to a state similar to late-stage retinal progenitors in developing retinas, there are transcriptional differences between reprogrammed MG/MGPCs and late progenitors, as well as reprogrammed MG in outer and inner retinal damage models. Validation of candidate genes confirmed that loss of different subtypes induces differences in transcription factor gene expression and regeneration outcomes. This work identifies major differences between gene regulatory networks activated following the selective loss of different subtypes of retina neurons, as well as between retinal regeneration and development.

3.
bioRxiv ; 2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37609307

ABSTRACT

Following acute retinal damage, zebrafish possess the ability to regenerate all neuronal subtypes. This regeneration requires Müller glia (MG) to reprogram and divide asymmetrically to produce a multipotent Müller glia-derived neuronal progenitor cell (MGPC). This raises three key questions. First, does loss of different retinal cell subtypes induce unique MG regeneration responses? Second, do MG reprogram to a developmental retinal progenitor cell state? And finally, to what extent does regeneration recapitulate retinal development? We examined these questions by performing single-nuclear and single-cell RNA-Seq and ATAC-Seq in both developing and regenerating retinas. While MG reprogram to a state similar to late-stage retinal progenitors in developing retinas, there are transcriptional differences between reprogrammed MG/MGPCs and late progenitors, as well as reprogrammed MG in outer and inner retinal damage models. Validation of candidate genes confirmed that loss of different subtypes induces differences in transcription factor gene expression and regeneration outcomes. This work identifies major differences between gene regulatory networks activated following the selective loss of different subtypes of retina neurons, as well as between retinal regeneration and development.

4.
Development ; 149(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35088848

ABSTRACT

Endothelial cells emerge from the atrioventricular canal to form coronary blood vessels in juvenile zebrafish hearts. We find that pdgfrb is first expressed in the epicardium around the atrioventricular canal and later becomes localized mainly in the mural cells. pdgfrb mutant fish show severe defects in mural cell recruitment and coronary vessel development. Single-cell RNA sequencing analyses identified pdgfrb+ cells as epicardium-derived cells (EPDCs) and mural cells. Mural cells associated with coronary arteries also express cxcl12b and smooth muscle cell markers. Interestingly, these mural cells remain associated with coronary arteries even in the absence of Pdgfrß, although smooth muscle gene expression is downregulated. We find that pdgfrb expression dynamically changes in EPDCs of regenerating hearts. Differential gene expression analyses of pdgfrb+ EPDCs and mural cells suggest that they express genes that are important for regeneration after heart injuries. mdka was identified as a highly upregulated gene in pdgfrb+ cells during heart regeneration. However, pdgfrb but not mdka mutants show defects in heart regeneration after amputation. Our results demonstrate that heterogeneous pdgfrb+ cells are essential for coronary development and heart regeneration.


Subject(s)
Coronary Vessels/growth & development , Coronary Vessels/metabolism , Heart/physiology , Organogenesis/physiology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Regeneration/physiology , Animals , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Myocytes, Smooth Muscle/metabolism , Pericardium/metabolism , Zebrafish/metabolism , Zebrafish/physiology
5.
Cells ; 10(4)2021 04 01.
Article in English | MEDLINE | ID: mdl-33916186

ABSTRACT

The ability to regenerate tissues varies between species and between tissues within a species. Mammals have a limited ability to regenerate tissues, whereas zebrafish possess the ability to regenerate almost all tissues and organs, including fin, heart, kidney, brain, and retina. In the zebrafish brain, injury and cell death activate complex signaling networks that stimulate radial glia to reprogram into neural stem-like cells that repair the injury. In the retina, a popular model for investigating neuronal regeneration, Müller glia, radial glia unique to the retina, reprogram into stem-like cells and undergo a single asymmetric division to generate multi-potent retinal progenitors. Müller glia-derived progenitors then divide rapidly, numerically matching the magnitude of the cell death, and differentiate into the ablated neurons. Emerging evidence reveals that inflammation plays an essential role in this multi-step process of retinal regeneration. This review summarizes the current knowledge of the inflammatory events during retinal regeneration and highlights the mechanisms whereby inflammatory molecules regulate the quiescence and division of Müller glia, the proliferation of Müller glia-derived progenitors and the survival of regenerated neurons.


