Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 632: 76-84, 2022 12 03.
Article in English | MEDLINE | ID: mdl-36206597

ABSTRACT

Hairy and enhancer of split homolog-1 (Hes1) is a member of an extensive family of basic helix-loop-helix (bHLH) proteins and plays a crucial role in neurogenesis, myogenesis, hematopoiesis, and sex determination. It has been reported that Hes1 is essential for precursors maintenance, optic cup-stalk boundary maintenance, and morphogenesis of the retina. However, it still reminds questions about the role and mechanism of Hes1 in the development of retinal pigment epithelial cells. In our study, We generated Hes1-/- human embrsyonic stem cells, and attempted to induce them into retinal pigment epithelial cells by our previous protocol, found that the cells induced by Hes1-/- hESCs hardly expressed RPE-related genes, and rarely appeared RPE cell morphology. Additionally, Hes1 may affect the development of RPE cells via Wnt4 pathway by analyzing the RNA-seq data of differently expressed genes between normal RPE cells development and Hes1-/- RPE cells development. Overall, depletion of Hes1 may result in the failure of Wnt4 signal activation, and contributed to the developmental disorder in retinal pigment epithelium morphogenesis and specification.


Subject(s)
Morphogenesis , Retinal Pigment Epithelium , Transcription Factor HES-1 , Child , Humans , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryonic Stem Cells , Morphogenesis/genetics , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism , Retinal Pigments/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
2.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34006636

ABSTRACT

Loss of the retinal pigment epithelium (RPE) because of dysfunction or disease can lead to blindness in humans. Harnessing the intrinsic ability of the RPE to self-repair is an attractive therapeutic strategy; however, mammalian RPE is limited in its regenerative capacity. Zebrafish possess tremendous intrinsic regenerative potential in ocular tissues, including the RPE, but little is known about the mechanisms driving RPE regeneration. Here, utilizing transgenic and mutant zebrafish lines, pharmacological manipulations, transcriptomics, and imaging analyses, we identified elements of the immune response as critical mediators of intrinsic RPE regeneration. After genetic ablation, the RPE express immune-related genes, including leukocyte recruitment factors such as interleukin 34 We demonstrate that macrophage/microglia cells are responsive to RPE damage and that their function is required for the timely progression of the regenerative response. These data identify the molecular and cellular underpinnings of RPE regeneration and hold significant potential for translational approaches aimed toward promoting a pro-regenerative environment in mammalian RPE.


Subject(s)
Blindness/genetics , Immunity/genetics , Interleukins/genetics , Regeneration/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Blindness/parasitology , Blindness/therapy , Disease Models, Animal , Gene Expression Regulation, Developmental/genetics , Humans , Microglia/metabolism , Microglia/pathology , Mutation/genetics , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/pathology , Transcriptome/genetics , Zebrafish/genetics , Zebrafish/growth & development
3.
Int J Mol Sci ; 22(8)2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33919990

ABSTRACT

Age-related macular degeneration (AMD) leads to gradual central vision loss and is the third leading cause of irreversible blindness worldwide. The underlying mechanisms for this progressive neurodegenerative disease remain unclear and there is currently no preventive treatment for dry AMD. Sodium iodate (NaIO3) has been reported to induce AMD-like retinal pathology in mice. We established a mouse model for AMD to evaluate the effects of quercetin on NaIO3-induced retinal apoptosis, and to investigate the pertinent underlying mechanisms. Our in vitro results indicated that quercetin protected human retinal pigment epithelium (ARPE-19) cells from NaIO3-induced apoptosis by inhibiting reactive oxygen species production and loss of mitochondrial membrane potential as detected by Annexin V-FITC/PI flow cytometry. We also evaluated the relative expression of proteins in the apoptosis pathway. Quercetin downregulated the protein expressions of Bax, cleaved caspase-3, and cleaved PARP and upregulated the expression of Bcl-2 through reduced PI3K and pAKT expressions. Furthermore, our in vivo results indicated that quercetin improved retinal deformation and increased the thickness of both the outer nuclear layer and inner nuclear layer, whereas the expression of caspase-3 was inhibited. Taken together, these results demonstrate that quercetin could protect retinal pigment epithelium and the retina from NaIO3-induced cell apoptosis via reactive oxygen species-mediated mitochondrial dysfunction, involving the PI3K/AKT signaling pathway. This suggests that quercetin has the potential to prevent and delay AMD and other retinal diseases involving NaIO3-mediated apoptosis.


