Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-37808650

ABSTRACT

Retinal degeneration in mammals causes permanent loss of vision, due to an inability to regenerate naturally. Some non-mammalian vertebrates show robust regeneration, via Muller glia (MG). We have recently made significant progress in stimulating adult mouse MG to regenerate functional neurons by transgenic expression of the proneural transcription factor Ascl1. While these results showed that MG can serve as an endogenous source of neuronal replacement, the efficacy of this process is limited. With the goal of improving this in mammals, we designed a small molecule screen using sci-Plex, a method to multiplex up to thousands of single nucleus RNA-seq conditions into a single experiment. We used this technology to screen a library of 92 compounds, identified, and validated two that promote neurogenesis in vivo. Our results demonstrate that high-throughput single-cell molecular profiling can substantially improve the discovery process for molecules and pathways that can stimulate neural regeneration and further demonstrate the potential for this approach to restore vision in patients with retinal disease.

2.
Proc Natl Acad Sci U S A ; 120(46): e2302089120, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37931105

ABSTRACT

Ongoing cell therapy trials have demonstrated the need for precision control of donor cell behavior within the recipient tissue. We present a methodology to guide stem cell-derived and endogenously regenerated neurons by engineering the microenvironment. Being an "approachable part of the brain," the eye provides a unique opportunity to study neuron fate and function within the central nervous system. Here, we focused on retinal ganglion cells (RGCs)-the neurons in the retina are irreversibly lost in glaucoma and other optic neuropathies but can potentially be replaced through transplantation or reprogramming. One of the significant barriers to successful RGC integration into the existing mature retinal circuitry is cell migration toward their natural position in the retina. Our in silico analysis of the single-cell transcriptome of the developing human retina identified six receptor-ligand candidates, which were tested in functional in vitro assays for their ability to guide human stem cell-derived RGCs. We used our lead molecule, SDF1, to engineer an artificial gradient in the retina, which led to a 2.7-fold increase in donor RGC migration into the ganglion cell layer (GCL) and a 3.3-fold increase in the displacement of newborn RGCs out of the inner nuclear layer. Only donor RGCs that migrated into the GCL were found to express mature RGC markers, indicating the importance of proper structure integration. Together, these results describe an "in silico-in vitro-in vivo" framework for identifying, selecting, and applying soluble ligands to control donor cell function after transplantation.


Subject(s)
Retina , Retinal Ganglion Cells , Infant, Newborn , Humans , Stem Cells , Neurogenesis , Cell Movement
3.
Adv Exp Med Biol ; 1415: 577-582, 2023.
Article in English | MEDLINE | ID: mdl-37440089

ABSTRACT

Endogenous regeneration strategies to replace lost neurons hold great promise for treating neurodegenerative disorders. In the majority of cases, neural regeneration is induced by converting resident glial cells into neurogenic precursors. This review will outline how proneural bHLH transcription factors can be used to reprogram glia in the brain and retina into a source for new neurons.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Neuroglia , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/physiology , Neuroglia/physiology , Nerve Regeneration/physiology , Neurons/physiology , Retina/physiology
4.
Sci Adv ; 8(47): eabq7219, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36417510

ABSTRACT

Many neurodegenerative diseases cause degeneration of specific types of neurons. For example, glaucoma leads to death of retinal ganglion cells, leaving other neurons intact. Neurons are not regenerated in the adult mammalian central nervous system. However, in nonmammalian vertebrates, glial cells spontaneously reprogram into neural progenitors and replace neurons after injury. We have recently developed strategies to stimulate regeneration of functional neurons in the adult mouse retina by overexpressing the proneural factor Ascl1 in Müller glia. Here, we test additional transcription factors (TFs) for their ability to direct regeneration to particular types of retinal neurons. We engineered mice to express different combinations of TFs in Müller glia, including Ascl1, Pou4f2, Islet1, and Atoh1. Using immunohistochemistry, single-cell RNA sequencing, single-cell assay for transposase-accessible chromatin sequencing, and electrophysiology, we find that retinal ganglion-like cells can be regenerated in the damaged adult mouse retina in vivo with targeted overexpression of developmental retinal ganglion cell TFs.