Subject(s)
Inflammation/pathology , Regeneration/physiology , Retina/physiopathology , Zebrafish/physiology , Animals , Cellular Reprogramming , Ependymoglial Cells/pathology , Neurogenesis
6.
PLoS Comput Biol ; 16(12): e1008437, 2020 12.
Article in English | MEDLINE | ID: mdl-33320887

ABSTRACT

The outer epithelial layer of zebrafish retinae contains a crystalline array of cone photoreceptors, called the cone mosaic. As this mosaic grows by mitotic addition of new photoreceptors at the rim of the hemispheric retina, topological defects, called "Y-Junctions", form to maintain approximately constant cell spacing. The generation of topological defects due to growth on a curved surface is a distinct feature of the cone mosaic not seen in other well-studied biological patterns like the R8 photoreceptor array in the Drosophila compound eye. Since defects can provide insight into cell-cell interactions responsible for pattern formation, here we characterize the arrangement of cones in individual Y-Junction cores as well as the spatial distribution of Y-junctions across entire retinae. We find that for individual Y-junctions, the distribution of cones near the core corresponds closely to structures observed in physical crystals. In addition, Y-Junctions are organized into lines, called grain boundaries, from the retinal center to the periphery. In physical crystals, regardless of the initial distribution of defects, defects can coalesce into grain boundaries via the mobility of individual particles. By imaging in live fish, we demonstrate that grain boundaries in the cone mosaic instead appear during initial mosaic formation, without requiring defect motion. Motivated by this observation, we show that a computational model of repulsive cell-cell interactions generates a mosaic with grain boundaries. In contrast to paradigmatic models of fate specification in mostly motionless cell packings, this finding emphasizes the role of cell motion, guided by cell-cell interactions during differentiation, in forming biological crystals. Such a route to the formation of regular patterns may be especially valuable in situations, like growth on a curved surface, where the resulting long-ranged, elastic, effective interactions between defects can help to group them into grain boundaries.


Subject(s)
Retinal Cone Photoreceptor Cells/metabolism , Zebrafish/anatomy & histology , Animals , Cell Communication , Cell Differentiation , Computer Simulation , Zebrafish/growth & development
7.
PLoS One ; 15(6): e0232308, 2020.
Article in English | MEDLINE | ID: mdl-32530962

ABSTRACT

Zebrafish have the ability to regenerate damaged cells and tissues by activating quiescent stem and progenitor cells or reprogramming differentiated cells into regeneration-competent precursors. Proliferation among the cells that will functionally restore injured tissues is a fundamental biological process underlying regeneration. Midkine-a is a cytokine growth factor, whose expression is strongly induced by injury in a variety of tissues across a range of vertebrate classes. Using a zebrafish Midkine-a loss of function mutant, we evaluated regeneration of caudal fin, extraocular muscle and retinal neurons to investigate the function of Midkine-a during epimorphic regeneration. In wildtype zebrafish, injury among these tissues induces robust proliferation and rapid regeneration. In Midkine-a mutants, the initial proliferation in each of these tissues is significantly diminished or absent. Regeneration of the caudal fin and extraocular muscle is delayed; regeneration of the retina is nearly completely absent. These data demonstrate that Midkine-a is universally required in the signaling pathways that convert tissue injury into the initial burst of cell proliferation. Further, these data highlight differences in the molecular mechanisms that regulate epimorphic regeneration in zebrafish.


Subject(s)
Midkine/metabolism , Regeneration/physiology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animal Fins/physiology , Animals , Animals, Genetically Modified/metabolism , Cell Differentiation , Cell Proliferation , Midkine/genetics , Mutagenesis , Neuroglia/cytology , Neuroglia/metabolism , Oculomotor Muscles/physiology , Retinal Neurons/physiology , Zebrafish Proteins/genetics
8.
Annu Rev Vis Sci ; 6: 171-193, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32343929

ABSTRACT

In humans, various genetic defects or age-related diseases, such as diabetic retinopathies, glaucoma, and macular degeneration, cause the death of retinal neurons and profound vision loss. One approach to treating these diseases is to utilize stem and progenitor cells to replace neurons in situ, with the expectation that new neurons will create new synaptic circuits or integrate into existing ones. Reprogramming non-neuronal cells in vivo into stem or progenitor cells is one strategy for replacing lost neurons. Zebrafish have become a valuable model for investigating cellular reprogramming and retinal regeneration. This review summarizes our current knowledge regarding spontaneous reprogramming of Müller glia in zebrafish and compares this knowledge to research efforts directed toward reprogramming Müller glia in mammals. Intensive research using these animal models has revealed shared molecular mechanisms that make Müller glia attractive targets for cellular reprogramming and highlighted the potential for curing degenerative retinal diseases from intrinsic cellular sources.


