Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Development ; 150(23)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37971210

ABSTRACT

Recent studies have demonstrated the impact of pro-inflammatory signaling and reactive microglia/macrophages on the formation of Müller glial-derived progenitor cells (MGPCs) in the retina. In chick retina, ablation of microglia/macrophages prevents the formation of MGPCs. Analyses of single-cell RNA-sequencing chick retinal libraries revealed that quiescent and activated microglia/macrophages have a significant impact upon the transcriptomic profile of Müller glia (MG). In damaged monocyte-depleted retinas, MG fail to upregulate genes related to different cell signaling pathways, including those related to Wnt, heparin-binding epidermal growth factor (HBEGF), fibroblast growth factor (FGF) and retinoic acid receptors. Inhibition of GSK3ß, to simulate Wnt signaling, failed to rescue the deficit in MGPC formation, whereas application of HBEGF or FGF2 completely rescued the formation of MGPCs in monocyte-depleted retinas. Inhibition of Smad3 or activation of retinoic acid receptors partially rescued the formation of MGPCs in monocyte-depleted retinas. We conclude that signals produced by reactive microglia/macrophages in damaged retinas stimulate MG to upregulate cell signaling through HBEGF, FGF and retinoic acid, and downregulate signaling through TGFß/Smad3 to promote the reprogramming of MG into proliferating MGPCs.


Subject(s)
Fibroblast Growth Factor 2 , Microglia , Animals , Microglia/metabolism , Fibroblast Growth Factor 2/metabolism , Neuroglia/metabolism , Ependymoglial Cells/metabolism , Stem Cells , Chickens , Retina/metabolism , Macrophages , Wnt Signaling Pathway , Receptors, Retinoic Acid/metabolism , EGF Family of Proteins/metabolism , Heparin/pharmacology , Heparin/metabolism , Cell Proliferation/genetics
2.
Differentiation ; 132: 51-58, 2023.
Article in English | MEDLINE | ID: mdl-37069005

ABSTRACT

Retinal development is initiated by multipotent retinal progenitor cells, which undergo several rounds of cell divisions and subsequently terminal differentiation. Retinal regeneration is usually considered as the recapitulation of retinal development, which share common mechanisms underlying the cell cycle re-entry of adult retinal stem cells and the differentiation of retinal neurons. However, how proliferative retinal progenitor cells perform a precise transition to postmitotic retinal cell types during the process of development and regeneration remains elusive. It is proposed that both the intrinsic and extrinsic programming are involved in the transcriptional regulation of the spatio-temporal fate commitment. Epigenetic modifications and the regulatory mechanisms at both DNA and chromatin levels are also postulated to play an important role in the timing of differentiation of specific retinal cells. In the present review, we have summarized recent knowledge of epigenetic regulation that underlies the commitment of retinal progenitor cells in the settings of retinal development and regeneration.


Subject(s)
Epigenesis, Genetic , Retina , Cell Differentiation/genetics , Stem Cells , Neurons
3.
Glia ; 71(12): 2866-2883, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37584502

ABSTRACT

The zebrafish retina possesses tremendous regenerative potential. Müller glia underlie retinal regeneration through their ability to reprogram and generate multipotent neuronal progenitors that re-differentiate into lost neurons. Many factors required for Müller glia reprogramming and proliferation have been identified; however, we know little about the epigenetic and transcriptional regulation of these genes during regeneration. Here, we determined whether transcriptional regulation by members of the Bromodomain (Brd) family is required for Müller glia-dependent retinal regeneration. Our data demonstrate that three brd genes were expressed in Müller glia upon injury. brd2a and brd2b were expressed in all Müller glia and brd4 was expressed only in reprogramming Müller glia. Utilizing (+)-JQ1, a pharmacological inhibitor of Brd function, we demonstrate that transcriptional regulation by Brds plays a critical role in Müller glia reprogramming and regeneration. (+)-JQ1 treatment prevented cell cycle re-entry of Müller glia and the generation of neurogenic progenitors. Modulating the (+)-JQ1 exposure window, we identified the first 48 h post-injury as the time-period during which Müller glia reprogramming occurs. (+)-JQ1 treatments after 48 h post-injury had no effect on the re-differentiation of UV cones, indicating that Brd function is required only for Müller glia reprogramming and not subsequent specification/differentiation events. Brd inhibition also prevented the expression of reprogramming genes like ascl1a and lepb in Müller glia, but not effector genes like mmp9, nor did it affect microglial recruitment after injury. These results demonstrate that transcriptional regulation by Brds plays a critical role during Müller glia-dependent retinal regeneration in zebrafish.