Subject(s)
Macular Degeneration/drug therapy , Quercetin/pharmacology , Retina/drug effects , Retinal Diseases/drug therapy , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/genetics , Cell Line , Gene Expression Regulation/drug effects , Humans , Iodates/toxicity , Macular Degeneration/genetics , Macular Degeneration/pathology , Mitochondria/drug effects , Poly(ADP-ribose) Polymerases/genetics , Reactive Oxygen Species/metabolism , Retina/pathology , Retinal Diseases/chemically induced , Retinal Diseases/genetics , Retinal Diseases/pathology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/growth & development , Signal Transduction/drug effects , bcl-2-Associated X Protein/genetics
4.
Mol Med Rep ; 23(5)2021 05.
Article in English | MEDLINE | ID: mdl-33760200

ABSTRACT

Proliferative vitreoretinopathy (PVR) is a disease leading to the formation of contractile preretinal membranes (PRMs) and is one of the leading causes of blindness. Connective tissue growth factor (CTGF) has been identified as a possible key determinant of progressive tissue fibrosis and excessive scarring. Therefore, the present study investigated the role and mechanism of action of CTGF in PVR. Immunohistochemical staining was performed to detect the expression of CTGF, fibronectin and collagen type III in PRMs from patients with PVR. The effects and mechanisms of recombinant human CTGF and its upstream regulator, TGF­ß1, on epithelial­mesenchymal transition (EMT) and the synthesis of extracellular matrix (ECM) by retinal pigment epithelium (RPE) cells were investigated using reverse transcription­quantitative PCR, western blotting and a [3H]proline incorporation assay. The data indicated that CTGF, fibronectin and collagen type III were highly expressed in PRMs. In vitro, CTGF significantly decreased the expression of the epithelial markers ZO­1 and E­cadherin and increased that of the mesenchymal markers fibronectin, N­cadherin and α­smooth muscle actin in a concentration­dependent manner. Furthermore, the expression of the ECM protein collagen type III was upregulated by CTGF. However, the trends in expression for the above­mentioned markers were reversed after knocking down CTGF. The incorporation of [3H]proline into RPE cells was also increased by CTGF. In addition, 8­Bromoadenosine cAMP inhibited CTGF­stimulated collagen synthesis and transient transfection of RPE cells with a CTGF antisense oligonucleotide inhibited TGF­ß1­induced collagen synthesis. The phosphorylation of PI3K and AKT in RPE cells was promoted by CTGF and TGF­ß1 and the latter promoted the expression of CTGF. The results of the present study indicated that CTGF may promote EMT and ECM synthesis in PVR via the PI3K/AKT signaling pathway and suggested that targeting CTGF signaling may have a therapeutic or preventative effect on PVR.


Subject(s)
Connective Tissue Growth Factor/genetics , Epithelial-Mesenchymal Transition/genetics , Retinal Pigments/genetics , Transforming Growth Factor beta1/genetics , Vitreoretinopathy, Proliferative/genetics , Blotting, Western , Cell Movement/genetics , Extracellular Matrix/genetics , Fibronectins/genetics , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism , Signal Transduction/genetics , Vitreoretinopathy, Proliferative/pathology
5.
Curr Stem Cell Res Ther ; 16(6): 710-717, 2021.
Article in English | MEDLINE | ID: mdl-33511959

ABSTRACT

Retinal degenerative diseases (RDDs) are irreversible ocular damages categorized as retinopathies. RDDs affect about 0.05% of individuals worldwide. The degenerations of RPE cells are involved in inherited and age-related RDDs. After the invention of induced pluripotent stem cells (iPSC) by Yamanaka, a promising avenue has been opened to regenerative medicine and disease modeling. Retinal pigment epithelium (RPE) degeneration related-RDDs are also affected by iPSCs. IPSC-derived RPE cells created a novel method for treating the RPE degeneration related- RDDs and retinal diseases modeling to find a new therapeutic approach or drug development. There are various studies based on iPSC-derived RPE cells reporting the investigation of the role of a specific mutation, protein, signaling pathway, etc., responsible for a type of RDD. Furthermore, iPSC-based RPE therapy is expanded to include some clinical trials. Despite the incredible growth rate in iPSC-based studies on RPE-related diseases, there are some challenges, i.e., teratoma formation potential of iPSCs, an expensive procedure of iPSC-based regeneration of RPEs, lack of a universal protocol or cellular product applicable in all patients, etc. This article reviews the iPSC-based RPE generation and their therapeutic applications, studies on RPE-related molecular and cellular pathophysiologic features of RDD in the iPSC-based models, future perspectives, and the challenges ahead.


