Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Adv Exp Med Biol ; 1185: 371-376, 2019.
Article in English | MEDLINE | ID: mdl-31884640

ABSTRACT

This study was designed to assess risk for retinal toxicity associated with administration of high-dose sildenafil citrate to dogs heterozygous for a functionally null mutation in Pde6a over a 4-month period. Three Pde6a +/- dogs were administered 14.3 mg/kg sildenafil per os and two Pde6a +/- dogs placebo once daily for 16 weeks. Three Pde6a +/+ dogs were administered sildenafil for 7 days. Ophthalmic examination, vision testing, and electroretinography (ERG) were regularly performed. At study termination, dogs were euthanized and globes collected. Retinal layer thickness and photoreceptor nuclei counts were determined from plastic sections. In both Pde6a +/- and Pde6a +/+ sildenafil-treated (ST) dogs, elevation of dark-adapted b-wave threshold and unmasking of the scotopic threshold response (STR) were observed. Sildenafil treated Pde6a +/- dogs had significantly thinner ONL (24.90 +/-1.88 µm, p = 0.004) and lower photoreceptor nuclei counts (273.6 +/- 29.3 cells/100 µm, p = 0.008) compared to measurements (35.90 +/- 1.63 µm) and counts (391.5 +/-27.0 cells/100 µm) from archived untreated Pde6a +/- dogs.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Eye Proteins/genetics , Retina/drug effects , Retina/pathology , Sildenafil Citrate/toxicity , Animals , Dogs , Electroretinography , Loss of Function Mutation , Photoreceptor Cells
2.
Mol Vis ; 24: 587-602, 2018.
Article in English | MEDLINE | ID: mdl-30210230

ABSTRACT

Purpose: Retinitis pigmentosa (RP) is a collection of genetic disorders that results in the degeneration of light-sensitive photoreceptor cells, leading to blindness. RP is associated with more than 70 loci that may display dominant or recessive modes of inheritance, but mutations in the gene encoding the visual pigment rhodopsin (RHO) are the most frequent cause. In an effort to develop precise mutations in zebrafish as novel models of photoreceptor degeneration, we describe the generation and germline transmission of a series of novel clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-induced insertion and deletion (indel) mutations in the major zebrafish rho locus, rh1-1. Methods: One- or two-cell staged zebrafish embryos were microinjected with in vitro transcribed mRNA encoding Cas9 and a single guide RNA (gRNA). Mutations were detected by restriction fragment length polymorphism (RFLP) and DNA sequence analyses in injected embryos and offspring. Immunolabeling with rod- and cone-specific antibodies was used to test for histological and cellular changes. Results: Using gRNAs that targeted highly conserved regions of rh1-1, a series of dominant and recessive alleles were recovered that resulted in the rapid degeneration of rod photoreceptors. No effect on cones was observed. Targeting the 5'-coding sequence of rh1-1 led to the recovery of several indels similar to disease-associated alleles. A frame shift mutation leading to a premature stop codon (T17*) resulted in rod degeneration when brought to homozygosity. Immunoblot and fluorescence labeling with a Rho-specific antibody suggest that this is indeed a null allele, illustrating that the Rho expression is essential for rod survival. Two in-frame mutations were recovered that disrupted the highly conserved N-linked glycosylation consensus sequence at N15. Larvae heterozygous for either of the alleles demonstrated rapid rod degeneration. Targeting of the 3'-coding region of rh1-1 resulted in the recovery of an allele encoding a premature stop codon (S347*) upstream of the conserved VSPA sorting sequence and a second in-frame allele that disrupted the putative phosphorylation site at S339. Both alleles resulted in rod death in a dominant inheritance pattern. Following the loss of the targeting sequence, immunolabeling for Rho was no longer restricted to the rod outer segment, but it was also localized to the plasma membrane. Conclusions: The efficiency of CRISPR/Cas9 for gene targeting, coupled with the large number of mutations associated with RP, provided a backdrop for the rapid isolation of novel alleles in zebrafish that phenocopy disease. These novel lines will provide much needed in-vivo models for high throughput screens of compounds or genes that protect from photoreceptor degeneration.