Subject(s)
Ependymoglial Cells/physiology , Nerve Regeneration/physiology , Retinal Neurons/physiology , Animals , Animals, Genetically Modified , Cell Differentiation , DNA Methylation , Epigenomics , Humans , Receptors, Notch/metabolism , Retinal Neurons/cytology , Signal Transduction , Stem Cells , Zebrafish
9.
Glia ; 68(7): 1445-1465, 2020 07.
Article in English | MEDLINE | ID: mdl-32034934

ABSTRACT

Brain injury activates complex inflammatory signals in dying neurons, surviving neurons, and glia. Here, we establish that inflammation regulates the regeneration of photoreceptors in the zebrafish retina and determine the cellular expression and function of the inflammatory protease, matrix metalloproteinase 9 (Mmp-9), during this regenerative neurogenesis. Following photoreceptor ablation, anti-inflammatory treatment suppresses the number of injury-induced progenitors and regenerated photoreceptors. Upon photoreceptor injury, mmp-9 is induced in Müller glia and Müller glia-derived photoreceptor progenitors. Deleting mmp-9 results in over production of injury-induced progenitors and regenerated photoreceptors, but over time the absence of Mmp-9 compromises the survival of the regenerated cones. At all time-points studied, the levels of tnf-α are significantly elevated in mutant retinas. Anti-inflammatory treatment in mutants rescues the defects in cone survival. These data provide a link between injury-induced inflammation in the vertebrate CNS, Mmp-9 function during neuronal regeneration and the requirement of Mmp-9 for the survival of regenerated cones.


Subject(s)
Inflammation/metabolism , Matrix Metalloproteinase 9/metabolism , Nerve Regeneration/physiology , Regeneration/physiology , Animals , Animals, Genetically Modified , Cell Differentiation/physiology , Cell Proliferation/physiology , Neuroglia/metabolism , Retina/metabolism , Retinal Rod Photoreceptor Cells/physiology , Stem Cells/physiology , Zebrafish
10.
J Neurosci ; 40(6): 1232-1247, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31882403

ABSTRACT

In the retina of zebrafish, Müller glia have the ability to reprogram into stem cells capable of regenerating all classes of retinal neurons and restoring visual function. Understanding the cellular and molecular mechanisms controlling the stem cell properties of Müller glia in zebrafish may provide cues to unlock the regenerative potential in the mammalian nervous system. Midkine is a cytokine/growth factor with multiple roles in neural development, tissue repair, and disease. In midkine-a loss-of-function mutants of both sexes, Müller glia initiate the appropriate reprogramming response to photoreceptor death by increasing expression of stem cell-associated genes, and entering the G1 phase of the cell cycle. However, transition from G1 to S phase is blocked in the absence of Midkine-a, resulting in significantly reduced proliferation and selective failure to regenerate cone photoreceptors. Failing to progress through the cell cycle, Müller glia undergo reactive gliosis, a pathological hallmark in the injured CNS of mammals. Finally, we determined that the Midkine-a receptor, anaplastic lymphoma kinase, is upstream of the HLH regulatory protein, Id2a, and of the retinoblastoma gene, p130, which regulates progression through the cell cycle. These results demonstrate that Midkine-a functions as a core component of the mechanisms that regulate proliferation of stem cells in the injured CNS.SIGNIFICANCE STATEMENT The death of retinal neurons and photoreceptors is a leading cause of vision loss. Regenerating retinal neurons is a therapeutic goal. Zebrafish can regenerate retinal neurons from intrinsic stem cells, Müller glia, and are a powerful model to understand how stem cells might be used therapeutically. Midkine-a, an injury-induced growth factor/cytokine that is expressed by Müller glia following neuronal death, is required for Müller glia to progress through the cell cycle. The absence of Midkine-a suspends proliferation and neuronal regeneration. With cell cycle progression stalled, Müller glia undergo reactive gliosis, a pathological hallmark of the mammalian retina. This work provides a unique insight into mechanisms that control the cell cycle during neuronal regeneration.