Subject(s)
Induced Pluripotent Stem Cells , Regeneration , Retinal Degeneration , Retinal Pigment Epithelium/growth & development , Humans , Induced Pluripotent Stem Cells/cytology , Regenerative Medicine , Retinal Degeneration/therapy
6.
J Vis Exp ; (165)2020 11 25.
Article in English | MEDLINE | ID: mdl-33311434

ABSTRACT

In ophthalmic research, there is a strong need for in vitro models of the neuroretina. Here, we present a detailed protocol for organotypic culturing of the mouse neuroretina with intact retinal pigment epithelium (RPE). Depending on the research question, retinas can be isolated from wild-type animals or from disease models, to study, for instance, diabetic retinopathy or hereditary retinal degeneration. Eyes from early postnatal day 2-9 animals are enucleated under aseptic conditions. They are partially digested in proteinase K to allow for a detachment of the choroid from the RPE. Under the stereoscope, a small incision is made in the cornea creating two edges from where the choroid and sclera can be gently peeled off from the RPE and neuroretina. The lens is then removed, and the eyecup is cut in four points to give it a four-wedged shape resembling a clover leaf. The tissue is finally transferred in a hanging drop into a cell culture insert holding a polycarbonate culturing membrane. The cultures are then maintained in R16 medium, without serum or antibiotics, under entirely defined conditions, with a medium change every second day. The procedure described enables the isolation of the retina and the preservation of its normal physiological and histotypic context for culturing periods of at least 2 weeks. These features make organotypic retinal explant cultures an excellent model with high predictive value, for studies into retinal development, disease mechanisms, and electrophysiology, while also enabling pharmacological screening.


Subject(s)
Organ Culture Techniques/methods , Retinal Pigment Epithelium/growth & development , Animals , Cell Culture Techniques , Culture Media, Serum-Free , Mice , Retinal Pigment Epithelium/cytology
7.
J Comp Neurol ; 528(17): 2864-2873, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32452548

ABSTRACT

We previously reported bidirectional gene expression regulation of the Bone Morphogenetic Proteins (BMP2, 4, and 7) in chick retinal pigment epithelium (RPE) in response to imposed optical defocus and form-deprivation (FD). This study investigated whether there are local (regional) differences in these effects. 19-day old White-Leghorn chicks wore monocular +10 or - 10 D lenses, or diffusers (FD) for 2 or 48 hr, after which RPE samples were collected from both eyes, from a central circular zone (3 mm radius), and 3 mm wide annular mid-peripheral and peripheral zones in all cases. BMP2, 4, and 7 gene expression levels in RPE from treated and fellow control eyes were compared as well as differences across zones. With the +10 D lens, increased expression of both BMP2 and BMP4 genes was observed in central and mid-peripheral zones but not the peripheral zone after 2 and 48 hr. In contrast, with the -10 D lens BMP2 gene expression was significantly decreased in all three zones after 2 and 48 hr. Similar patterns of BMP2 gene expression were observed in all three zones after 48 hr of FD. Smaller changes were recorded for BMP4 and BMP7 gene expression for both myopia-inducing treatments. That optical defocus- and FD-induced changes in BMP gene expression in chick RPE show treatment-dependent local (regional) differences suggest important differences in the nature and contributions of local retinal and underlying RPE regions to eye growth regulation.


Subject(s)
Bone Morphogenetic Proteins/biosynthesis , Form Perception/physiology , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Chickens , Gene Expression Regulation/physiology , Retina/metabolism , Reverse Transcription/physiology
8.
J Exp Zool B Mol Dev Evol ; 334(7-8): 438-449, 2020 11.
Article in English | MEDLINE | ID: mdl-31930686

ABSTRACT

Astyanax mexicanus consists of two forms, a sighted surface dwelling form (surface fish) and a blind cave-dwelling form (cavefish). Embryonic eyes are initially formed in cavefish but they are subsequently arrested in growth and degenerate during larval development. Previous lens transplantation studies have shown that the lens plays a central role in cavefish eye loss. However, several lines of evidence suggest that additional factors, such as the retinal pigment epithelium (RPE), which is morphologically altered in cavefish, could also be involved in the eye regression process. To explore the role of the RPE in cavefish eye degeneration, we generated an albino eyed (AE) strain by artificial selection for hybrid individuals with large eyes and a depigmented RPE. The AE strain exhibited an RPE lacking pigment granules and showed reduced expression of the RPE specific enzyme retinol isomerase, allowing eye development to be studied by lens ablation in an RPE background resembling cavefish. We found that lens ablation in the AE strain had stronger negative effects on eye growth than in surface fish, suggesting that an intact RPE is required for normal eye development. We also found that the AE strain develops a cartilaginous sclera lacking boney ossicles, a trait similar to cavefish. Extrapolation of the results to cavefish suggests that the RPE and lens have dual roles in eye degeneration, and that deficiencies in the RPE may be associated with evolutionary changes in scleral ossification.