Subject(s)
Retina , Transcription Factors , Mice , Animals , Transcription Factors/genetics , Neuroglia , Neurons , Mammals
5.
Glia ; 70(7): 1380-1401, 2022 07.
Article in English | MEDLINE | ID: mdl-35388544

ABSTRACT

Müller glia (MG) in mammalian retinas are incapable of regenerating neurons after damage, whereas the MG in lower vertebrates regenerate functional neurons. Identification of cell signaling pathways and gene regulatory networks that regulate MG-mediated regeneration is key to harnessing the regenerative potential of MG. Here, we study how NFkB-signaling influences glial responses to damage and reprogramming of MG into neurons in the rodent retina. We find activation of NFkB and dynamic expression of NFkB-associated genes in MG after damage, however damage-induced NFkB activation is inhibited by microglia ablation. Knockout of NFkB in MG suppressed the accumulation of immune cells after damage. Inhibition of NFkB following NMDA-damage significantly enhanced the reprogramming of Ascl1-overexpressing MG into neuron-like cells. scRNA-seq of retinal glia following inhibition of NFkB reveals coordination with signaling via TGFß2 and suppression of NFI and Id transcription factors. Inhibition of Smad3 signal transducer or Id transcription factors increased numbers of neuron-like cells produced by Ascl1-overexpressing MG. We conclude that NFkB is a key signaling hub that is activated in MG after damage, mediates the accumulation of immune cells, and suppresses the neurogenic potential of MG.


Subject(s)
Ependymoglial Cells , Neuroglia , Animals , Cell Proliferation/physiology , Ependymoglial Cells/metabolism , Mammals/metabolism , NF-kappa B/metabolism , Neuroglia/metabolism , Neurons/metabolism , Regeneration , Retina , Signal Transduction , Transcription Factors/metabolism
6.
Article in English | MEDLINE | ID: mdl-34580118

ABSTRACT

The regenerative capacity of the vertebrate retina varies substantially across species. Whereas fish and amphibians can regenerate functional retina, mammals do not. In this perspective piece, we outline the various strategies nonmammalian vertebrates use to achieve functional regeneration of vision. We review key differences underlying the regenerative potential across species including the cellular source of postnatal progenitors, the diversity of cell fates regenerated, and the level of functional vision that can be achieved. Finally, we provide an outlook on the field of engineering the mammalian retina to replace neurons lost to injury or disease.


Subject(s)
Cellular Reprogramming , Vertebrates , Animals , Biology , Mammals , Nerve Regeneration/physiology , Retina/physiology
7.
Glia ; 70(4): 661-674, 2022 04.
Article in English | MEDLINE | ID: mdl-34939240

ABSTRACT

Ischemic preconditioning (IPC) is a phenomenon whereby a brief, non-injurious ischemic exposure enhances tolerance to a subsequent ischemic challenge. The mechanism of IPC has mainly been studied in rodent stroke models where gray matter (GM) constitutes about 85% of the cerebrum. In humans, white matter (WM) is 50% of cerebral volume and is a critical component of stroke damage. We developed a novel CNS WM IPC model using the mouse optic nerve (MON) and identified the involved immune signaling pathways. Here we tested the hypothesis that microglia are necessary for WM IPC. Microglia were depleted by treatment with the colony stimulating factor 1 receptor (CSF1R) inhibitor PLX5622. MONs were exposed to transient ischemia in vivo, acutely isolated 72 h later, and subjected to oxygen-glucose deprivation (OGD) to simulate a severe ischemic injury (i.e., stroke). Functional and structural axonal recovery was assessed by recording compound action potentials (CAPs) and by microscopy using quantitative stereology. Microglia depletion eliminated IPC-mediated protection. In control mice, CAP recovery was improved in preconditioned MONs compared with non-preconditioned MONs, however, in PLX5622-treated mice, we observed no difference in CAP recovery between preconditioned and non-preconditioned MONs. Microgliadepletion also abolished IPC protective effects on axonal integrity and survival of mature (APC+ ) oligodendrocytes after OGD. IPC-mediated protection was independent of retinal injury suggesting it results from mechanistic processes intrinsic to ischemia-exposed WM. We conclude that preconditioned microglia are critical for IPC in WM. The "preconditioned microglia" phenotype might protect against other CNS pathologies and is a neurotherapeutic horizon worth exploring.