Subject(s)
Disease Models, Animal , Retinal Degeneration/genetics , Retinal Rod Photoreceptor Cells/pathology , Rhodopsin/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Codon, Terminator/genetics , Frameshift Mutation/genetics , Gene Targeting , Immunoblotting , Polymorphism, Restriction Fragment Length , RNA, Messenger/genetics , Retinal Degeneration/pathology
4.
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
5.
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
6.
Exp Neurol ; 273: 114-25, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26272753

ABSTRACT

Reactive microglia and macrophages are prevalent in damaged retinas. Glucocorticoid signaling is known to suppress inflammation and the reactivity of microglia and macrophages. In the vertebrate retina, the glucocorticoid receptor (GCR) is known to be activated and localized to the nuclei of Müller glia (Gallina et al., 2014). Accordingly, we investigated how signaling through GCR influences the survival of neurons using the chick retina in vivo as a model system. We applied intraocular injections of GCR agonist or antagonist, assessed microglial reactivity, and the survival of retinal neurons following different damage paradigms. Microglial reactivity was increased in retinas from eyes that were injected with vehicle, and this reactivity was decreased by GCR-agonist dexamethasone (Dex) and increased by GCR-antagonist RU486. We found that activation of GCR suppresses the reactivity of microglia and inhibited the loss of retinal neurons resulting from excitotoxicity. We provide evidence that the protection-promoting effects of Dex were maintained when the microglia were selectively ablated. Similarly, intraocular injections of Dex protected ganglion cells from colchicine-treatment and protected photoreceptors from damage caused by retinal detachment. We conclude that activation of GCR promotes the survival of ganglion cells in colchicine-damaged retinas, promotes the survival of amacrine and bipolar cells in excitotoxin-damaged retinas, and promotes the survival of photoreceptors in detached retinas. We propose that suppression of microglial reactivity is secondary to activation of GCR in Müller glia, and this mode of signaling is an effective means to lessen the damage and vision loss resulting from different types of retinal damage.


Subject(s)
Ependymoglial Cells/metabolism , Neurons/physiology , Receptors, Glucocorticoid/metabolism , Retina/cytology , Animals , Animals, Newborn , Cell Count , Chickens , Colchicine/pharmacology , Cytokines/metabolism , Ependymoglial Cells/drug effects , Excitatory Amino Acid Agonists/metabolism , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Injections, Intraocular , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Receptors, Glucocorticoid/genetics , Retinal Detachment/chemically induced , Retinal Detachment/metabolism , Retinal Detachment/pathology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Tubulin Modulators/pharmacology , Visual Pathways/drug effects , Visual Pathways/metabolism , Zebrafish Proteins
7.
Glia ; 63(2): 313-27, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25231952

ABSTRACT

Reactive microglia and macrophages are prevalent in damaged retinas. Accordingly, we investigate how the activation or ablation of microglia/macrophages influences the survival of neurons in the chick retina in vivo. We applied intraocular injections of interleukin 6 (IL6) to stimulate the reactivity of microglia/macrophages and clodronate-liposomes to ablate microglia/macrophages. Activation of the microglia/macrophages with IL6 delays the death of retinal neurons from N-methyl-D-aspartate (NMDA) -induced excitotoxicity. In addition, activation of microglia/macrophages combined with colchicine-mediated retinal damage diminished the survival of ganglion cells. Application of IL6 after an excitotoxic insult greatly exacerbates the damage, and causes widespread retinal detachments and folds, accompanied by accumulation of microglia/macrophages in the subretinal space. Damage-induced retinal folds and detachments were significantly reduced by the ablation of microglia/macrophages. We conclude that microglial reactivity is detrimental to the survival of ganglion cells in colchicine-damaged retinas and detrimental to the survival of photoreceptors in retinal folds. In addition, we conclude that IL6-treatment transiently protects amacrine and bipolar cells against an excitotoxic insult. We propose that suppressing reactivity of microglia/macrophages may be an effective means to lessen the damage and vision loss resulting from damage, in particular during retinal detachment injuries.