Subject(s)
Characidae/embryology , Eye/embryology , Lens, Crystalline/embryology , Retinal Pigment Epithelium/embryology , Animals , Caves , Characidae/anatomy & histology , Characidae/growth & development , Eye/growth & development , Eye Abnormalities/embryology , Female , Lens, Crystalline/growth & development , Male , Retinal Pigment Epithelium/anatomy & histology , Retinal Pigment Epithelium/growth & development
9.
Graefes Arch Clin Exp Ophthalmol ; 257(11): 2401-2427, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31529323

ABSTRACT

PURPOSE: This study was conducted in order to study Sostdc1 expression in rat and human developing and adult eyes. METHODS: Using the yeast signal sequence trap screening method, we identified the Sostdc1 cDNA encoding a protein secreted by the adult rat retinal pigment epithelium. We determined by in situ hybridization, RT-PCR, immunohistochemistry, and western blot analysis Sostdc1 gene and protein expression in developing and postnatal rat ocular tissue sections. We also investigated Sostdc1 immunohistolocalization in developing and adult human ocular tissues. RESULTS: We demonstrated a prominent Sostdc1 gene expression in the developing rat central nervous system (CNS) and eyes at early developmental stages from E10.5 days postconception (dpc) to E13 dpc. Specific Sostdc1 immunostaining was also detected in most adult cells of rat ocular tissue sections. We also identified the rat ocular embryonic compartments characterized by a specific Sostdc1 immunohistostaining and specific Pax6, Sox2, Otx2, and Vsx2 immunohistostaining from embryonic stages E10.5 to E13 dpc. Furthermore, we determined the localization of SOSTDC1 immunoreactivity in ocular tissue sections of developing and adult human eyes. Indeed, we detected SOSTDC1 immunostaining in developing and adult human retinal pigment epithelium (RPE) and neural retina (NR) as well as in several developing and adult human ocular compartments, including the walls of choroidal and scleral vessels. Of utmost importance, we observed a strong SOSTDC1 expression in a pathological ocular specimen of type 2 Peters' anomaly complicated by retinal neovascularization as well in the walls ofother pathological extra-ocular vessels.  CONCLUSION: As rat Sostdc1 and human SOSTDC1 are dual antagonists of the Wnt/ß-catenin and BMP signaling pathways, these results underscore the potential crucial roles of these pathways and their antagonists, such as Sostdc1 and SOSTDC1, in developing and adult mammalian normal eyes as well as in syndromic and nonsyndromic congenital eye diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Eye Diseases, Hereditary/genetics , Gene Expression Regulation, Developmental , RNA/genetics , Retinal Pigment Epithelium/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Aged , Animals , Blotting, Western , Child, Preschool , Disease Models, Animal , Eye Diseases, Hereditary/metabolism , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Rats , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/growth & development
10.
Biochem Biophys Res Commun ; 515(4): 524-530, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31176487

ABSTRACT

Stem cells have regenerative potentials that can be used for the treatment of critical and incurable diseases. Age-related macular degeneration (ARMD) and diabetic retinopathy are one of the most severe retinal disorders, which are mostly attributed to impairment of retinal pigmented epithelium (RPE). Thus, restoration of RPE is the main therapeutic approach to prevent the development of ocular diseases, such as ARMD. In this study, we have investigated the role of substance P (SP) on bone marrow mesenchymal stem cell (MSC)-mediated RPE regeneration in vitro. The MSCs were primed with SP followed by the addition of conditioned medium (MSCSP-CM) to RPE. The effects of MSCSP-CM on RPE activity was evaluated by assessing viability, proliferation rate, and migration of RPE. Ex vivo long-term culture led to altered cellular characteristics of MSCs by weakening cell viability, cytokine secretion, and differentiation potential. The conditioned medium of early passage MSC (E-MSCCM) enhanced the RPE viability and migration, whereas the late passage MSC (L-MSCCM) hardly influenced the RPE activity. SP priming, however, facilitated the inductive effects of MSC, and SP effect was more distinct in the late passage than in the early passage. Moreover, it was revealed that SP could exert its effects by modulating PDGF-BB secretion in the MSCs. Taken together, these results suggested that SP could restore the therapeutic effects of MSCs on retinal diseases by elevating their proliferative and paracrine activities through PDGF-PDGFR signaling in ex vivo culture.


Subject(s)
Becaplermin/metabolism , Mesenchymal Stem Cells/cytology , Retinal Pigment Epithelium/growth & development , Substance P/pharmacology , Adipocytes/cytology , Cell Differentiation , Cell Line , Cell Movement , Cell Proliferation , Cell Survival , Cellular Senescence , Culture Media, Conditioned/chemistry , Cytokines/metabolism , Humans , Paracrine Communication , Regeneration , Signal Transduction
11.
Am J Ophthalmol ; 206: 113-131, 2019 10.
Article in English | MEDLINE | ID: mdl-31078532

ABSTRACT

PURPOSE: To investigate the role of fibroblast growth factors (FGFs) in the production of neural retina (NR) and retinal pigmented epithelium (RPE) in a human pluripotent stem cell model of early retinal development. METHODS: Human induced pluripotent stem cell (hiPSC) lines from an individual with microphthalmia caused by a functional null mutation (R200Q) in visual system homeobox 2 (VSX2), a transcription factor involved in early NR progenitor cell (NRPC) production, and a normal sibling were differentiated along the retinal and forebrain lineages using an established protocol. Quantitative and global gene expression analyses (microarray and RNAseq) were used to investigate endogenous FGF expression profiles in these cultures over time. Based on these results, mutant and control hiPSC cultures were treated exogenously with selected FGFs and subjected to gene and protein expression analyses to determine their effects on RPE and NR production. RESULTS: We found that FGF9 and FGF19 were selectively increased in early hiPSC-derived optic vesicles (OVs) when compared to isogenic cultures of hiPSC-derived forebrain neurospheres. Furthermore, these same FGFs were downregulated over time in (R200Q)VSX2 hiPSC-OVs relative to sibling control hiPSC-OVs. Interestingly, long-term supplementation with FGF9, but not FGF19, partially rescued the mutant retinal phenotype of the (R200Q)VSX2 hiPSC-OV model. However, antagonizing FGF9 in wild-type control hiPSCs did not alter OV development. CONCLUSIONS: Our results show that FGF9 acts in concert with VSX2 to promote NR differentiation in hiPSC-OVs and has potential to be used to manipulate early retinogenesis and mitigate ocular defects caused by functional loss of VSX2 activity. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.


Subject(s)
Fibroblast Growth Factor 9/genetics , Gene Expression Regulation, Developmental , Induced Pluripotent Stem Cells/cytology , Microphthalmos/genetics , Retinal Pigment Epithelium/growth & development , Blotting, Western , Cell Differentiation , Cells, Cultured , Fibroblast Growth Factor 9/biosynthesis , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Microphthalmos/metabolism , Microphthalmos/pathology , Phenotype , RNA/genetics , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism
12.
Sci Rep ; 9(1): 3860, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30846751

ABSTRACT

The epigenetic plasticity of amphibian retinal pigment epithelium (RPE) allows them to regenerate the entire retina, a trait known to be absent in mammals. In this study, we investigated the epigenetic plasticity of adult murine RPE to identify possible mechanisms that prevent mammalian RPE from regenerating retinal tissue. RPE were analyzed using microarray, ChIP-seq, and whole-genome bisulfite sequencing approaches. We found that the majority of key genes required for progenitor phenotypes were in a permissive chromatin state and unmethylated in RPE. We observed that the majority of non-photoreceptor genes had promoters in a repressive chromatin state, but these promoters were in unmethylated or low-methylated regions. Meanwhile, the majority of promoters for photoreceptor genes were found in a permissive chromatin state, but were highly-methylated. Methylome states of photoreceptor-related genes in adult RPE and embryonic retina (which mostly contain progenitors) were very similar. However, promoters of these genes were demethylated and activated during retinal development. Our data suggest that, epigenetically, adult murine RPE cells are a progenitor-like cell type. Most likely two mechanisms prevent adult RPE from reprogramming and differentiating into retinal neurons: 1) repressive chromatin in the promoter regions of non-photoreceptor retinal neuron genes; 2) highly-methylated promoters of photoreceptor-related genes.


Subject(s)
Epigenesis, Genetic , Regeneration/physiology , Retinal Pigment Epithelium/metabolism , Animals , DNA Methylation , Male , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Retinal Pigment Epithelium/growth & development , Stem Cells/metabolism
13.
PLoS Genet ; 15(1): e1007939, 2019 01.
Article in English | MEDLINE | ID: mdl-30695061

ABSTRACT

The retinal pigment epithelium (RPE) is a specialized monolayer of pigmented cells within the eye that is critical for maintaining visual system function. Diseases affecting the RPE have dire consequences for vision, and the most prevalent of these is atrophic (dry) age-related macular degeneration (AMD), which is thought to result from RPE dysfunction and degeneration. An intriguing possibility for treating RPE degenerative diseases like atrophic AMD is the stimulation of endogenous RPE regeneration; however, very little is known about the mechanisms driving successful RPE regeneration in vivo. Here, we developed a zebrafish transgenic model (rpe65a:nfsB-eGFP) that enabled ablation of large swathes of mature RPE. RPE ablation resulted in rapid RPE degeneration, as well as degeneration of Bruch's membrane and underlying photoreceptors. Using this model, we demonstrate for the first time that zebrafish are capable of regenerating a functional RPE monolayer after RPE ablation. Regenerated RPE cells first appear at the periphery of the RPE, and regeneration proceeds in a peripheral-to-central fashion. RPE ablation elicits a robust proliferative response in the remaining RPE. Subsequently, proliferative cells move into the injury site and differentiate into RPE. BrdU incorporation assays demonstrate that the regenerated RPE is likely derived from remaining peripheral RPE cells. Pharmacological disruption using IWR-1, a Wnt signaling antagonist, significantly reduces cell proliferation in the RPE and impairs overall RPE recovery. These data demonstrate that the zebrafish RPE possesses a robust capacity for regeneration and highlight a potential mechanism through which endogenous RPE regenerate in vivo.


Subject(s)
Macular Degeneration/genetics , Regeneration/genetics , Retinal Pigment Epithelium/growth & development , cis-trans-Isomerases/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Apoptosis/genetics , Bruch Membrane/growth & development , Bruch Membrane/metabolism , Cell Differentiation/genetics , Disease Models, Animal , Green Fluorescent Proteins/genetics , Humans , Imides/administration & dosage , Larva/genetics , Larva/growth & development , Macular Degeneration/pathology , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Quinolines/administration & dosage , Retina/growth & development , Retina/pathology , Retinal Pigment Epithelium/metabolism , Wnt Signaling Pathway/drug effects , Zebrafish/genetics , Zebrafish/growth & development
14.
Stem Cells ; 37(5): 593-598, 2019 05.
Article in English | MEDLINE | ID: mdl-30548510

ABSTRACT

The rapid improvements in single cell sequencing technologies and analyses afford greater scope for dissecting organoid cultures composed of multiple cell types and create an opportunity to interrogate these models to understand tissue biology, cellular behavior and interactions. To this end, retinal organoids generated from human embryonic stem cells (hESCs) were analyzed by single cell RNA-sequencing (scRNA-Seq) at three time points of differentiation. Combinatorial data from all time points revealed the presence of nine clusters, five of which corresponded to key retinal cell types: retinal pigment epithelium (RPE), retinal ganglion cells (RGCs), cone and rod photoreceptors, and Müller glia. The remaining four clusters expressed genes typical of mitotic cells, extracellular matrix components and those involved in homeostasis. The cell clustering analysis revealed the decreasing presence of mitotic cells and RGCs, formation of a distinct RPE cluster, the emergence of cone and rod photoreceptors from photoreceptor precursors, and an increasing number of Müller glia cells over time. Pseudo-time analysis resembled the order of cell birth during retinal development, with the mitotic cluster commencing the trajectory and the large majority of Müller glia completing the time line. Together, these data demonstrate the feasibility and potential of scRNA-Seq to dissect the inherent complexity of retinal organoids and the orderly birth of key retinal cell types. Stem Cells 2019;37:593-598.


Subject(s)
Cell Differentiation/genetics , Organoids/cytology , Pluripotent Stem Cells/cytology , Retina/growth & development , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Human Embryonic Stem Cells/cytology , Humans , RNA-Seq/methods , Retina/cytology , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/metabolism , Single-Cell Analysis/methods
15.
PLoS One ; 13(11): e0207222, 2018.
Article in English | MEDLINE | ID: mdl-30440011

ABSTRACT

The retinal pigment epithelium (RPE) is an epithelial monolayer in the back of the vertebrate eye. RPE dysfunction is associated with retinal degeneration and blindness. In order to fully understand how dysregulation affects visual function, RPE-specific gene knockouts are indispensable. Since the currently available RPE-specific Cre recombinases show lack of specificity or poor recombination, we sought to generate an alternative. We generated a tamoxifen-inducible RPE-specific Cre transgenic mouse line under transcriptional control of an RPE-specific Tyrosinase enhancer. We characterized the Cre-mediated recombinant expression by crossing our RPE-Tyrosinase-CreErT2 mouse line with the tdTomato reporter line, Ai14. Detected fluorescence was quantified via high-content image analysis. Recombination was predominantly observed in the RPE and adjacent ciliary body. RPE flatmount preparations revealed a high level of recombination in adult mice (47.25-69.48%). Regional analysis of dorsal, ventral, nasal and temporal areas did not show significant changes in recombination. However, recombination was higher in the central RPE compared to the periphery. Higher levels of Cre-mediated recombinant expression was observed in embryonic RPE (~83%). Compared to other RPE-specific Cre transgenic mouse lines, this newly generated RPE-Tyrosinase-CreErT2 line shows a more uniform and higher level of recombination with the advantage to initiate recombination in both, prenatal and postnatal animals. This line can serve as a valuable tool for researches exploring the role of individual gene functions, in both developing and differentiated RPE.


Subject(s)
Mice, Transgenic , Retinal Pigment Epithelium/metabolism , Animals , Female , Integrases , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Inbred C57BL , Models, Animal , Monophenol Monooxygenase/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/ultrastructure
16.
Fluids Barriers CNS ; 15(1): 22, 2018 Aug 16.
Article in English | MEDLINE | ID: mdl-30111340

ABSTRACT

BACKGROUND: Choroid plexus epithelial cells express high levels of transthyretin, produce cerebrospinal fluid and many of its proteins, and make up the blood-cerebrospinal fluid barrier. Choroid plexus epithelial cells are vital to brain health and may be involved in neurological diseases. Transgenic mice containing fluorescent and luminescent reporters of these cells would facilitate their study in health and disease, but prior transgenic reporters lost expression over the early postnatal period. METHODS: Human bacterial artificial chromosomes in which the transthyretin coding sequence was replaced with DNA for tdTomato or luciferase 2 were used in pronuclear injections to produce transgenic mice. These mice were characterized by visualizing red fluorescence, immunostaining, real-time reverse transcription polymerase chain reaction, and luciferase enzyme assay. RESULTS: Reporters were faithfully expressed in cells that express transthyretin constitutively, including choroid plexus epithelial cells, retinal pigment epithelium, pancreatic islets, and liver. Expression of tdTomato in choroid plexus began at the appropriate embryonic age, being detectable by E11.5. Relative levels of tdTomato transcript in the liver and choroid plexus paralleled relative levels of transcripts for transthyretin. Expression remained robust over the first postnatal year, although choroid plexus transcripts of tdTomato declined slightly with age whereas transthyretin remained constant. TdTomato expression patterns were consistent across three founder lines, displayed no sex differences, and were stable across several generations. Two of the tdTomato lines were bred to homozygosity, and homozygous mice are healthy and fertile. The usefulness of tdTomato reporters in visualizing and analyzing live Transwell cultures was demonstrated. Luciferase activity was very high in homogenates of choroid plexus and continued to be expressed through adulthood. Luciferase also was detectable in eye and pancreas. CONCLUSIONS: Transgenic mice bearing fluorescent and luminescent reporters of transthyretin should prove useful for tracking transplanted choroid plexus epithelial cells, for purifying the cells, and for reporting their derivation from stem cells. They also should prove useful for studying transthyretin synthesis by other cell types, as transthyretin has been implicated in many functions and conditions, including clearance of ß-amyloid peptides associated with Alzheimer's disease, heat shock in neurons, processing of neuropeptides, nerve regeneration, astrocyte metabolism, and transthyretin amyloidosis.


Subject(s)
Choroid Plexus/cytology , Epithelial Cells/cytology , Luminescent Proteins/metabolism , Mice, Transgenic , Models, Animal , Prealbumin/metabolism , Animals , Cell Culture Techniques , Cells, Cultured , Choroid Plexus/growth & development , Choroid Plexus/metabolism , Chromosomes, Artificial, Bacterial , Epithelial Cells/metabolism , Humans , Islets of Langerhans/cytology , Islets of Langerhans/growth & development , Islets of Langerhans/metabolism , Liver/cytology , Liver/growth & development , Liver/metabolism , Luminescent Proteins/genetics , Prealbumin/genetics , RNA, Messenger/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism
17.
Development ; 145(8)2018 04 25.
Article in English | MEDLINE | ID: mdl-29615467

ABSTRACT

In the adult central nervous system, endothelial and neuronal cells engage in tight cross-talk as key components of the so-called neurovascular unit. Impairment of this important relationship adversely affects tissue homeostasis, as observed in neurodegenerative conditions including Alzheimer's and Parkinson's disease. In development, the influence of neuroprogenitor cells on angiogenesis is poorly understood. Here, we show in mouse that these cells interact intimately with the growing retinal vascular network, and we identify a novel regulatory mechanism of vasculature development mediated by hypoxia-inducible factor 2a (Hif2a). By Cre-lox gene excision, we show that Hif2a in retinal neuroprogenitor cells upregulates the expression of the pro-angiogenic mediators vascular endothelial growth factor and erythropoietin, whereas it locally downregulates the angiogenesis inhibitor endostatin. Importantly, absence of Hif2a in retinal neuroprogenitor cells causes a marked reduction of proliferating endothelial cells at the angiogenic front. This results in delayed retinal vascular development, fewer major retinal vessels and reduced density of the peripheral deep retinal vascular plexus. Our findings demonstrate that retinal neuroprogenitor cells are a crucial component of the developing neurovascular unit.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Retinal Vessels/growth & development , Retinal Vessels/innervation , Animals , Astrocytes/cytology , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Endostatins/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neovascularization, Physiologic/genetics , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism , Retinal Vessels/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Nat Biotechnol ; 36(4): 328-337, 2018 04.
Article in English | MEDLINE | ID: mdl-29553577

ABSTRACT

Age-related macular degeneration (AMD) remains a major cause of blindness, with dysfunction and loss of retinal pigment epithelium (RPE) central to disease progression. We engineered an RPE patch comprising a fully differentiated, human embryonic stem cell (hESC)-derived RPE monolayer on a coated, synthetic basement membrane. We delivered the patch, using a purpose-designed microsurgical tool, into the subretinal space of one eye in each of two patients with severe exudative AMD. Primary endpoints were incidence and severity of adverse events and proportion of subjects with improved best-corrected visual acuity of 15 letters or more. We report successful delivery and survival of the RPE patch by biomicroscopy and optical coherence tomography, and a visual acuity gain of 29 and 21 letters in the two patients, respectively, over 12 months. Only local immunosuppression was used long-term. We also present the preclinical surgical, cell safety and tumorigenicity studies leading to trial approval. This work supports the feasibility and safety of hESC-RPE patch transplantation as a regenerative strategy for AMD.


Subject(s)
Human Embryonic Stem Cells/transplantation , Macular Degeneration/therapy , Retinal Pigment Epithelium/transplantation , Visual Acuity/physiology , Aged , Animals , Basement Membrane/diagnostic imaging , Basement Membrane/growth & development , Cell Differentiation/genetics , Female , Humans , Macular Degeneration/diagnostic imaging , Macular Degeneration/pathology , Male , Mice , Middle Aged , Retinal Pigment Epithelium/diagnostic imaging , Retinal Pigment Epithelium/growth & development , Stem Cell Transplantation/adverse effects , Swine , Tomography, Optical Coherence
19.
Nat Cell Biol ; 20(2): 175-185, 2018 02.
Article in English | MEDLINE | ID: mdl-29335527

ABSTRACT

Primary cilia play essential roles in signal transduction and development. The docking of preciliary vesicles at the distal appendages of a mother centriole is an initial/critical step of ciliogenesis, but the mechanisms are unclear. Here, we demonstrate that myosin-Va mediates the transportation of preciliary vesicles to the mother centriole and reveal the underlying mechanism. We also show that the myosin-Va-mediated transportation of preciliary vesicles is the earliest event that defines the onset of ciliogenesis. Depletion of myosin-Va significantly inhibits the attachment of preciliary vesicles to the distal appendages of the mother centriole and decreases cilia assembly. Myosin-Va functions upstream of EHD1- and Rab11-mediated ciliary vesicle formation. Importantly, dynein mediates myosin-Va-associated preciliary vesicle transportation to the pericentrosomal region along microtubules, while myosin-Va mediates preciliary vesicle transportation from the pericentrosomal region to the distal appendages of the mother centriole via the Arp2/3-associated branched actin network.


Subject(s)
Cilia/genetics , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics , Actin-Related Protein 2/genetics , Actins/genetics , Animals , Biological Transport/genetics , Centrioles/genetics , Centrioles/metabolism , Cilia/metabolism , Humans , Mice , Microtubules/genetics , Microtubules/metabolism , Myosin Heavy Chains/antagonists & inhibitors , Myosin Type V/antagonists & inhibitors , NIH 3T3 Cells , Primary Cell Culture , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism , Signal Transduction
20.
Exp Mol Med ; 49(12): e411, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29244789

ABSTRACT

Clinical expansion of mesenchymal stem cells (MSCs) is hampered by the lack of knowledge regarding how to prevent MSC apoptosis and promote their proliferation in serum-free medium. Our in vitro studies demonstrated that human umbilical cord MSCs (HUCMSCs) underwent apoptosis in the serum-free medium. When HUCMSCs were co-cultured with retinal pigment epithelial cells (ARPE19), however, HUCMSCs exhibited normal growth and morphology in serum-free medium. Their colony formation was promoted by the conditioned medium (CM) of ARPE19 cells on Matrigel. Proteomics analysis showed that pigment epithelium-derived factor (PEDF) was one of the most abundant extracellular proteins in the ARPE19 CM, whereas enzyme-linked immunosorbent assay confirmed that large amounts of PEDF was secreted from ARPE19 cells. Adding anti-PEDF-blocking antibodies to the co-culture of HUCMSCs with ARPE19 cells increased apoptosis of HUCMSCs. Conversely, treatment with PEDF significantly reduced apoptosis and increased proliferation of HUCMSCs in serum-free medium. PEDF was further demonstrated to exert this anti-apoptotic effect by inhibiting P53 expression to suppress caspase activation. In vivo studies demonstrated that co-injection of HUCMSCs with ARPE19 cells in immunocompromised NOD-SCID mice also increased survival and decreased apoptosis of HUCMSCs. PEDF also showed no negative effect on the mesoderm differentiation capability of HUCMSCs. In conclusion, this study is the first to demonstrate that PEDF promotes HUCMSC proliferation and protects them from apoptosis by reducing p53 expression in the serum-free medium. This study provides crucial information for clinical-scale expansion of HUCMSCs.


Subject(s)
Cell Differentiation/drug effects , Eye Proteins/drug effects , Mesenchymal Stem Cells/cytology , Nerve Growth Factors/drug effects , Serpins/drug effects , Umbilical Cord/cytology , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Collagen/pharmacology , Culture Media, Serum-Free/pharmacology , Drug Combinations , Gene Expression Regulation, Developmental/drug effects , Humans , Laminin/pharmacology , Mesenchymal Stem Cells/drug effects , Mesoderm/cytology , Mesoderm/growth & development , Proteoglycans/pharmacology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/growth & development , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...