Subject(s)
Ischemic Preconditioning , Stroke , White Matter , Animals , Cerebral Cortex/metabolism , Ischemic Preconditioning/methods , Mice , Microglia/metabolism , Stroke/metabolism , White Matter/metabolism
8.
Cell Rep ; 37(3): 109857, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686336

ABSTRACT

Regenerative neuroscience aims to stimulate endogenous repair in the nervous system to replace neurons lost from degenerative diseases. Recently, we reported that overexpressing the transcription factor Ascl1 in Müller glia (MG) is sufficient to stimulate MG to regenerate functional neurons in the adult mouse retina. However, this process is inefficient, and only a third of the Ascl1-expressing MG generate new neurons. Here, we test whether proneural transcription factors of the Atoh1/7 class can further promote the regenerative capacity of MG. We find that the combination of Ascl1:Atoh1 is remarkably efficient at stimulating neurogenesis, even in the absence of retinal injury. Using electrophysiology and single-cell RNA sequencing (scRNA-seq), we demonstrate that Ascl1:Atoh1 generates a diversity of retinal neuron types, with the majority expressing characteristics of retinal ganglion cells. Our results provide a proof of principle that combinations of developmental transcription factors can substantially improve glial reprogramming to neurons and expand the repertoire of regenerated cell fates.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Ependymoglial Cells/metabolism , Nerve Regeneration , Nerve Tissue Proteins/metabolism , Neurogenesis , Retina/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Ependymoglial Cells/pathology , Female , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Phenotype , RNA-Seq , Retina/pathology , Signal Transduction , Single-Cell Analysis
9.
Cell Rep ; 33(11): 108507, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326790

ABSTRACT

The innate immune system plays key roles in tissue regeneration. For example, microglia promote neurogenesis in Müller glia in birds and fish after injury. Although mammalian retina does not normally regenerate, neurogenesis can be induced in mouse Müller glia by Ascl1, a proneural transcription factor. We show that in mice, microglia inhibit the Ascl1-mediated retinal regeneration, suggesting that the innate immune system limits the regenerative response to injury.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Microglia/immunology , Nerve Regeneration/immunology , Retina/physiopathology , Animals , Mice
10.
Science ; 370(6519)2020 11 20.
Article in English | MEDLINE | ID: mdl-33004674

ABSTRACT

Injury induces retinal Müller glia of certain cold-blooded vertebrates, but not those of mammals, to regenerate neurons. To identify gene regulatory networks that reprogram Müller glia into progenitor cells, we profiled changes in gene expression and chromatin accessibility in Müller glia from zebrafish, chick, and mice in response to different stimuli. We identified evolutionarily conserved and species-specific gene networks controlling glial quiescence, reactivity, and neurogenesis. In zebrafish and chick, the transition from quiescence to reactivity is essential for retinal regeneration, whereas in mice, a dedicated network suppresses neurogenic competence and restores quiescence. Disruption of nuclear factor I transcription factors, which maintain and restore quiescence, induces Müller glia to proliferate and generate neurons in adult mice after injury. These findings may aid in designing therapies to restore retinal neurons lost to degenerative diseases.


Subject(s)
Cellular Reprogramming/genetics , Ependymoglial Cells/cytology , Gene Regulatory Networks , Nerve Regeneration/genetics , Neurogenesis/genetics , Animals , Chickens , Gene Expression Regulation, Developmental , Mice , RNA-Seq , Zebrafish
11.
Cell Rep ; 30(7): 2195-2208.e5, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075759