Subject(s)
Microglia/physiology , Neurons/physiology , Retina/physiology , Retinal Detachment/pathology , Age Factors , Animals , Animals, Newborn , Cell Death/drug effects , Chickens , Clodronic Acid/administration & dosage , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , In Situ Nick-End Labeling , Interleukin-6/therapeutic use , Leukocyte Common Antigens , Liposomes/metabolism , Macrophages/drug effects , Microglia/drug effects , Microglia/pathology , N-Methylaspartate/toxicity , Neurons/drug effects , Plant Lectins , Retina/drug effects , Retina/pathology , Retinal Detachment/chemically induced , Retinal Detachment/drug therapy , SOX9 Transcription Factor
8.
Development ; 141(17): 3340-51, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25085975

ABSTRACT

Identification of the signaling pathways that influence the reprogramming of Müller glia into neurogenic retinal progenitors is key to harnessing the potential of these cells to regenerate the retina. Glucocorticoid receptor (GCR) signaling is commonly associated with anti-inflammatory responses and GCR agonists are widely used to treat inflammatory diseases of the eye, even though the cellular targets and mechanisms of action in the retina are not well understood. We find that signaling through GCR has a significant impact upon the ability of Müller glia to become proliferating Müller glia-derived progenitor cells (MGPCs). The primary amino acid sequence and pattern of GCR expression in the retina is highly conserved across vertebrate species, including chickens, mice, guinea pigs, dogs and humans. In all of these species we find GCR expressed by the Müller glia. In the chick retina, we find that GCR is expressed by progenitors in the circumferential marginal zone (CMZ) and is upregulated by Müller glia in acutely damaged retinas. Activation of GCR signaling inhibits the formation of MGPCs and antagonizes FGF2/MAPK signaling in the Müller glia. By contrast, we find that inhibition of GCR signaling stimulates the formation of proliferating MGPCs in damaged retinas, and enhances the neuronal differentiation while diminishing glial differentiation. Given the conserved expression pattern of GCR in different vertebrate retinas, we propose that the functions and mechanisms of GCR signaling are highly conserved and are mediated through the Müller glia. We conclude that GCR signaling directly inhibits the formation of MGPCs, at least in part, by interfering with FGF2/MAPK signaling.


Subject(s)
Ependymoglial Cells/metabolism , Receptors, Glucocorticoid/metabolism , Retina/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Proliferation/drug effects , Dexamethasone/pharmacology , Ependymoglial Cells/drug effects , Ependymoglial Cells/enzymology , Ependymoglial Cells/pathology , Fibroblast Growth Factor 2/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , N-Methylaspartate/pharmacology , Retina/drug effects , Retina/pathology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Signal Transduction/drug effects , Stem Cells/drug effects , TOR Serine-Threonine Kinases/metabolism
9.
Glia ; 62(10): 1608-28, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24916856

ABSTRACT

In retinas where Müller glia have been stimulated to become progenitor cells, reactive microglia are always present. Thus, we investigated how the activation or ablation of microglia/macrophage influences the formation of Müller glia-derived progenitor cells (MGPCs) in the retina in vivo. Intraocular injections of the Interleukin-6 (IL6) stimulated the reactivity of microglia/macrophage, whereas other types of retinal glia appear largely unaffected. In acutely damaged retinas where all of the retinal microglia/macrophage were ablated, the formation of proliferating MGPCs was greatly diminished. With the microglia ablated in damaged retinas, levels of Notch and related genes were unchanged or increased, whereas levels of ascl1a, TNFα, IL1ß, complement component 3 (C3) and C3a receptor were significantly reduced. In the absence of retinal damage, the combination of insulin and Fibroblast growth factor 2 (FGF2) failed to stimulate the formation of MGPCs when the microglia/macrophage were ablated. In addition, intraocular injections of IL6 and FGF2 stimulated the formation of MGPCs in the absence of retinal damage, and this generation of MGPCs was blocked when the microglia/macrophage were absent. We conclude that the activation of microglia and/or infiltrating macrophage contributes to the formation of proliferating MGPCs, and these effects may be mediated by components of the complement system and inflammatory cytokines.


Subject(s)
Ependymoglial Cells/physiology , Macrophages/physiology , Microglia/physiology , Neural Stem Cells/physiology , Animals , Avian Proteins/metabolism , Cell Proliferation/physiology , Chickens , Complement C3/metabolism , Excitatory Amino Acid Agonists/toxicity , Fibroblast Growth Factor 2/metabolism , Insulin/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , N-Methylaspartate/toxicity , Receptors, Complement/metabolism , Receptors, Notch/metabolism , Retina/injuries , Retina/physiopathology , Tumor Necrosis Factor-alpha/metabolism
10.
PLoS One ; 7(9): e44477, 2012.
Article in English | MEDLINE | ID: mdl-22973454