Subject(s)
Cell Dedifferentiation/physiology , Cellular Reprogramming/physiology , Midkine/metabolism , Nerve Regeneration/physiology , Neuroglia , Retina , Animals , Animals, Genetically Modified , Cell Cycle/physiology , Cell Proliferation/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Retina/cytology , Retina/metabolism , Zebrafish , Zebrafish Proteins/metabolism
11.
Bio Protoc ; 10(24): e3848, 2020 Dec 20.
Article in English | MEDLINE | ID: mdl-33659497

ABSTRACT

Immunohistochemistry is a widely used technique to examine the expression and subcellular localization of proteins. This technique relies on the specificity of antibodies and requires adequate penetration of antibodies into tissues. The latter is especially challenging for thick specimens, such as embryos and other whole-mount preparations. Here we describe an improved method of immunohistochemistry for retinal whole-mount preparations. We report that a cocktail of three reagents, Triton X-100, Tween-20, and DMSO, in blocking and antibody dilution buffers strongly enhances immunolabeling in whole-mount retinas from adult zebrafish. In addition, we establish that in whole retinal tissues, a classic epitope retrieval method, based on citrate buffer, is effective for immunolabeling membrane-associated proteins. Overall, this simple modification allows precise and reproducible immunolabeling of proteins in retinal whole-mounts.

12.
Invest Ophthalmol Vis Sci ; 59(1): 505-518, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29368007

ABSTRACT

Purpose: Human Crb1 is implicated in some forms of retinal degeneration, suggesting a role in photoreceptor maintenance. Multiple Crumbs (Crb) polarity genes are expressed in vertebrate retina, although their functional roles are not well understood. To gain further insight into Crb and photoreceptor maintenance, we compared retinal cell densities between wild-type and Tg(RH2-2:Crb2b-sfEX/RH2-2:GFP)pt108b transgenic zebrafish, in which the extracellular domain of Crb2b-short form (Crb2b-sfEX) is expressed in the retina as a secreted protein, which disrupts the planar organization of RGB cones (red, green, and blue) by interfering with Crb2a/2b-based cone-cone adhesion. Methods: We used standard morphometric techniques to assess age-related changes in retinal cell densities in adult zebrafish (3 to 27 months old), and to assess effects of the Crb2b-sfEX transgene on retinal structure and photoreceptor densities. Linear cell densities were measured in all retinal layers in radial sections with JB4-Feulgen histology. Planar (surface) densities of cones were determined in retinal flat-mounts. Cell counts from wild-type and pt108b transgenic fish were compared with both a "photoreceptor maintenance index" and statistical analysis of cell counts. Results: Age-related changes in retinal cell linear densities and cone photoreceptor planar densities in wild-type adult zebrafish provided a baseline for analysis. Expression of Crb2b-sfEX caused progressive and selective degeneration of RGB cones, but had no effect on ultraviolet-sensitive (UV) cones, and increased numbers of rod photoreceptors. Conclusions: These differential responses of RGB cones, UV cones, and rods to sustained exposure to Crb2b-sfEX suggest that Crb-based photoreceptor maintenance mechanisms are highly selective.


Subject(s)
Disease Models, Animal , Gene Expression Regulation/physiology , Membrane Proteins/genetics , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/physiopathology , Zebrafish Proteins/genetics , Aging/physiology , Animals , Animals, Genetically Modified , Cell Count , Immunohistochemistry , Zebrafish
13.
Neural Dev ; 12(1): 20, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29141686