ABSTRACT

Müller glia (MG) serve as sources for retinal regeneration in non-mammalian vertebrates. We find that this process can be induced in mouse MG, after injury, by transgenic expression of the proneural transcription factor Ascl1 and the HDAC inhibitor TSA. However, new neurons are generated only from a subset of MG. Identifying factors that limit Ascl1-mediated MG reprogramming could make this process more efficient. In this study, we test whether injury-induced STAT activation hampers the ability of Ascl1 to reprogram MG into retinal neurons. Single-cell RNA-seq shows that progenitor-like cells derived from Ascl1-expressing MG have a higher level of STAT signaling than do those cells that become neurons. Ascl1-ChIPseq and ATAC-seq show that STAT potentially directs Ascl1 to developmentally inappropriate targets. Using a STAT inhibitor, in combination with our previously described reprogramming paradigm, we found a large increase in the ability of MG to generate neurons.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin/metabolism , Nerve Regeneration/physiology , Neuroglia/physiology , Neurons/metabolism , STAT Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Mice , Mice, Inbred C57BL , Neurogenesis , Signal Transduction
12.
J Neuroinflammation ; 16(1): 118, 2019 Jun 06.
Article in English | MEDLINE | ID: mdl-31170999

ABSTRACT

BACKGROUND: Microglia and inflammation have context-specific impacts upon neuronal survival in different models of central nervous system (CNS) disease. Herein, we investigate how inflammatory mediators, including microglia, interleukin 1 beta (IL1ß), and signaling through interleukin 1 receptor type 1 (IL-1R1), influence the survival of retinal neurons in response to excitotoxic damage. METHODS: Excitotoxic retinal damage was induced via intraocular injections of NMDA. Microglial phenotype and neuronal survival were assessed by immunohistochemistry. Single-cell RNA sequencing was performed to obtain transcriptomic profiles. Microglia were ablated by using clodronate liposome or PLX5622. Retinas were treated with IL1ß prior to NMDA damage and cell death was assessed in wild type, IL-1R1 null mice, and mice expressing IL-1R1 only in astrocytes. RESULTS: NMDA-induced damage included neuronal cell death, microglial reactivity, upregulation of pro-inflammatory cytokines, and genes associated with IL1ß-signaling in different types of retinal neurons and glia. Expression of the IL1ß receptor, IL-1R1, was evident in astrocytes, endothelial cells, some Müller glia, and OFF bipolar cells. Ablation of microglia with clodronate liposomes or Csf1r antagonist (PLX5622) resulted in elevated cell death and diminished neuronal survival in excitotoxin-damaged retinas. Exogenous IL1ß stimulated the proliferation and reactivity of microglia in the absence of damage, reduced numbers of dying cells in damaged retinas, and increased neuronal survival following an insult. IL1ß failed to provide neuroprotection in the IL-1R1-null retina, but IL1ß-mediated neuroprotection was rescued when expression of IL-1R1 was restored in astrocytes. CONCLUSIONS: We conclude that reactive microglia provide protection to retinal neurons, since the absence of microglia is detrimental to survival. We propose that, at least in part, the survival-influencing effects of microglia may be mediated by IL1ß, IL-1R1, and interactions of microglia and other macroglia.


Subject(s)
Interleukin-1beta/metabolism , Microglia/metabolism , Neuroprotection/physiology , Receptors, Interleukin-1 Type I/metabolism , Retina/pathology , Animals , Excitatory Amino Acid Agonists/toxicity , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , N-Methylaspartate/toxicity , Neurotoxins/toxicity , Receptors, Interleukin-1 Type I/immunology , Retina/immunology
13.
Exp Neurol ; 320: 112984, 2019 10.
Article in English | MEDLINE | ID: mdl-31251936

ABSTRACT

Gelatinases are a class of matrix metalloproteinases (MMPs) that degrade the extracellular matrix (ECM) to regulate intercellular signaling and cell migration. Gelatinase activity is tightly regulated via proteolytic activation and through the expression of tissue inhibitors of matrix metalloproteinases (TIMPs). Gelatinase activity has been implicated in retinal pathophysiology in different animal models and human disease. However, the role of gelatinases in retinal regeneration remains uncertain. In this study we investigated the dynamic changes in gelatinase activity in response to excitotoxic damage and how this enzymatic activity influenced the formation of Müller glia progenitor cells (MGPCs) in the avian retina. This study used hydrogels containing a gelatinase-degradable fluorescent peptide to measure gelatinase activity in vitro and dye quenched gelatin to localize enzymatic activity in situ. These data were corroborated by using single cell RNA sequencing (scRNA-seq). Gelatinase mRNA, specifically MMP2, was detected in oligodendrocytes and Non-Astrocytic Inner Retinal Glia (NIRG). Total retinal gelatinase activity was reduced following NMDA-treatment, and sustained inhibition of MMP2 prior to damage or growth factor treatment increased the formation of proliferating MGPCs and c-fos signaling. We observed that microglia, Müller glia (MG), and NIRG cells were involved in regulating changes in gelatinase activity through TIMP2 and TIMP3. Collectively, these findings implicate MMP2 in reprogramming of Muller glia into MGPCs.