4.
Adv Exp Med Biol ; 1415: 309-317, 2023.
Article in English | MEDLINE | ID: mdl-37440050

ABSTRACT

Nearly a billion people worldwide are affected by vision-impairing conditions, with retinal degenerative diseases being a major cause of blindness. Unfortunately, such diseases are often permanent and progressive, resulting in further degeneration and loss of sight, due to the human retina possessing little, if any, regenerative capacity. Despite numerous efforts and great progress being made to understand the molecular mechanisms of these diseases and possible therapies, the majority of investigations focused on cell-intrinsic factors. However, the microenvironment surrounding retinal cells throughout these processes also plays an important role, though our current understanding of its involvement remains limited. Here we present a brief overview of the current state of the field of extracellular matrix studies within the retina and its potential roles in retinal diseases and potential therapeutic approaches.


Subject(s)
Extracellular Matrix , Retinal Degeneration , Humans , Extracellular Matrix Proteins , Retina
5.
Development ; 146(23)2019 12 02.
Article in English | MEDLINE | ID: mdl-31792065

ABSTRACT

As with all glial cells, the major role of retinal Müller glia (MG) is to provide essential neuronal support. However, the MG of some non-mammalian species have the additional ability to generate new retinal neurons capable of sight restoration. Unfortunately, mammalian MG do not possess this ability. However, if we could understand the reasons why, we may be able to devise strategies to confer regenerative potential. The recent discovery that the Hippo signaling pathway acts as an intrinsic block to mammalian MG proliferation, along with reports of adeno-associated virus (AAV)-based MG reprogramming and functional photoreceptor differentiation, may indicate a watershed moment in the field of mammalian retinal regeneration. However, as researchers delve deeper into the cellular and molecular mechanisms, and further refine MG reprogramming strategies, we should recall past misinterpretations of data in this field and proceed with caution. Here, we provide a summary of these emerging data and a discussion of technical concerns specific to AAV-mediated reprogramming experiments that must be addressed in order for the field to move forward.


Subject(s)
Cell Proliferation , Cellular Reprogramming Techniques , Cellular Reprogramming , Ependymoglial Cells/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Regeneration , Animals , Dependovirus , Genetic Vectors , Humans
6.
Exp Eye Res ; 207: 108569, 2021 06.
Article in English | MEDLINE | ID: mdl-33839111

ABSTRACT

Retinal regeneration research offers hope to people affected by visual impairment due to disease and injury. Ongoing research has explored many avenues towards retinal regeneration, including those that utilizes implantation of devices, cells or targeted viral-mediated gene therapy. These results have so far been limited, as gene therapy only has applications for rare single-gene mutations and implantations are invasive and in the case of cell transplantation donor cells often fail to integrate with adult neurons. An alternative mode of retinal regeneration utilizes a stem cell population unique to vertebrate retina - Müller glia (MG). Endogenous MG can readily regenerate lost neurons spontaneously in zebrafish and to a very limited extent in mammalian retina. The use of adenosine triphosphate (ATP) has been shown to induce retinal degeneration and activation of the MG in mammals, but whether this is conserved to other vertebrate species including those with higher regenerative capacity remains unknown. In our study, we injected a single dose of ATP intravitreal in zebrafish to characterize the cell death and MG induced regeneration. We used TUNEL labelling on retinal sections to show that ATP caused localised death of photoreceptors and ganglion cells within 24 h. Histology of GFP-transgenic zebrafish and BrdU injected fish demonstrated that MG proliferation peaked at days 3 and 4 post-ATP injection. Using BrdU labelling and photoreceptor markers (Zpr1) we observed regeneration of lost rod photoreceptors at day 14. This study has been undertaken to allow for comparative studies between mammals and zebrafish that use the same specific induction method of injury, i.e. ATP induced injury to allow for direct comparison of across species to narrow down resulting differences that might reflect the differing regenerative capacity. The ultimate aim of this work is to recapitulate pro-neurogenesis Müller glia signaling in mammals to produce new neurons that integrate with the existing retinal circuit to restore vision.