ABSTRACT

BACKGROUND: The multiplex, lattice mosaic of cone photoreceptors in the adult fish retina is a compelling example of a highly ordered epithelial cell pattern, with single cell width rows and columns of cones and precisely defined neighbor relationships among different cone types. Cellular mechanisms patterning this multiplex mosaic are not understood. Physical models can provide new insights into fundamental mechanisms of biological patterning. In earlier work, we developed a mathematical model of photoreceptor cell packing in the zebrafish retina, which predicted that anisotropic mechanical tension in the retinal epithelium orients planar polarized adhesive interfaces to align the columns as cone photoreceptors are generated at the retinal margin during post-embryonic growth. METHODS: With cell-specific fluorescent reporters and in vivo imaging of the growing retinal margin in transparent juvenile zebrafish we provide the first view of how cell packing, spatial arrangement, and cell identity are coordinated to build the lattice mosaic. With targeted laser ablation we probed the tissue mechanics of the retinal epithelium. RESULTS: Within the lattice mosaic, planar polarized Crumbs adhesion proteins pack cones into a single cell width column; between columns, N-cadherin-mediated adherens junctions stabilize Müller glial apical processes. The concentration of activated pMyosin II at these punctate adherens junctions suggests that these glial bands are under tension, forming a physical barrier between cone columns and contributing to mechanical stress anisotropies in the epithelial sheet. Unexpectedly, we discovered that the appearance of such parallel bands of Müller glial apical processes precedes the packing of cones into single cell width columns, hinting at a possible role for glia in the initial organization of the lattice mosaic. Targeted laser ablation of Müller glia directly demonstrates that these glial processes support anisotropic mechanical tension in the planar dimension of the retinal epithelium. CONCLUSIONS: These findings uncovered a novel structural feature of Müller glia associated with alignment of photoreceptors into a lattice mosaic in the zebrafish retina. This is the first demonstration, to our knowledge, of planar, anisotropic mechanical forces mediated by glial cells.


Subject(s)
Ependymoglial Cells/cytology , Neurogenesis/physiology , Retina/growth & development , Retinal Cone Photoreceptor Cells/cytology , Animals , Zebrafish
14.
PLoS One ; 9(8): e106010, 2014.
Article in English | MEDLINE | ID: mdl-25162623

ABSTRACT

Fish retinal ganglion cells (RGCs) can regenerate their axons after optic nerve injury, whereas mammalian RGCs normally fail to do so. Interleukin 6 (IL-6)-type cytokines are involved in cell differentiation, proliferation, survival, and axon regrowth; thus, they may play a role in the regeneration of zebrafish RGCs after injury. In this study, we assessed the expression of IL-6-type cytokines and found that one of them, leukemia inhibitory factor (LIF), is upregulated in zebrafish RGCs at 3 days post-injury (dpi). We then demonstrated the activation of signal transducer and activator of transcription 3 (STAT3), a downstream target of LIF, at 3-5 dpi. To determine the function of LIF, we performed a LIF knockdown experiment using LIF-specific antisense morpholino oligonucleotides (LIF MOs). LIF MOs, which were introduced into zebrafish RGCs via a severed optic nerve, reduced the expression of LIF and abrogated the activation of STAT3 in RGCs after injury. These results suggest that upregulated LIF drives Janus kinase (Jak)/STAT3 signaling in zebrafish RGCs after nerve injury. In addition, the LIF knockdown impaired axon sprouting in retinal explant culture in vitro; reduced the expression of a regeneration-associated molecule, growth-associated protein 43 (GAP-43); and delayed functional recovery after optic nerve injury in vivo. In this study, we comprehensively demonstrate the beneficial role of LIF in optic nerve regeneration and functional recovery in adult zebrafish.


Subject(s)
Leukemia Inhibitory Factor/genetics , Nerve Regeneration/genetics , Optic Nerve Injuries/genetics , Retinal Ganglion Cells/metabolism , STAT3 Transcription Factor/genetics , Zebrafish Proteins/genetics , Animals , Diffusion , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , Gene Expression Regulation , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Leukemia Inhibitory Factor/antagonists & inhibitors , Leukemia Inhibitory Factor/metabolism , Morpholinos/genetics , Morpholinos/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Optic Nerve Injuries/rehabilitation , Recovery of Function/physiology , Retinal Ganglion Cells/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction , Time Factors , Tissue Culture Techniques , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
15.
PLoS One ; 9(1): e85325, 2014.
Article in English | MEDLINE | ID: mdl-24465536