ABSTRACT

Recent studies have described a novel type of glial cell that is scattered across the inner layers of the avian retina and possibly the retinas of primates. These cells have been termed Non-astrocytic Inner Retinal Glial (NIRG) cells. These cells are stimulated by insulin-like growth factor 1 (IGF1) to proliferate, migrate distally into the retina, and become reactive. These changes in glial activity correlate with increased susceptibility of retinal neurons and Müller glia to excitotoxic damage. The purpose of this study was to further study the NIRG cells in retinas treated with IGF1 or acute damage. In response to IGF1, the reactivity, proliferation and migration of NIRG cells persists through 3 days after treatment. At 7 days after treatment, the numbers and distribution of NIRG cells returns to normal, suggesting that homeostatic mechanisms are in place within the retina to maintain the numbers and distribution of these glial cells. By comparison, IGF1-induced microglial reactivity persists for at least 7 days after treatment. In damaged retinas, we find a transient accumulation of NIRG cells, which parallels the accumulation of reactive microglia, suggesting that the reactivity of NIRG cells and microglia are linked. When the microglia are selectively ablated by the combination of interleukin 6 and clodronate-liposomes, the NIRG cells down-regulate transitin and perish within the following week, suggesting that the survival and phenotype of NIRG cells are somehow linked to the microglia. We conclude that the abundance, reactivity and retinal distribution of NIRG cells can be dynamic, are regulated by homoestatic mechanisms and are tethered to the microglia.


Subject(s)
Gene Expression Regulation/drug effects , Homeostasis/physiology , Insulin-Like Growth Factor I/pharmacology , Microglia/drug effects , Neuroglia/metabolism , Retina/cytology , Animals , Bromodeoxyuridine , Cell Count , Cell Movement/drug effects , Cell Proliferation/drug effects , Chickens , Clodronic Acid/administration & dosage , Clodronic Acid/toxicity , Colchicine/toxicity , DNA Primers/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Immunohistochemistry , Injections, Intraocular , Insulin-Like Growth Factor I/administration & dosage , Interleukin-6/administration & dosage , Interleukin-6/toxicity , Intermediate Filament Proteins/metabolism , Liposomes/administration & dosage , Liposomes/toxicity , Microglia/physiology , Microscopy, Fluorescence , N-Methylaspartate/toxicity , Nerve Tissue Proteins/metabolism , Nestin , Neuroglia/drug effects , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Zebrafish Proteins
11.
Exp Eye Res ; 102: 59-69, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22824538

ABSTRACT

Visual experience is known to guide ocular growth. We tested the hypothesis that vision-guided ocular growth is disrupted in a model system with diminished visual acuity. We examine whether ocular elongation is influenced by form-deprivation (FD) and lens-imposed defocus in the Retinopathy, Globe Enlarged (RGE) chicken. Young RGE chicks have poor visual acuity, without significant retinal pathology, resulting from a mutation in guanine nucleotide-binding protein ß3 (GNB3), also known as transducin ß3 or Gß3. The mutation in GNB3 destabilizes the protein and causes a loss of Gß3 from photoreceptors and ON-bipolar cells (Ritchey et al., 2010). FD increased ocular elongation in RGE eyes in a manner similar to that seen in wild-type (WT) eyes. By comparison, the excessive ocular elongation that results from hyperopic defocus was increased, whereas myopic defocus failed to significantly decrease ocular elongation in RGE eyes. Brief daily periods of unrestricted vision interrupting FD prevented ocular elongation in RGE chicks in a manner similar to that seen in WT chicks. Glucagonergic amacrine cells differentially expressed the immediate early gene Egr1 in response to growth-guiding stimuli in RGE retinas, but the defocus-dependent up-regulation of Egr1 was lesser in RGE retinas compared to that of WT retinas. We conclude that high visual acuity, and the retinal signaling mediated by Gß3, is not required for emmetropization and the excessive ocular elongation caused by FD and hyperopic defocus. However, the loss of acuity and Gß3 from RGE retinas causes enhanced responses to hyperopic defocus and diminished responses to myopic defocus.


Subject(s)
Chickens/genetics , Disease Models, Animal , Eye/growth & development , Myopia/physiopathology , Vision Disorders/physiopathology , Vision, Ocular/physiology , Visual Acuity/physiology , Amacrine Cells/metabolism , Animals , Axial Length, Eye , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Eye/diagnostic imaging , Fluorescent Antibody Technique, Indirect , Glucagon/genetics , Glucagon/metabolism , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Microscopy, Confocal , Myopia/genetics , Myopia/metabolism , RNA, Messenger/metabolism , Refraction, Ocular/physiology , Retinoscopy , Reverse Transcriptase Polymerase Chain Reaction , Sensory Deprivation , Ultrasonography , Vision Disorders/genetics , Vision Disorders/metabolism
12.
Exp Eye Res ; 99: 1-16, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22695224

ABSTRACT

Different growth factors have been shown to influence the development of form-deprivation myopia and lens-induced ametropias. However, growth factors have relatively little effect on the growth of eyes with unrestricted vision. We investigate whether the combination of insulin-like growth factor 1 (IGF1) and fibroblast growth factor 2 (FGF2) influence ocular growth in eyes with unrestricted vision. Different doses of IGF1 and FGF2 were injected into the vitreous chamber of postnatal chicks. Measurements of ocular dimensions and intraocular pressure (IOP) were made during and at the completion of different treatment paradigms. Histological and immunocytochemical analyses were performed to assess cell death, cellular proliferation and integrity of ocular tissues. Treated eyes had significant increases in equatorial diameter and vitreous chamber depth. With significant variability between individuals, IGF1/FGF2-treatment caused hypertrophy of lens and ciliary epithelia, lens thickness was increased, and anterior chamber depth was decreased. Treated eyes developed myopia, in excess of 15 diopters of refractive error. Shortly after treatment, eyes had increased intraocular pressure (IOP), which was increased in a dose-dependent manner. Seven days after treatment with IGF1 and FGF2 changes to anterior chamber depth, lens thickness and elevated IOP were reduced, whereas increases in the vitreous chamber were persistent. Some damage to ganglion cells was detected in peripheral regions of the retina at 7 days after treatment. We conclude that the extreme myopia in IGF1/FGF2-treated eyes results from increased vitreous chamber depth, decreased anterior chamber depth, and changes in the lens. We propose that factor-induced ocular enlargement and myopia result from changes to the sclera, lens and anterior chamber depth.


Subject(s)
Disease Models, Animal , Eye/drug effects , Fibroblast Growth Factor 2/toxicity , Insulin-Like Growth Factor I/toxicity , Myopia, Degenerative/chemically induced , Animals , Animals, Newborn , Anterior Chamber/drug effects , Anterior Chamber/pathology , Apoptosis/drug effects , Cell Proliferation/drug effects , Chickens , Ciliary Body/drug effects , Ciliary Body/pathology , Dose-Response Relationship, Drug , Drug Combinations , Eye/growth & development , Hypertrophy , In Situ Nick-End Labeling , Intraocular Pressure/drug effects , Intravitreal Injections , Lens, Crystalline/drug effects , Lens, Crystalline/pathology , Myopia, Degenerative/pathology , Organ Size/drug effects , RNA, Messenger/metabolism , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Receptors, Fibroblast Growth Factor/genetics , Retina/drug effects , Retina/pathology , Retinoscopy , Reverse Transcriptase Polymerase Chain Reaction
13.
Mol Vis ; 17: 2440-54, 2011.
Article in English | MEDLINE | ID: mdl-21976955

ABSTRACT

PURPOSE: The cornea is the major refractive component of the eye and serves as a barrier to the external environment. Understanding how the cornea responds to injury is important to developing therapies to treat vision disorders that affect the integrity and refractive properties of the cornea. Thus, investigation of the wound healing responses of the cornea to injury in a cost-effective animal model is a valuable tool for research. This study characterizes the wound healing responses in the corneas of White Leghorn chicken. METHODS: Linear corneal wounds were induced in post-natal day 7 (P7) chicks and cellular proliferation, apoptosis and regulation of structural proteins were assessed using immunohistochemical techniques. We describe the time course of increased expression of different scar-related markers, including vimentin, vinculin, perlecan and smooth muscle actin. RESULTS: We find evidence for acute necrotic cell death in the corneal region immediately surrounding cite of incision, whereas we failed to find evidence of delayed cell death or apoptosis. We find that the neuronal re-innervation of SV2-positive axon terminals within the corneal stroma and epithelium occurs very quickly after the initial scarring insult. We describe an accumulation of cells within the stroma immediately underlying the scar, which results, at least in part, from the local proliferation of keratocytes. Further, we provide evidence for scar-induced accumulations of CD45-positive monocytes in injured corneas. CONCLUSIONS: We conclude that the chick cornea is an excellent model system in which to study wound healing, formation of scar tissue, and neuronal re-innervation of sensory endings.


Subject(s)
Biomarkers/analysis , Cicatrix/metabolism , Cornea/metabolism , Cornea/pathology , Corneal Keratocytes/metabolism , Neurons/metabolism , Wound Healing/physiology , Actins/analysis , Actins/biosynthesis , Animals , Animals, Newborn , Bromodeoxyuridine/analysis , Cell Proliferation , Chickens , Cornea/innervation , Corneal Injuries , Corneal Keratocytes/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/biosynthesis , Immunohistochemistry , Leukocyte Common Antigens/analysis , Microscopy , Monocytes/cytology , Monocytes/metabolism , Necrosis , Neurons/cytology , Vimentin/analysis , Vimentin/biosynthesis , Vinculin/analysis , Vinculin/biosynthesis
14.
PLoS One ; 5(6): e10774, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20567503

ABSTRACT

We have recently described a novel type of glial cell that is scattered across the inner layers of the avian retina [1]. These cells are stimulated by insulin-like growth factor 1 (IGF1) to proliferate, migrate distally into the retina, and up-regulate the nestin-related intermediate filament transition. These changes in glial activity correspond with increased susceptibility of neurons to excitotoxic damage. This novel cell-type has been termed the Non-astrocytic Inner Retinal Glia-like (NIRG) cells. The purpose of the study was to investigate whether the retinas of non-avian species contain cells that resemble NIRG cells. We assayed for NIRG cells by probing for the expression of Sox2, Sox9, Nkx2.2, vimentin and nestin. NIRG cells were distinguished from astrocytes by a lack of expression for Glial Fibrilliary Acidic Protein (GFAP). We examined the retinas of adult mice, guinea pigs, dogs and monkeys (Macaca fasicularis). In the mouse retina and optic nerve head, we identified numerous astrocytes that expressed GFAP, S100beta, Sox2 and Sox9; however, we found no evidence for NIRG-like cells that were positive for Nkx2.2, nestin, and negative for GFAP. In the guinea pig retina, we did not find astrocytes or NIRG cells in the retina, whereas we identified astrocytes in the optic nerve. In the eyes of dogs and monkeys, we found astrocytes and NIRG-like cells scattered across inner layers of the retina and within the optic nerve. We conclude that NIRG-like cells are present in the retinas of canines and non-human primates, whereas the retinas of mice and guinea pigs do not contain NIRG cells.


Subject(s)
Neuroglia/cytology , Optic Nerve/cytology , Retina/cytology , Animals , Birds , Gene Expression Profiling , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Mammals , Neuroglia/metabolism , Nuclear Proteins , Optic Nerve/metabolism , Retina/metabolism , Species Specificity , Transcription Factors
15.
J Neurosci ; 30(8): 3101-12, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181607

ABSTRACT

Notch signaling is known to play important roles during retinal development. Recently, Notch signaling has been shown to be active in proliferating Müller glia in acutely damaged chick retina (Hayes et al., 2007). However, the roles of Notch in mature, undamaged retina remain unknown. Thus, the purpose of this study was to determine the role of the Notch-signaling pathway in the postnatal retina. Here we show that components of the Notch-signaling pathway are expressed in most Müller glia at low levels in undamaged retina. The expression of Notch-related genes varies during early postnatal development and across regions, with higher expression in peripheral versus central retina. Blockade of Notch activity with a small molecule inhibitor before damage was protective to retinal interneurons (amacrine and bipolar cells) and projection neurons (ganglion cells). In the absence of damage, Notch is upregulated in retinas treated with insulin and FGF2; the combination of these factors is known to stimulate the proliferation and dedifferentiation of Müller glia (Fischer et al., 2002b). Inhibition of Notch signaling during FGF2 treatment reduces levels of the downstream effectors of the MAPK-signaling pathway-p38 MAPK and pCREB in Müller glia. Further, inhibition of Notch activity potently inhibits FGF2-induced proliferation of Müller glia. Together, our data indicate that Notch signaling is downstream of, and is required for, FGF2/MAPK signaling to drive the proliferation of Müller glia. In addition, our data suggest that low levels of Notch signaling in Müller glia diminish the neuroprotective activities of these glial cells.


Subject(s)
Cell Proliferation/drug effects , Cytoprotection/physiology , Neuroglia/metabolism , Receptors, Notch/metabolism , Retina/metabolism , Signal Transduction/physiology , Amacrine Cells/drug effects , Amacrine Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival/physiology , Chickens , Cyclic AMP Response Element-Binding Protein/metabolism , Cytoprotection/drug effects , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Neuroglia/cytology , Neuroglia/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retina/cytology , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Signal Transduction/drug effects , Transcription Factor HES-1 , Triglycerides/pharmacology , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/pharmacology
16.
Glia ; 58(6): 633-49, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-19941335

ABSTRACT

Recent studies have demonstrated that insulin can have profound affects on the survival of neurons within the retina. The purpose of this study was to determine how insulin-like growth factor 1 (IGF1) influences retinal cells; in particular, the glial cells. We identify a novel type of glial cell in the avian retina and provide evidence that these cells can respond to acute damage and IGF1. In normal retinas, we found a distinct cell-type, scattered across the ganglion cell and inner plexiform layers that express Sox2, Sox9, Nkx2.2, vimentin, and transitin, the avian homologue of mammalian nestin. These glial cells have a unique immunohistochemical profile, morphology, and distribution that are distinct among other known types of retinal glia, including microglia, oligodendrocytes, astrocytes, and Muller glia. We termed these cells nonastrocytic inner retinal glia-like (NIRG) cells. We found that the NIRG cells may express the IGF1 receptor and respond to IGF1 by proliferating, migrating distally into the retina, and upregulating transitin. In addition, IGF1 stimulated microglia to become reactive and upregulate lysosomal membrane glycoprotein and CD45. With microglia and NIRG cells stimulated by IGF1 there were elevated levels of cell death and numerous focal detachments across the retina in response to excitotoxic damage. Cell death was prominent within the areas of detachment coinciding with a stark loss of Müller glia and accumulation of NIRG cells. We conclude that NIRG cells are a novel type of retinal glia that is sensitive to IGF1 and whose activity may impact the survival of neurons and Müller glia.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Neuroglia/drug effects , Neurons/drug effects , Retina/cytology , Animals , Animals, Newborn , Cell Count/methods , Cell Proliferation/drug effects , Chickens , Drug Synergism , Excitatory Amino Acid Agonists/toxicity , Glial Fibrillary Acidic Protein/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , In Situ Nick-End Labeling/methods , Injections, Intraocular/methods , Leukocyte Common Antigens/metabolism , N-Methylaspartate/toxicity , Plant Lectins/metabolism , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/metabolism , SOX Transcription Factors/metabolism , Transcription Factors/metabolism , Vimentin/metabolism , Zebrafish Proteins
17.
J Neurochem ; 111(1): 1-14, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19619137

ABSTRACT

The neurotransmitter serotonin is synthesized in the retina by one type of amacrine neuron but accumulates in bipolar neurons in many vertebrates. The mechanisms, functions and purpose underlying serotonin accumulation in bipolar cells remain unknown. Here, we demonstrate that exogenous serotonin transiently accumulates in a distinct type of bipolar neuron. KCl-mediated depolarization causes the depletion of serotonin from amacrine neurons and, subsequently, serotonin is taken-up by bipolar neurons. The accumulation of endogenous and exogenous serotonin by bipolar neurons is blocked by selective reuptake inhibitors. Exogenous serotonin is specifically taken-up by bipolar neurons even when serotonin-synthesizing amacrine neurons are destroyed; excluding the possibility that serotonin diffuses through gap junctions from amacrine into bipolar neurons. Further, inhibition of monoamine oxidase A prevents the degradation of serotonin in bipolar neurons, suggesting that monoamine oxidase A is present in these neurons. However, the vesicular monoamine transporter 2 is present only in amacrine cells suggesting that serotonin is not transported into synaptic vesicles and reused as a transmitter in the bipolar neurons. We conclude that the serotonin-accumulating bipolar neurons perform glial functions in the retina by actively transporting and degrading serotonin that is synthesized in neighboring amacrine cells.


Subject(s)
Amacrine Cells/metabolism , Retina/cytology , Retinal Bipolar Cells/metabolism , Serotonin/metabolism , Amacrine Cells/drug effects , Animals , Animals, Newborn , Chickens , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Homeodomain Proteins/metabolism , Potassium Chloride/pharmacology , Retinal Bipolar Cells/drug effects , Serotonin/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Sertraline/pharmacology , Vesicular Monoamine Transport Proteins/genetics , Vesicular Monoamine Transport Proteins/metabolism , Zimeldine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...