Subject(s)
Cellular Reprogramming/physiology , Ependymoglial Cells/enzymology , Matrix Metalloproteinase 2/metabolism , Nerve Regeneration/physiology , Retina/enzymology , Animals , Cell Proliferation/physiology , Chickens , Gelatinases/metabolism , Stem Cells/enzymology
14.
Stem Cells ; 36(3): 392-405, 2018 03.
Article in English | MEDLINE | ID: mdl-29193451

ABSTRACT

In the retina, Müller glia have the potential to become progenitor cells with the ability to proliferate and regenerate neurons. However, the ability of Müller glia-derived progenitor cells (MGPCs) to proliferate and produce neurons is limited in higher vertebrates. Using the chick model system, we investigate how retinoic acid (RA)-signaling influences the proliferation and the formation of MGPCs. We observed an upregulation of cellular RA binding proteins (CRABP) in the Müller glia of damaged retinas where the formation of MGPCs is known to occur. Activation of RA-signaling was stimulated, whereas inhibition suppressed the proliferation of MGPCs in damaged retinas and in fibroblast growth factor 2-treated undamaged retinas. Furthermore, inhibition of RA-degradation stimulated the proliferation of MGPCs. Levels of Pax6, Klf4, and cFos were upregulated in MGPCs by RA agonists and downregulated in MGPCs by RA antagonists. Activation of RA-signaling following MGPC proliferation increased the percentage of progeny that differentiated as neurons. Similarly, the combination of RA and insulin-like growth factor 1 (IGF1) significantly increased neurogenesis from retinal progenitors in the circumferential marginal zone (CMZ). In summary, RA-signaling stimulates the formation of proliferating MGPCs and enhances the neurogenic potential of MGPCs and stem cells in the CMZ. Stem Cells 2018;36:392-405.


Subject(s)
Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Retina/cytology , Retina/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Tretinoin/metabolism , Animals , Neurogenesis/physiology , Neuroglia/cytology , Neuroglia/metabolism , Signal Transduction
15.
Glia ; 65(10): 1640-1655, 2017 10.
Article in English | MEDLINE | ID: mdl-28703293

ABSTRACT

Müller glia-derived progenitor cells (MGPCs) have the capability to regenerate neurons in the retinas of different vertebrate orders. The formation of MGPCs is regulated by a network of cell-signaling pathways. The purpose of this study was to investigate how BMP/Smad1/5/8- and TGFß/Smad2/3-signaling are coordinated to influence the formation of MGPCs in the chick model system. We find that pSmad1/5/8 is selectively up-regulated in the nuclei of Müller glia following treatment with BMP4, FGF2, or NMDA-induced damage, and this up-regulation is blocked by a dorsomorphin analogue DMH1. By comparison, Smad2/3 is found in the nuclei of Müller glia in untreated retinas, and becomes localized to the cytoplasm following NMDA- or FGF2-treatment. These findings suggest a decrease in TGFß- and increase in BMP-signaling when MGPCs are known to form. In both NMDA-damaged and FGF2-treated retinas, inhibition of BMP-signaling suppressed the proliferation of MGPCs, whereas inhibition of TGFß-signaling stimulated the proliferation of MGPCs. Consistent with these findings, TGFß2 suppressed the formation of MGPCs in NMDA-damaged retinas. Our findings indicate that BMP/TGFß/Smad-signaling is recruited into the network of signaling pathways that controls the formation of proliferating MGPCs. We conclude that signaling through BMP4/Smad1/5/8 promotes the formation of MGPCs, whereas signaling through TGFß/Smad2/3 suppresses the formation of MGPCs.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Ependymoglial Cells/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Retina/cytology , Signal Transduction/physiology , Stem Cells/metabolism , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chickens , Enzyme Inhibitors/pharmacology , Ependymoglial Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , In Situ Nick-End Labeling , N-Methylaspartate/toxicity , RNA, Messenger/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Retina/drug effects , Signal Transduction/drug effects , Smad Proteins/genetics , Smad Proteins/metabolism , Stem Cells/drug effects , Urea/analogs & derivatives , Urea/metabolism
17.
Sci Rep ; 6: 35703, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759082