ABSTRACT

Cone photoreceptors in teleost fish are organized in precise, crystalline arrays in the epithelial plane of the retina. In zebrafish, four distinct morphological/spectral cone types occupy specific, invariant positions within a regular lattice. The cone lattice is aligned orthogonal and parallel to circumference of the retinal hemisphere: it emerges as cones generated in a germinal zone at the retinal periphery are incorporated as single-cell columns into the cone lattice. Genetic disruption of the transcription factor Tbx2b eliminates most of the cone subtype maximally sensitive to ultraviolet (UV) wavelengths and also perturbs the long-range organization of the cone lattice. In the tbx2b mutant, the other three cone types (red, green, and blue cones) are specified in the correct proportion, differentiate normally, and acquire normal, planar polarized adhesive interactions mediated by Crumbs 2a and Crumbs 2b. Quantitative image analysis of cell adjacency revealed that the cones in the tbx2b mutant primarily have two nearest neighbors and align in single-cell-wide column fragments that are separated by rod photoreceptors. Some UV cones differentiate at the dorsal retinal margin in the tbx2b mutant, although they are severely dysmorphic and are eventually eliminated. Incorporating loss of UV cones during formation of cone columns at the margin into our previously published mathematical model of zebrafish cone mosaic formation (which uses bidirectional interactions between planar cell polarity proteins and anisotropic mechanical stresses in the plane of the retinal epithelium to generate regular columns of cones parallel to the margin) reproduces many features of the pattern disruptions seen in the tbx2b mutant.


Subject(s)
Morphogenesis/genetics , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Rod Photoreceptor Cells/ultrastructure , T-Box Domain Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Cell Adhesion , Cell Communication/radiation effects , Cell Differentiation , Cell Polarity/radiation effects , Embryo, Nonmammalian , Gene Deletion , Gene Expression Regulation, Developmental , In Situ Hybridization , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/radiation effects , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/radiation effects , Signal Transduction , T-Box Domain Proteins/deficiency , Ultraviolet Rays , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/deficiency , Zebrafish Proteins/metabolism
16.
Development ; 140(22): 4510-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24154521

ABSTRACT

Müller glia function as retinal stem cells in adult zebrafish. In response to loss of retinal neurons, Müller glia partially dedifferentiate, re-express neuroepithelial markers and re-enter the cell cycle. We show that the immunoglobulin superfamily adhesion molecule Alcama is a novel marker of multipotent retinal stem cells, including injury-induced Müller glia, and that each Müller glial cell divides asymmetrically only once to produce an Alcama-negative, proliferating retinal progenitor. The initial mitotic division of Müller glia involves interkinetic nuclear migration, but mitosis of retinal progenitors occurs in situ. Rapidly dividing retinal progenitors form neurogenic clusters tightly associated with Alcama/N-cadherin-labeled Müller glial radial processes. Genetic suppression of N-cadherin function interferes with basal migration of retinal progenitors and subsequent regeneration of HuC/D(+) inner retinal neurons.


Subject(s)
Asymmetric Cell Division , Cadherins/metabolism , Ependymoglial Cells/cytology , Neural Stem Cells/cytology , Regeneration , Retinal Neurons/cytology , Zebrafish/metabolism , Animals , Asymmetric Cell Division/drug effects , Biomarkers/metabolism , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Dedifferentiation/drug effects , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Heterozygote , Models, Biological , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neuroepithelial Cells/cytology , Neuroepithelial Cells/metabolism , Neurogenesis/drug effects , Ouabain/pharmacology , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Regeneration/drug effects , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Neurons/drug effects , Retinal Neurons/metabolism , Zebrafish Proteins/metabolism
18.
Neurochem Int ; 58(8): 888-95, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21338645

ABSTRACT

Fish retinal ganglion cells (RGCs) can survive and regrow their axons after optic nerve injury. Injured RGCs express anti-apoptotic proteins, such as Bcl-2, after nerve injury; however, upstream effectors of this anti-apoptotic protein are not yet fully understood. Heat shock proteins (HSPs) play a crucial role in cell survival against various stress conditions. In this study, we focused on HSP70 expression in the zebrafish retina after optic nerve injury. HSP70 mRNA and protein levels increased rapidly 2.3-fold in RGCs by 1-6 h after injury and returned to control levels by 1-3 days. HSP70 transcription is regulated by heat shock factor 1 (HSF1). HSF1 mRNA and phosphorylated-HSF1 protein rapidly increased by 2.2-fold in RGCs 0.5-6 h after injury. Intraocular injection of HSP inhibitor I significantly suppressed the induction of HSP70 expression after nerve injury. It also suppressed Bcl-2 protein induction and resulted in TUNEL-positive cell death of RGCs at 5 days post-injury. Zebrafish treated with HSP inhibitor I retarded axonal elongation or visual function after injury, as analyzed by GAP43 expression and behavioral analysis of optomotor response, respectively. These results strongly indicate that HSP70, the earliest induced gene in the zebrafish retina after optic nerve injury, is a crucial factor for RGCs survival and optic nerve regeneration in fish.