Subject(s)
Adenosine Triphosphate/toxicity , Ependymoglial Cells/physiology , Nerve Regeneration/physiology , Neuroglia/physiology , Retinal Degeneration/chemically induced , Retinal Rod Photoreceptor Cells/physiology , Zebrafish/physiology , Animals , Apoptosis/drug effects , Cell Proliferation , Disease Models, Animal , Female , In Situ Nick-End Labeling , Intravitreal Injections , Male , Retinal Degeneration/physiopathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/pathology
7.
Cell Mol Neurobiol ; 41(1): 43-62, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32219603

ABSTRACT

Müller cells may have stem cell-like capability as they regenerate photoreceptor loss upon injury in some vertebrates, but not in mammals. Indeed, mammalian Müller cells undergo major cellular and molecular changes summarized as reactive gliosis. Transforming growth factor beta (TGFß) isoforms are multifunctional cytokines that play a central role, both in wound healing and in tissue repair. Here, we studied the role of TGFß isoforms and their signaling pathways in response to injury induction during tissue regeneration in zebrafish and scar formation in mouse. Our transcriptome analysis showed a different activation of canonical and non-canonical signaling pathways and how they shaped the injury response. In particular, TGFß3 promotes retinal regeneration via Smad-dependent canonical pathway upon regulation of junb gene family and mycb in zebrafish Müller cells. However, in mice, TGFß1 and TGFß2 evoke the p38MAPK signaling pathway. The activation of this non-canonical pathway leads to retinal gliosis. Thus, the regenerative versus reparative effect of the TGFß pathway observed may rely on the activation of different signaling cascades. This provides one explanation of the different injury response in zebrafish and mouse retina.


Subject(s)
Gliosis/pathology , Retinal Degeneration/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Fibrinolysis , Fibrosis , Gliosis/complications , Gliosis/diagnostic imaging , Green Fluorescent Proteins/metabolism , Kinetics , Lasers , MAP Kinase Signaling System , Mice, Transgenic , Plasminogen Activator Inhibitor 1/metabolism , Protein Isoforms/metabolism , Regeneration , Retinal Degeneration/complications , Retinal Degeneration/diagnostic imaging , Tomography, Optical Coherence , Transforming Growth Factor beta2/metabolism , Up-Regulation , Zebrafish
8.
J Cell Mol Med ; 22(12): 6176-6189, 2018 12.
Article in English | MEDLINE | ID: mdl-30255622

ABSTRACT

Retinal ganglion cell apoptosis and optic nerve degeneration are prevalent in aged patients, which may be related to the decrease in bone marrow (BM) stem cell number/function because of the possible cross-talk between the two organs. This pathological process is accelerated by retinal ischaemia-reperfusion (I/R) injury. This study investigated whether young BM stem cells can regenerate and repair the aged retina after acute I/R injury. Young BM stem cell antigen 1 positive (Sca-1+ ) or Sca-1- cells were transplanted into lethally irradiated aged recipient mice to generate Sca-1+ and Sca-1- chimaeras, respectively. The animals were housed for 3 months to allow the young Sca-1 cells to repopulate in the BM of aged mice. Retinal I/R was then induced by elevation of intraocular pressure. Better preservation of visual function was found in Sca-1+ than Sca-1- chimaeras 7 days after injury. More Sca-1+ cells homed to the retina than Sca-1- cells and more cells differentiated into glial and microglial cells in the Sca-1+ chimaeras. After injury, Sca-1+ cells in the retina reduced host cellular apoptosis, which was associated with higher expression of fibroblast growth factor 2 (FGF2) in the Sca-1+ chimaeras. Young Sca-1+ cells repopulated the stem cells in the aged retina and diminished cellular apoptosis after acute I/R injury through FGF2 and Akt signalling pathways.


Subject(s)
Antigens, Ly/genetics , Fibroblast Growth Factor 2/genetics , Membrane Proteins/genetics , Reperfusion Injury/therapy , Stem Cell Transplantation , Aging/metabolism , Aging/pathology , Animals , Apoptosis/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Gene Expression Regulation, Developmental , Humans , Mice , Optic Nerve/metabolism , Optic Nerve/pathology , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Retina/growth & development , Retina/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
9.
J Neurosci Res ; 96(3): 362-370, 2018 03.
Article in English | MEDLINE | ID: mdl-29030949

ABSTRACT

The neural retina hosts a unique specialized type of macroglial cell that not only preserves retinal homeostasis, function, and integrity but also may serve as a source of new neurons during regenerative processes: the Müller cell. Precise microRNA-driven mechanisms of gene regulation impel and direct the processes of Müller glia lineage acquisition from retinal progenitors during development, the triggering of their response to retinal degeneration and, in some cases, Müller cell reprogramming and regenerative events. In this review we survey the recent reports describing, through functional assays, the regulatory role of microRNAs in Müller cell physiology, differentiation potential, and retinal pathology. We discuss also the evidence based on expression analysis that points out the relevance of a Müller glia-specific microRNA signature that would orchestrate these processes.


Subject(s)
Diabetic Retinopathy/metabolism , MicroRNAs/biosynthesis , Neuroglia/metabolism , Retina/metabolism , Retina/pathology , Animals , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Humans , MicroRNAs/genetics , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Regeneration
10.
J Neurosci ; 35(47): 15612-34, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26609156

ABSTRACT

Loss of retinal neurons in adult zebrafish (Danio rerio) induces a robust regenerative response mediated by the reentry of the resident Müller glia into the cell cycle. Upon initiating Müller glia proliferation, their nuclei migrate along the apicobasal axis of the retina in phase with the cell cycle in a process termed interkinetic nuclear migration (INM). We examined the mechanisms governing this cellular process and explored its function in regenerating the adult zebrafish retina. Live-cell imaging revealed that the majority of Müller glia nuclei migrated to the outer nuclear layer (ONL) to divide. These Müller glia formed prominent actin filaments at the rear of nuclei that had migrated to the ONL. Inhibiting actin filament formation or Rho-associated coiled-coil kinase (Rock) activity, which is necessary for phosphorylation of myosin light chain and actin myosin-mediated contraction, disrupted INM with increased numbers of mitotic nuclei remaining in the basal inner nuclear layer, the region where Müller glia typically reside. Double knockdown of Rho-associated coiled-coil kinase 2a (Rock2a) and Rho-associated coiled-coil kinase 2b (Rock2b) similarly disrupted INM and reduced Müller glial cell cycle reentry. In contrast, Rock inhibition immediately before the onset of INM did not affect Müller glia proliferation, but subsequently reduced neuronal progenitor cell proliferation due to early cell cycle exit. Long-term, Rock inhibition increased the generation of mislocalized ganglion/amacrine cells at the expense of rod and cone photoreceptors. In summary, INM is driven by an actin-myosin-mediated process controlled by Rock2a and Rock2b activity, which is required for sufficient proliferation and regeneration of photoreceptors after light damage. SIGNIFICANCE STATEMENT: The human retina does not replace lost or damaged neurons, ultimately causing vision impairment. In contrast, zebrafish are capable of regenerating lost neurons. Understanding the mechanisms that regulate retinal regeneration in these organisms will help to elucidate approaches to stimulate a similar response in humans. In the damaged zebrafish retina, Müller glia dedifferentiate and proliferate to generate neuronal progenitor cells (NPCs) that differentiate into the lost neurons. We show that the nuclei of Müller glia and NPCs migrate apically and basally in phase with the cell cycle. This migration is facilitated by the actin cytoskeleton and Rho-associated coiled-coil kinases (Rocks). We demonstrate that Rock function is required for sufficient proliferation and the regeneration of photoreceptors, likely via regulating nuclear migration.


Subject(s)
Actins/physiology , Cell Movement/physiology , Cell Nucleus/physiology , Cytoskeleton/physiology , Nerve Regeneration/physiology , Retinal Cone Photoreceptor Cells/physiology , Zebrafish Proteins/physiology , rho-Associated Kinases/physiology , Age Factors , Animals , Animals, Genetically Modified , Cells, Cultured , Female , Male , Retina/cytology , Retina/physiology , Zebrafish
11.
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
12.
Zoolog Sci ; 33(1): 21-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26853865

ABSTRACT

The adult newt has the remarkable ability to regenerate a functional retina from retinal pigment epithelium (RPE) cells, even when the neural retina (NR) is completely lost from the eye. In this system, RPE cells are reprogrammed into a unique state of multipotent cells, named RPESCs, in an early phase of retinal regeneration. However, the signals that trigger reprogramming remain unknown. Here, to approach this issue we focused on Pax6, a transcription factor known to be expressed in RPESCs. We first identified four classes (v1, v2, v3 and v4) of Pax6 variants in the eye of adult newt, Cynops pyrrhogaster. These variants were expressed in most tissues of the intact eye in different combinations but not in the RPE, choroid or sclera. On the basis of this information, we investigated the expression of Pax6 in RPE cells after the NR was removed from the eye by surgery (retinectomy), and found that two classes (v1 and v2) of Pax6 variants were newly expressed in RPE cells 10 days after retinectomy, both in vivo and in vitro (RLEC system). In the RLEC system, we found that Pax6 expression is mediated through a pathway separate from the MEK-ERK pathway, which is required for cell cycle re-entry of RPE cells. These results predict the existence of a pathway that may be of fundamental importance to a better understanding of the reprogramming of RPE cells in vivo.


Subject(s)
Eye Proteins/metabolism , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/physiology , Salamandridae/physiology , Animals , Base Sequence , Butadienes/pharmacology , DNA/genetics , Enzyme Inhibitors/pharmacology , Eye Proteins/genetics , Gene Expression Regulation/drug effects , Genetic Variation , Homeodomain Proteins/genetics , Nitriles/pharmacology , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics
13.
Adv Exp Med Biol ; 854: 587-93, 2016.
Article in English | MEDLINE | ID: mdl-26427463

ABSTRACT

In the adult zebrafish, death of retinal neurons stimulates Müller glia to re-enter the cell cycle to produce neuronal progenitor cells (NPCs) that undergo further cell divisions and differentiate to replace lost neurons in the correct spatial locations. Understanding the mechanisms regulating retinal regeneration will ultimately provide avenues to overcome vision loss in human. Recently, the observation of interkinetic nuclear migration (INM) of Müller glia in the regenerating zebrafish retina resulted in the inclusion of an additional complex step to the regeneration process. The pathways regulating INM and its function in the regenerating retina have not been well studied. Here, we summarize the evidence for INM in the regenerating retina and review mechanisms that control INM during neuro-epithelial development in the context of pathways known to be critical during retinal regeneration.


Subject(s)
Cell Movement/physiology , Ependymoglial Cells/physiology , Retina/physiology , Retinal Neurons/physiology , Animals , Cell Nucleus/physiology , Humans , Neurogenesis , Regeneration , Retina/cytology , Zebrafish/physiology
14.
J Neurosci ; 34(43): 14403-19, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25339752

ABSTRACT

Retinal damage in teleosts, unlike mammals, induces robust Müller glia-mediated regeneration of lost neurons. We examined whether Notch signaling regulates Müller glia proliferation in the adult zebrafish retina and demonstrated that Notch signaling maintains Müller glia in a quiescent state in the undamaged retina. Repressing Notch signaling, through injection of the γ-secretase inhibitor RO4929097, stimulates a subset of Müller glia to reenter the cell cycle without retinal damage. This RO4929097-induced Müller glia proliferation is mediated by repressing Notch signaling because inducible expression of the Notch Intracellular Domain (NICD) can reverse the effect. This RO4929097-induced proliferation requires Ascl1a expression and Jak1-mediated Stat3 phosphorylation/activation, analogous to the light-damaged retina. Moreover, coinjecting RO4929097 and TNFα, a previously identified damage signal, induced the majority of Müller glia to reenter the cell cycle and produced proliferating neuronal progenitor cells that committed to a neuronal lineage in the undamaged retina. This demonstrates that repressing Notch signaling and activating TNFα signaling are sufficient to induce Müller glia proliferation that generates neuronal progenitor cells that differentiate into retinal neurons, mimicking the responses observed in the regenerating retina.


Subject(s)
Cell Proliferation/physiology , Ependymoglial Cells/physiology , Nerve Regeneration/physiology , Neural Stem Cells/physiology , Receptors, Notch/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Animals, Genetically Modified , Cell Proliferation/drug effects , Ependymoglial Cells/drug effects , Female , Gene Expression Regulation , Male , Nerve Regeneration/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurogenesis/physiology , Neuroglia/drug effects , Neuroglia/physiology , Retinal Neurons/drug effects , Retinal Neurons/physiology , Tumor Necrosis Factor-alpha/pharmacology , Zebrafish
15.
Dev Dyn ; 243(12): 1591-605, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25220904

ABSTRACT

BACKGROUND: Adult zebrafish spontaneously regenerate their retinas after damage. Although a number of genes and signaling pathways involved in regeneration have been identified, the exact mechanisms regulating various aspects of regeneration are unclear. microRNAs (miRNAs) were examined for their potential roles in regulating zebrafish retinal regeneration. RESULTS: To investigate the requirement of miRNAs during zebrafish retinal regeneration, we knocked down the expression of Dicer in retinas prior to light-induced damage. Reduced Dicer expression significantly decreased the number of proliferating Müller glia-derived neuronal progenitor cells during regeneration. To identify individual miRNAs with roles in neuronal progenitor cell proliferation, we collected retinas at different stages of light damage and performed small RNA high-throughput sequencing. We identified subsets of miRNAs that were differentially expressed during active regeneration but returned to basal levels once regeneration was completed. We then knocked down five different miRNAs that increased in expression and assessed the effects on retinal regeneration. Reduction of miR-142b and miR-146a expression significantly reduced INL proliferation at 51 h of light treatment, while knockdown of miR-7a, miR-27c, and miR-31 expression significantly reduced INL proliferation at 72 h of constant light. CONCLUSIONS: miRNAs exhibit dynamic expression profiles during retinal regeneration and are necessary for neuronal progenitor cell proliferation.


Subject(s)
Cell Proliferation/physiology , Gene Expression Regulation , MicroRNAs/biosynthesis , Neural Stem Cells/metabolism , Neuroglia/metabolism , Regeneration/physiology , Retina/physiology , Ribonuclease III/metabolism , Zebrafish Proteins/biosynthesis , Zebrafish/metabolism , Animals , Gene Knockdown Techniques , MicroRNAs/genetics , Ribonuclease III/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
16.
Angew Chem Int Ed Engl ; 54(46): 13555-60, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26383645

ABSTRACT

Continued activation of the photocycle of the dim-light receptor rhodopsin leads to the accumulation of all-trans-retinal in the rod outer segments (ROS). This accumulation can damage the photoreceptor cell. For retinal homeostasis, deactivation processes are initiated in which the release of retinal is delayed. One of these processes involves the binding of arrestin to rhodopsin. Here, the interaction of pre-activated truncated bovine visual arrestin (Arr(Tr)) with rhodopsin in 1,2-diheptanoyl-sn-glycero-3-phosphocholine (DHPC) micelles is investigated by solution NMR techniques and flash photolysis spectroscopy. Our results show that formation of the rhodopsin-arrestin complex markedly influences partitioning in the decay kinetics of rhodopsin, which involves the simultaneous formation of a meta II and a meta III state from the meta I state. Binding of Arr(Tr) leads to an increase in the population of the meta III state and consequently to an approximately twofold slower release of all-trans-retinal from rhodopsin.


Subject(s)
Arrestin/chemistry , Arrestin/metabolism , Photochemical Processes , Rhodopsin/chemistry , Rhodopsin/metabolism , Animals , Cattle , Rhodopsin/radiation effects
17.
Exp Eye Res ; 123: 107-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23880527

ABSTRACT

The retinal pigment epithelium (RPE) is a partner of the neural retina and is indispensable for vision. In humans, proliferation and transformation (cell-type switching) of RPE cells after a traumatic injury of the neural retina causes a retinal disorder leading to loss of vision. In contrast, in certain adult amphibians such as Xenopus laevis and the newt, a similar process in RPE cells leads to regeneration of the entire retina. In this review, on the basis of accumulating evidence in basic biology and medical sciences, similarities and differences between these RPE-mediated retinal disorders and regeneration in adult vertebrates are highlighted, providing a connection to future research that should be designed to establish clues for the treatment of pathogenesis caused by RPE while promoting RPE-mediated retinal regeneration in a patient's eyes.


Subject(s)
Regeneration/physiology , Retina/physiology , Retinal Pigment Epithelium/physiopathology , Vitreoretinopathy, Proliferative/physiopathology , Animals , Humans
18.
Int J Radiat Biol ; 100(1): 131-138, 2024.
Article in English | MEDLINE | ID: mdl-37555698

ABSTRACT

PURPOSE: Zebrafish, a small fish model, exhibits a multipotent ability for retinal regeneration after damage throughout its lifetime. Compared with zebrafish, birds and mammals exhibit such a regenerative capacity only during the embryonic period, and this capacity decreases with age. In medaka, another small fish model that has also been used extensively in biological research, the retina's inner nuclear layer (INL) failed to regenerate after injury in the hatchling at eight days postfertilization (dpf). We characterized the regenerative process of the embryonic retina when the retinal injury occurred during the early embryonic period in medaka. METHODS: We employed a 10 Gy dose of gamma-ray irradiation to initiate retinal injury in medaka embryos at 3 dpf and performed histopathological analyses up to 21 dpf. RESULTS: One day after irradiation, numerous apoptotic neurons were observed in the INL; however, these neurons were rarely observed in the ciliary marginal zone and the photoreceptor layer. Numerous pyknotic cells were clustered in the irradiated retina until two days after irradiation. These disappeared four days after irradiation, but the abnormal bridging structures between the INL and ganglion cell layer (GCL) were present until 11 days after irradiation, and the neural layers were completely regenerated 18 days after irradiation. After gamma-ray irradiation, the spindle-like Müller glial cells in the INL became rounder but did not lose their ability to express SOX2. CONCLUSIONS: Irradiated retina at 3 dpf of medaka embryos could be completely regenerated at 18 days after irradiation (21 dpf), although the abnormal layer structures bridging the INL and GCL were transiently formed in the retinas of all the irradiated embryos. Four days after irradiation, embryonic medaka Müller glia were reduced in number but maintained SOX2 expression as in nonirradiated embryos. This finding contrasts with previous reports that 8 dpf medaka larvae could not fully regenerate damaged retinas because of loss of SOX2 expression.


Subject(s)
Oryzias , Animals , Zebrafish , Retina/injuries , Retina/pathology , Neuroglia , Embryonic Development , Mammals
19.
Cureus ; 16(2): e53479, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38440034

ABSTRACT

Retinitis pigmentosa and age-related macular degeneration are the most frequent causes of irreversible visual impairment in the world. Existing therapeutic methods could be more effective, underscoring the necessity of new treatments. Reconstructing the retinal photoreceptors through the transplantation of human pluripotent stem cells, representing an attractive approach for restoring vision, has gained momentum. This paper gives an exhaustive account of what has been known in this field, the discoveries made, and the recent progress. This review paper outlines the retina's organisation, cell types, the pathophysiology of retinal injury/degeneration, and the reasoning behind using pluripotent stem cells in retinal regeneration. This article investigates differentiation strategies, molecular components that dictate cell type specification, and the recreation of retinal development in vitro, genetically engineering and manipulating epigenetic marks using various techniques for driving specific cell fates and improving therapy efficacy. Subretinal injection methods, cell encapsulation techniques, scaffold-based approaches, cell sheet transplantation, and their impact on integrating implanted cells into a functional retina are thoroughly reviewed. Using bioengineering approaches, biomaterials and growth factors form a favourable micro-ambience for grafted cells. Issues around safety and efficacy (tumorigenicity, immunological rejection, and long-term integration/functionality) are explored. Moreover, the paper emphasises the significance of rigorous characterisation, immunomodulatory strategies, and clinical and pre-clinical studies to ensure the safety and effectiveness of retinal regeneration therapy. Future perspectives and challenges are presented, looking at fine-tuning differentiation strategies, improving functional integration and regulatory aspects, and using co-therapy and supportive treatments.

20.
Stem Cell Res ; 66: 103006, 2023 02.
Article in English | MEDLINE | ID: mdl-36563542

ABSTRACT

Müller glia are non-neuronal support cells that play a vital role in the homeostasis of the eye. Their radial-oriented processes span the width of the retina and respond to injury through a cellular response that can be detrimental or protective depending on the context. In some species, protective responses include the expression of stem cell-like genes which help to fuel new neuron formation and even restoration of vision. In many lower vertebrates including fish and amphibians, this response is well documented, however, in mammals it is severely limited. The remarkable plasticity of cellular reprogramming in lower vertebrates has inspired studies in mammals for repairing the retina and restoring sight, and recent studies suggest that mammals are also capable of regeneration, albeit to a lesser degree. Endogenous regeneration, whereby new retinal neurons are created from existing support cells, offers an exciting alternative approach to existing tissue transplant, gene therapy, and neural prosthetic approaches being explored in parallel. This review will highlight the role of Müller glia during retinal injury and repair. In the end, prospects for advancing retinal regeneration research will be considered.


Subject(s)
Cellular Reprogramming , Neuroglia , Animals , Neuroglia/metabolism , Retina/metabolism , Ependymoglial Cells/metabolism , Neurons , Cell Proliferation/physiology , Mammals
SELECTION OF CITATIONS
SEARCH DETAIL