ABSTRACT

Müller glia are capable of de-differentiating and proliferating to become Müller glia-derived progenitor cells (MGPCs) with the ability to regenerate retinal neurons. One of the cell-signaling pathways that drives the reprogramming of Müller glia into MGPCs in the zebrafish retina is the Jak/Stat-pathway. However, nothing is known about the influence of Jak/Stat-signaling during the formation of MGPCs in the retinas of warm-blooded vertebrates. Accordingly, we examined whether Jak/Stat-signaling influences the formation of MGPCs and differentiation of progeny in the avian retina. We found that Jak/Stat-signaling is activated in Müller glia in response to NMDA-induced retinal damage or by CNTF or FGF2 in the absence of retinal damage. Inhibition of gp130, Jak2, or Stat3 suppressed the formation of proliferating MGPCs in NMDA-damaged and FGF2-treated retinas. Additionally, CNTF combined with FGF2 enhanced the formation of proliferating MGPCs in the absence of retinal damage. In contrast to the zebrafish model, where activation of gp130/Jak/Stat is sufficient to drive neural regeneration from MGPCs, signaling through gp130 inhibits the neurogenic potential of MGPCs and promotes glial differentiation. We conclude that gp130/Jak/Stat-signaling plays an important role in the network of pathways that drives the formation of proliferating MGPCs; however, this pathway inhibits the neural differentiation of the progeny.


Subject(s)
Cell Differentiation , Janus Kinases/metabolism , Neuroglia/physiology , Retina/cytology , STAT Transcription Factors/metabolism , Signal Transduction , Stem Cells/physiology , Animals , Chickens , Regeneration , Retina/injuries
18.
Development ; 143(11): 1859-73, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27068108

ABSTRACT

We investigate the roles of mTor signaling in the formation of Müller glia-derived progenitor cells (MGPCs) in the chick retina. During embryonic development, pS6 (a readout of active mTor signaling) is present in early-stage retinal progenitors, differentiating amacrine and ganglion cells, and late-stage progenitors or maturing Müller glia. By contrast, pS6 is present at low levels in a few scattered cell types in mature, healthy retina. Following retinal damage, in which MGPCs are known to form, mTor signaling is rapidly activated in Müller glia. Inhibition of mTor in damaged retinas prevented the accumulation of pS6 in Müller glia and reduced numbers of proliferating MGPCs. Inhibition of mTor had no effect on MAPK signaling or on upregulation of the stem cell factor Klf4, whereas Pax6 upregulation was significantly reduced. Inhibition of mTor potently blocked the MGPC-promoting effects of Hedgehog, Wnt and glucocorticoid signaling in damaged retinas. In the absence of retinal damage, insulin, IGF1 and FGF2 induced pS6 in Müller glia, and this was blocked by mTor inhibitor. In FGF2-treated retinas, in which MGPCs are known to form, inhibition of mTor blocked the accumulation of pS6, the upregulation of Pax6 and the formation of proliferating MGPCs. We conclude that mTor signaling is required, but not sufficient, to stimulate Müller glia to give rise to proliferating progenitors, and the network of signaling pathways that drive the formation of MGPCs requires activation of mTor.


Subject(s)
Ependymoglial Cells/cytology , Neuroglia/cytology , Retina/metabolism , Signal Transduction , Stem Cells/cytology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Chickens , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , MAP Kinase Signaling System/drug effects , Models, Biological , N-Methylaspartate/pharmacology , Neuroglia/drug effects , Neuroglia/metabolism , PAX6 Transcription Factor/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Retina/pathology , Signal Transduction/drug effects , Sirolimus/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism
19.
Dev Neurobiol ; 76(9): 983-1002, 2016 09.
Article in English | MEDLINE | ID: mdl-26663639

ABSTRACT

Müller glia can be stimulated to de-differentiate, proliferate, and form Müller glia-derived progenitor cells (MGPCs) that are capable of producing retinal neurons. The signaling pathways that influence the de-differentiation of mature Müller glia and proliferation of MGPCs may include the Wnt-pathway. The purpose of this study was to investigate how Wnt-signaling influences the formation of MGPCs in the chick retina in vivo. In NMDA-damaged retinas where MGPCs are known to form, we find dynamic changes in retinal levels of potential readouts of Wnt-signaling, including dkk1, dkk3, axin2, c-myc, tcf-1, and cd44. We find accumulations of nuclear ß-catenin in MGPCs that peaks at 3 days and rapidly declines by 5 days after NMDA-treatment. Inhibition of Wnt-signaling with XAV939 in damaged retinas suppressed the formation of MGPCs, increased expression of ascl1a and decreased hes5, but had no effect upon the differentiation of progeny produced by MGPCs. Activation of Wnt-signaling, with GSK3ß-inhibitors, in the absence of retinal damage, failed to stimulate the formation of MGPCs, whereas activation of Wnt-signaling in damaged retinas stimulated the formation of MGPCs. In the absence of retinal damage, FGF2/MAPK-signaling stimulated the formation of MGPCs by activating a signaling network that includes Wnt/ß-catenin. In FGF2-treated retinas, inhibition of Wnt-signaling reduced numbers of proliferating MGPCs, whereas activation of Wnt-signaling failed to influence the formation of proliferating MGPCs. Our findings indicate that Wnt-signaling is part of a network initiated by FGF2/MAPK or retinal damage, and activation of canonical Wnt-signaling is required for the formation of proliferating MGPCs. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 983-1002, 2016.


Subject(s)
Ependymoglial Cells/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Retina/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Chick Embryo , Chickens , Retina/cytology , Retina/growth & development , Retina/pathology , Wnt Signaling Pathway/physiology
20.
J Comp Neurol ; 524(1): 74-89, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26053997

ABSTRACT

Retinal progenitors in the circumferential marginal zone (CMZ) and Müller glia-derived progenitors have been well described for the eyes of fish, amphibians, and birds. However, there is no information regarding a CMZ and the nature of retinal glia in species phylogenetically bridging amphibians and birds. The purpose of this study was to examine the retinal glia and investigate whether a CMZ is present in the eyes of reptilian species. We used immunohistochemical analyses to study retinal glia, neurons that could influence CMZ progenitors, the retinal margin, and the nonpigmented epithelium of ciliary body of garter snakes, queen snakes, anole lizards, snapping turtles, and painted turtles. We compare our observations on reptile eyes to the CMZ and glia of fish, amphibians, and birds. In all species, Sox9, Pax6, and the glucocorticoid receptor are expressed by Müller glia and cells at the retinal margin. However, proliferating cells were found only in the CMZ of turtles and not in the eyes of anoles and snakes. Similar to eyes of chickens, the retinal margin in turtles contains accumulations of GLP1/glucagonergic neurites. We find that filamentous proteins, vimentin and GFAP, are expressed by Müller glia, but have different patterns of subcellular localization in the different species of reptiles. We provide evidence that the reptile retina may contain nonastrocytic inner retinal glial cells, similar to those described in the avian retina. We conclude that the retinal glia, glucagonergic neurons, and CMZ of turtles appear to be most similar to those of fish, amphibians, and birds.


Subject(s)
Glucagon/metabolism , Lizards/anatomy & histology , Neuroglia/cytology , Retina/cytology , Snakes/anatomy & histology , Turtles/anatomy & histology , Amphibians/anatomy & histology , Amphibians/metabolism , Animals , Cell Proliferation , Chickens/anatomy & histology , Chickens/metabolism , Female , Fishes/anatomy & histology , Fishes/metabolism , Glucagon-Like Peptide 1/metabolism , Lizards/metabolism , Male , Neuroglia/metabolism , Retina/metabolism , Snakes/metabolism , Species Specificity , Turtles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...