Subject(s)
HSP70 Heat-Shock Proteins/biosynthesis , Nerve Regeneration/physiology , Optic Nerve/physiology , Transcription, Genetic/physiology , Zebrafish Proteins/biosynthesis , Animals , Cell Survival/physiology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/physiology , Zebrafish
19.
Adv Exp Med Biol ; 664: 97-104, 2010.
Article in English | MEDLINE | ID: mdl-20238007

ABSTRACT

In adult visual system, goldfish can regrow their axons and fully restore their visual function even after optic nerve transection. The optic nerve regeneration process in goldfish is very long and it takes about a half year to fully recover visual function via synaptic refinement. Therefore, we investigated time course of growth-associated protein 43 (GAP43) expression in the goldfish retina for over 6 months after axotomy. In the control retina, very weak immunoreactivity could be seen in the retinal ganglion cells (RGCs). The immunoreactivity of GAP43 started to increase in the RGCs at 5 days, peaked at 7-20 days and then gradually decreased at 30-40 days after axotomy. The weak but significant immunoreactivity of GAP43 in the RGCs continued during 50-90 days and slowly returned to the control level by 180 days after lesion. The levels of GAP43 mRNA showed a biphasic pattern; a short-peak increase (9-folds) at 1-3 weeks and a long plateau increase (5-folds) at 50-120 days after axotomy. Thereafter, the levels declined to the control value by 180 days after axotomy. The changes of chasing behavior of pair of goldfish with bilaterally axotomized optic nerve also showed a slow biphasic recovery pattern in time course. Although further experiment is needed to elucidate the role of GAP43 in the regrowing axon terminals, the GAP43 is a good biochemical marker for monitoring the whole period of optic nerve regeneration in fish.


Subject(s)
GAP-43 Protein/metabolism , Goldfish/metabolism , Nerve Regeneration/physiology , Optic Nerve/metabolism , Optic Nerve/physiopathology , Animals , Axotomy , Behavior, Animal , Biomarkers/metabolism , GAP-43 Protein/genetics , Gene Expression Regulation , Immunohistochemistry , Optic Nerve/pathology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Retina/pathology , Time Factors
20.
Adv Exp Med Biol ; 664: 517-24, 2010.
Article in English | MEDLINE | ID: mdl-20238054

ABSTRACT

Recently, we cloned a photoreceptor-specific purpurin cDNA from axotomized goldfish retina. In the present study, we investigate the structure of zebrafish purpurin genomic DNA and its function during retinal development. First, we cloned a 3.7-kbp genomic DNA fragment including 1.4-kbp 5'-flanking region and 2.3-kbp full-length coding region. In the 1.4-kbp 5'-upstream region, there were some cone-rod homeobox (crx) protein binding motifs. The vector of the 1.4-kbp 5'-flanking region combined with the reporter GFP gene showed specific expression of this gene only in the photoreceptors. Although the first appearance time of purpurin mRNA expression was a little bit later (40 hpf) than that of crx (17-24 hpf), the appearance site was identical to the ventral part of the retina. Next, we made purpurin or crx knock down embryos with morpholino antisense oligonucleotides. The both morphants (purpurin and crx) showed similar abnormal phenotypes in the eye development; small size of eyeball and lacking of retinal lamination. Furthermore, co-injection of crx morpholino and purpurin mRNA significantly rescued these abnormalities. These data strongly indicate that purpurin is a key molecule for the cell differentiation during early retinal development in zebrafish under transcriptional crx regulation.


Subject(s)
Embryo, Nonmammalian/abnormalities , Gene Knockdown Techniques , Retina/abnormalities , Retinol-Binding Proteins/deficiency , Retinol-Binding Proteins/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Organ Specificity/drug effects , Organ Specificity/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology , Tolonium Chloride , Trans-Activators/metabolism , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL