Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Neurosci ; 43(49): 8367-8384, 2023 12 06.
Article in English | MEDLINE | ID: mdl-37775301

ABSTRACT

The nuclear factor one (NFI) transcription factor genes Nfia, Nfib, and Nfix are all enriched in late-stage retinal progenitor cells, and their loss has been shown to retain these progenitors at the expense of later-generated retinal cell types. Whether they play any role in the specification of those later-generated fates is unknown, but the expression of one of these, Nfia, in a specific amacrine cell type may intimate such a role. Here, Nfia conditional knockout (Nfia-CKO) mice (both sexes) were assessed, finding a massive and largely selective absence of AII amacrine cells. There was, however, a partial reduction in type 2 cone bipolar cells (CBCs), being richly interconnected to AII cells. Counts of dying cells showed a significant increase in Nfia-CKO retinas at postnatal day (P)7, after AII cell numbers were already reduced but in advance of the loss of type 2 CBCs detected by P10. Those results suggest a role for Nfia in the specification of the AII amacrine cell fate and a dependency of the type 2 CBCs on them. Delaying the conditional loss of Nfia to the first postnatal week did not alter AII cell number nor differentiation, further suggesting that its role in AII cells is solely associated with their production. The physiological consequences of their loss were assessed using the ERG, finding the oscillatory potentials to be profoundly diminished. A slight reduction in the b-wave was also detected, attributed to an altered distribution of the terminals of rod bipolar cells, implicating a role of the AII amacrine cells in constraining their stratification.SIGNIFICANCE STATEMENT The transcription factor NFIA is shown to play a critical role in the specification of a single type of retinal amacrine cell, the AII cell. Using an Nfia-conditional knockout mouse to eliminate this population of retinal neurons, we demonstrate two selective bipolar cell dependencies on the AII cells; the terminals of rod bipolar cells become mis-stratified in the inner plexiform layer, and one type of cone bipolar cell undergoes enhanced cell death. The physiological consequence of this loss of the AII cells was also assessed, finding the cells to be a major contributor to the oscillatory potentials in the electroretinogram.


Subject(s)
Amacrine Cells , NFI Transcription Factors , Retina , Animals , Female , Male , Mice , Amacrine Cells/metabolism , Electroretinography , NFI Transcription Factors/metabolism , Retina/metabolism , Retinal Bipolar Cells , Transcription Factors/metabolism
2.
Stem Cell Reports ; 18(3): 782-797, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36801002

ABSTRACT

Chromosome 20 abnormalities are some of the most frequent genomic changes acquired by human pluripotent stem cell (hPSC) cultures worldwide. Yet their effects on differentiation remain largely unexplored. We investigated a recurrent abnormality also found on amniocentesis, the isochromosome 20q (iso20q), during a clinical retinal pigment epithelium differentiation. Here we show that the iso20q abnormality interrupts spontaneous embryonic lineage specification. Isogenic lines revealed that under conditions that promote the spontaneous differentiation of wild-type hPSCs, the iso20q variants fail to differentiate into primitive germ layers and to downregulate pluripotency networks, resulting in apoptosis. Instead, iso20q cells are highly biased for extra-embryonic/amnion differentiation following inhibition of DNMT3B methylation or BMP2 treatment. Finally, directed differentiation protocols can overcome the iso20q block. Our findings reveal in iso20q a chromosomal abnormality that impairs the developmental competency of hPSCs toward germ layers but not amnion, which models embryonic developmental bottlenecks in the presence of aberrations.


Subject(s)
Isochromosomes , Pluripotent Stem Cells , Humans , Cell Differentiation/genetics , Pluripotent Stem Cells/metabolism , Retinal Pigment Epithelium , Germ Layers
3.
EClinicalMedicine ; 51: 101604, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35996565

ABSTRACT

Background: A potential immunotherapeutic role for AZD1656 (a glucokinase activator) in the treatment of COVID-19 was hypothesized. The ARCADIA trial investigated the safety and efficacy of AZD1656 in diabetic patients admitted to hospital with COVID-19. Methods: The ARCADIA trial was a Phase II randomised, double-blind, placebo-controlled clinical trial. Adult diabetic patients, admitted with COVID-19, were recruited at 28 hospitals in the UK, Romania and Czech Republic and randomly assigned (1:1) to receive AZD1656 tablets (100mg twice a day), or matched placebo, for up to 21 days, in addition to usual care. All involved were masked to treatment allocation. The primary endpoint was clinical improvement measured at Day 14. The Full Analysis Set (FAS) included all patients who received at least one dose of assigned treatment. ARCADIA is complete and registered with ClinicalTrials.gov (NCT04516759). Findings: Between 29 September 2020 to 16 April 2021, 170 patients were screened and 156 patients were randomised, three of whom did not commence treatment. Of the remaining 153, 80 were assigned to AZD1656 and 73 were assigned to placebo and included in the Full Analysis Set (FAS). The primary analysis showed no statistically significant difference between groups (AZD1656: 76·3%; Placebo: 69·9%, p=0·19). There was no difference in the number of adverse events between groups (AZD1656: 35·7%; Placebo: 33·3%). Mortality was lower in the AZD1656 group compared to the placebo group (AZD1656: four (5%); Placebo: nine (12·3%), p=0·090)). At Day 7 there were zero deaths in the AZD1656 group compared to six deaths in the placebo group (p=0·011, post hoc). A difference between groups in time to hospital discharge was also seen (p=0·16). Immunophenotyping data suggested that AZD1656-treated patients had a less pro-inflammatory immune response and a better adaptive immune response than those treated with placebo. Interpretation: Although the trial did not achieve its primary endpoint, AZD1656 was associated with a decrease in deaths and a reduction in the duration of hospitalisation, as compared to Placebo. Immunophenotyping and immunochemistry indicated an immunomodulatory effect of AZD1656. The trial suggests a beneficial therapeutic effect of AZD1656 and identifies a new therapeutic concept: small molecule activation of endogenous homeostatic immune cells which themselves become the therapeutic agent within the body. Phase 2 trials of this size carry the risk of false positive results and confirmation of these results in a larger clinical trial is now required. Funding: UK Research and Innovation (UKRI) 'Innovate UK' programme and Excalibur Medicines Ltd.

4.
PLoS One ; 15(7): e0236298, 2020.
Article in English | MEDLINE | ID: mdl-32701996

ABSTRACT

Degeneration of the retinal pigment epithelium (RPE) plays a central role in age-related macular degeneration (AMD). Throughout life, RPE cells are challenged by a variety of cytotoxic stressors, some of which are cumulative with age and may ultimately contribute to drusen and lipofuscin accumulation. Stressors such as these continually damage RPE cells resulting in a state of chronic wounding. Current cell-based platforms that model a state of chronic RPE cell wounding are limited, and the RPE cellular response is not entirely understood. Here, we used the electric cell-substrate impedance sensing (ECIS) system to induce a state of acute or chronic wounding on differentiated human fetal RPE cells to analyze changes in the wound repair response. RPE cells surrounding the lesioned area employ both cell migration and proliferation to repair wounds but fail to reestablish their original cell morphology or density after repetitive wounding. Chronically wounded RPE cells develop phenotypic AMD characteristics such as loss of cuboidal morphology, enlarged size, and multinucleation. Transcriptomic analysis suggests a systemic misregulation of RPE cell functions in bystander cells, which are not directly adjacent to the wound. Genes associated with the major RPE cell functions (LRAT, MITF, RDH11) significantly downregulate after wounding, in addition to differential expression of genes associated with the cell cycle (CDK1, CDC6, CDC20), inflammation (IL-18, CCL2), and apoptosis (FAS). Interestingly, repetitive wounding resulted in prolonged misregulation of genes, including FAS, LRAT, and PEDF. The use of ECIS to induce wounding resulted in an over-representation of AMD-associated genes among those dysregulated genes, particularly genes associated with advanced AMD. This simple system provides a new model for further investigation of RPE cell wound response in AMD pathogenesis.


Subject(s)
Macular Degeneration/pathology , Models, Biological , Acute Disease , Bystander Effect , Cell Cycle , Cell Differentiation , Cell Proliferation , Chronic Disease , Fetus/pathology , Gene Expression Profiling , Gene Ontology , Humans , Kinetics , Macular Degeneration/genetics , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/pathology , Transcriptome/genetics , Wound Healing
5.
Curr Eye Res ; 45(3): 361-371, 2020 03.
Article in English | MEDLINE | ID: mdl-31777296

ABSTRACT

Retinal pigment epithelium (RPE) degradation is central to the onset and progression of age-related macular degeneration (AMD), a growing and currently incurable form of blindness.Due to its key role in maintaining the retinal structure and homeostasis, cell replacement of the RPE monolayer has emerged as a promising therapy to rescue visual acuity in AMD patients.Thanks to the tremendous progress of pluripotent stem cell technologies over the last decade, a potentially unlimited new source for RPE transplantation has reached clinical trials. This review summarizes the methods by which pluripotent stem cell-based RPE cells are produced for transplantation, the delivery methods currently being adopted and the latest clinical outcomes with regard to the treatment of AMD.


Subject(s)
Macular Degeneration/therapy , Retinal Pigment Epithelium/pathology , Stem Cell Transplantation/methods , Visual Acuity , Disease Progression , Humans , Macular Degeneration/pathology
6.
Stem Cells Transl Med ; 8(8): 775-784, 2019 08.
Article in English | MEDLINE | ID: mdl-31037833

ABSTRACT

Glaucoma is one of the leading causes of blindness, and there is an ongoing need for new therapies. Recent studies indicate that cell transplantation using Müller glia may be beneficial, but there is a need for novel sources of cells to provide therapeutic benefit. In this study, we have isolated Müller glia from retinal organoids formed by human induced pluripotent stem cells (hiPSCs) in vitro and have shown their ability to partially restore visual function in rats depleted of retinal ganglion cells by NMDA. Based on the present results, we suggest that Müller glia derived from retinal organoids formed by hiPSC may provide an attractive source of cells for human retinal therapies, to prevent and treat vision loss caused by retinal degenerative conditions. Stem Cells Translational Medicine 2019;8:775&784.


Subject(s)
Cell Transplantation/methods , Ependymoglial Cells/transplantation , Induced Pluripotent Stem Cells/cytology , Retinal Degeneration/therapy , Retinal Ganglion Cells/physiology , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Ependymoglial Cells/cytology , Humans , Induced Pluripotent Stem Cells/transplantation , Organoids/cytology , Phenotype , Rats , Regeneration , Retinal Ganglion Cells/pathology
7.
Invest Ophthalmol Vis Sci ; 58(1): 430-441, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28118667

ABSTRACT

Purpose: Transforming growth factor ß-mediated epithelial-to-mesenchymal transition (EMT) is a major component of the wound healing response and a negative determinant of retinal pigment epithelium (RPE) differentiation. We have shown previously that inhibition of TGFß signaling restored the capacity of mesenchymal RPE to differentiate; however, the potential lessens with extensive passaging. We investigated TGFß-independent mechanisms that regulate RPE differentiation following repetitive passaging. Methods: Retinal pigment epithelium-EMT was induced by repetitive passaging of fetal RPE at subconfluence. Suppression of EMT was achieved by the addition of A-83-01, a TGFß receptor kinase inhibitor. Transcriptomic analysis was used to identify potential TGFß independent processes that prevent differentiation after extensive passage. Downregulated transcription factors were identified and transduced into highly passaged RPE to restore cell differentiation. Restoration was evaluated according to morphology, expression of RPE/mesenchymal markers, transcriptomic analysis, cell doubling time, and senescence-associated ß-galactosidase (SA-ß-gal) activity. Results: A-83-01-treated RPE failed to differentiate after 7 passages (P7). This failure was concomitant with downregulation of RPE genes, misregulation of cell cycle genes, a decline in proliferative potential, and cell senescence. Exogenous expression of MYCN and OTX2 in conjunction with A-83-01 restored P7-RPE differentiation to a status similar to minimally passaged RPE. Moreover, the treatment allowed cells to maintain their differentiation capacity after extended passaging. Conclusions: Retinal pigment epithelium subjected to chronic wound stimulus undergoes TGFß-mediated EMT, loss of expression of signature RPE genes, and senescence. Targeting these aspects with a TGFß receptor kinase inhibitor, a RPE transcription factor, and a cell cycle regulator restores the capacity of highly passaged RPE cells to regenerate and differentiate.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Retinal Pigment Epithelium/metabolism , Transcription Factors/genetics , Cell Differentiation , Cell Movement , Cells, Cultured , Epithelial-Mesenchymal Transition/drug effects , Humans , Immunohistochemistry , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Signal Transduction/drug effects , Transcription Factors/metabolism , Transforming Growth Factor beta2/pharmacology
8.
Genome Med ; 7(1): 58, 2015.
Article in English | MEDLINE | ID: mdl-26150894

ABSTRACT

BACKGROUND: Age-related macular degeneration (AMD) is a leading cause of blindness. Most vision loss occurs following the transition from a disease of deposit formation and inflammation to a disease of neovascular fibrosis and/or cell death. Here, we investigate how repeated wound stimulus leads to seminal changes in gene expression and the onset of a perpetual state of stimulus-independent wound response in retinal pigmented epithelial (RPE) cells, a cell-type central to the etiology of AMD. METHODS: Transcriptome wide expression profiles of human fetal RPE cell cultures as a function of passage and time post-plating were determined using Agilent 44 K whole genome microarrays and RNA-Seq. Using a systems level analysis, differentially expressed genes and pathways of interest were identified and their role in the establishment of a persistent mesenchymal state was assessed using pharmacological-based experiments. RESULTS: Using a human fetal RPE cell culture model that considers monolayer disruption and subconfluent culture as a proxy for wound stimulus, we show that prolonged wound stimulus leads to terminal acquisition of a mesenchymal phenotype post-confluence and altered expression of more than 40 % of the transcriptome. In contrast, at subconfluence fewer than 5 % of expressed transcripts have two-fold or greater expression differences after repeated passage. Protein-protein and pathway interaction analysis of the genes with passage-dependent expression levels in subconfluent cultures reveals a 158-node interactome comprised of two interconnected modules with functions pertaining to wound response and cell division. Among the wound response genes are the TGFß pathway activators: TGFB1, TGFB2, INHBA, INHBB, GDF6, CTGF, and THBS1. Significantly, inhibition of TGFBR1/ACVR1B mediated signaling using receptor kinase inhibitors both forestalls and largely reverses the passage-dependent loss of epithelial potential; thus extending the effective lifespan by at least four passages. Moreover, a disproportionate number of RPE wound response genes have altered expression in neovascular and geographic AMD, including key members of the TGFß pathway. CONCLUSIONS: In RPE cells the switch to a persistent mesenchymal state following prolonged wound stimulus is driven by lasting activation of the TGFß pathway. Targeted inhibition of TGFß signaling may be an effective approach towards retarding AMD progression and producing RPE cells in quantity for research and cell-based therapies.

9.
Cell Stem Cell ; 16(1): 13-7, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25575079

ABSTRACT

Induced pluripotent stem cells (iPSCs) have the potential to transform drug discovery and healthcare in the 21(st) century. However, successful commercialization will require standardized manufacturing platforms. Here we highlight the need to define standardized practices for iPSC generation and processing and discuss current challenges to the robust manufacture of iPSC products.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Animals , Humans , Reproducibility of Results , Stem Cell Transplantation
10.
Stem Cells Transl Med ; 3(11): 1295-304, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25273541

ABSTRACT

Human embryonic stem cells (hESCs) are a promising source of retinal pigment epithelium (RPE) cells: cells that can be used for the treatment of common and incurable forms of blindness, such as age-related macular degeneration. Although most hESC lines will produce a number of clusters of pigmented RPE cells within 30-50 days when allowed to spontaneously differentiate, the timing and efficiency of differentiation is highly variable. This could prove problematic in the design of robust processes for the large scale production of RPE cells for cell therapy. In this study we sought to identify, quantify, and reduce the sources of variability in hESC-RPE differentiation. By monitoring the emergence of pigmented cells over time, we show how the cell line, passaging method, passage number, and seeding density have a significant and reproducible effect on the RPE yield. To counter this variability, we describe the production of RPE cells from two cell lines in feeder-free, density controlled conditions using single cell dissociation and seeding that is more amenable to scaled up production. The efficacy of small molecules in directing differentiation toward the RPE lineage was tested in two hESC lines with divergent RPE differentiation capacities. Neural induction by treatment with a bone morphogenetic protein inhibitor, dorsomorphin, significantly enhanced the RPE yield in one cell line but significantly reduce it in another, generating instead a Chx10 positive neural progenitor phenotype. This result underlines the necessity to tailor differentiation protocols to suit the innate properties of different cell lines.


Subject(s)
Cell Differentiation , Neural Stem Cells , Pluripotent Stem Cells , Retinal Pigment Epithelium , Cell Culture Techniques , Cell Line , Homeodomain Proteins/biosynthesis , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Transcription Factors/biosynthesis
11.
PLoS One ; 8(11): e81622, 2013.
Article in English | MEDLINE | ID: mdl-24303062

ABSTRACT

A systematic evaluation of three different methods for generating induced pluripotent stem (iPS) cells was performed using the same set of parental cells in our quest to develop a feeder independent and xeno-free method for somatic cell reprogramming that could be transferred into a GMP environment. When using the BJ fibroblast cell line, the highest reprogramming efficiency (1.89% of starting cells) was observed with the mRNA based method which was almost 20 fold higher than that observed with the retrovirus (0.2%) and episomal plasmid (0.10%) methods. Standard characterisation tests did not reveal any differences in an array of pluripotency markers between the iPS lines derived using the various methods. However, when the same methods were used to reprogram three different primary fibroblasts lines, two derived from patients with rapid onset parkinsonism dystonia and one from an elderly healthy volunteer, we consistently observed higher reprogramming efficiencies with the episomal plasmid method, which was 4 fold higher when compared to the retroviral method and over 50 fold higher than the mRNA method. Additionally, with the plasmid reprogramming protocol, recombinant vitronectin and synthemax® could be used together with commercially available, fully defined, xeno-free essential 8 medium without significantly impacting the reprogramming efficiency. To demonstrate the robustness of this protocol, we reprogrammed a further 2 primary patient cell lines, one with retinosa pigmentosa and the other with Parkinsons disease. We believe that we have optimised a simple and reproducible method which could be used as a starting point for developing GMP protocols, a prerequisite for generating clinically relevant patient specific iPS cells.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Animals , Cell Culture Techniques , Cell Line , Fibroblasts/metabolism , Gene Expression , Humans , Mice , Plasmids/genetics , Transduction, Genetic , Transfection , Transgenes
12.
PLoS One ; 8(11): e79188, 2013.
Article in English | MEDLINE | ID: mdl-24223903

ABSTRACT

Multiple sclerosis is an immune-mediated, demyelinating and neurodegenerative disease that currently lacks any neuroprotective treatments. Innovative neuroprotective trial designs are required to hasten the translational process of drug development. An ideal target to monitor the efficacy of strategies aimed at treating multiple sclerosis is the visual system, which is the most accessible part of the human central nervous system. A novel C57BL/6 mouse line was generated that expressed transgenes for a myelin oligodendrocyte glycoprotein-specific T cell receptor and a retinal ganglion cell restricted-Thy1 promoter-controlled cyan fluorescent protein. This model develops spontaneous or induced optic neuritis, in the absence of paralytic disease normally associated with most rodent autoimmune models of multiple sclerosis. Demyelination and neurodegeneration could be monitored longitudinally in the living animal using electrophysiology, visual sensitivity, confocal scanning laser ophthalmoscopy and optical coherence tomography all of which are relevant to human trials. This model offers many advantages, from a 3Rs, economic and scientific perspective, over classical experimental autoimmune encephalomyelitis models that are associated with substantial suffering of animals. Optic neuritis in this model led to inflammatory damage of axons in the optic nerve and subsequent loss of retinal ganglion cells in the retina. This was inhibited by the systemic administration of a sodium channel blocker (oxcarbazepine) or intraocular treatment with siRNA targeting caspase-2. These novel approaches have relevance to the future treatment of neurodegeneration of MS, which has so far evaded treatment.


Subject(s)
Carbamazepine/analogs & derivatives , Disease Models, Animal , Multiple Sclerosis/therapy , Optic Neuritis/therapy , RNA Interference , Animals , Anticonvulsants/pharmacology , Carbamazepine/pharmacology , Caspase 2/genetics , Caspase 2/immunology , Caspase 2/metabolism , Evoked Potentials, Visual/immunology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Ophthalmoscopy , Optic Nerve/immunology , Optic Nerve/metabolism , Optic Neuritis/genetics , Optic Neuritis/immunology , Oxcarbazepine , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Retina/immunology , Retina/metabolism , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/metabolism , Tomography, Optical Coherence
13.
PLoS One ; 8(6): e65518, 2013.
Article in English | MEDLINE | ID: mdl-23799019

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of legal blindness in the Western world. There are effective treatments for the vascular complications of neo-vascular AMD, but no effective therapies are available for the dry/atrophic form of the disease. A previously described transgenic CFH-gene deficient mouse model, (cfh-/-), shows hallmarks of early AMD. The ocular phenotype has been further analysed to demonstrate amyloid beta (Aß) rich basement membrane deposits associated with activated complement C3. Cfh-/- mice were treated systemically in both prophylactic and therapeutic regimes with an anti-Aß monoclonal antibody (mAb), 6F6, to determine the effect on the cfh-/- retinal phenotype. Prophylactic treatment with 6F6 demonstrated a dose dependent reduction in the accumulation of both Aß and activated C3 deposition. A similar reduction in the retinal endpoints could be seen after therapeutic treatment. Serum Aß levels after systemic administration of 6F6 show accumulation of Aß in the periphery suggestive of a peripheral sink mechanism. In summary, anti-Aß mAb treatment can partially prevent or reverse ocular phenotypes of the cfh-/- mouse. The data support this therapeutic approach in humans potentially modulating two key elements in the pathogenesis of AMD - Aß and activated, complement C3.


Subject(s)
Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Complement C3/metabolism , Macular Degeneration/drug therapy , Peptide Fragments/metabolism , Retina/metabolism , Amyloid beta-Peptides/immunology , Animals , Disease Models, Animal , Humans , Hybridomas , Macular Degeneration/immunology , Macular Degeneration/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/immunology , Retina/pathology
14.
Invest Ophthalmol Vis Sci ; 54(2): 1490-500, 2013 Feb 27.
Article in English | MEDLINE | ID: mdl-23385800

ABSTRACT

PURPOSE: Two noninvasive delivery strategies for VEGF/PDGF receptor tyrosine kinase inhibitors (RTKI) were explored that exploited uveal retention as a means for establishing an ocular drug depot: a single oral "loading" dose and topical administration. METHODS: Melanin binding was confirmed by centrifugation and mass spectrometry. Ocular retention was examined in pigmented and albino rats. Ocular release kinetics were measured 3 to 28 days postdosing in pigmented rats. Microautoradiography was used to demonstrate retention of RTKI in the uveal tract. A uveal drug depot of pazopanib was created by a single oral dose prior to induction of laser choroidal neovascularization (CNV). Choroid/retinal pigmented epithelium (RPE) retention of a related RTKI with enhanced topical bioavailability, GW771806, was confirmed by bioanalytics, and its ability to regress CNV compared with pazopanib. RESULTS: Pazopanib and GW771806 directly bound melanin and were retained within the uveal tract of pigmented rats for weeks following a single oral dose. Pazopanib was undetectable systemically following a single oral administration prior to CNV induction, and reduced CNV as well as twice daily dosing. Topical ocular delivery of GW771806 at 5 mg/mL led to high choroidal/RPE exposure and significantly regressed CNV lesions; 2 mg/mL prevented lesion progression. CONCLUSIONS: Uveal retention of drugs such as pazopanib can be used to create a sustained-release depot. Topical GW771806 regressed CNV. These data indicate that topical or infrequent oral loading dose treatment with VEGF/PDGF RTKI retained in the choroid/RPE might allow noninvasive treatments for ocular neovascular disease.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Choroidal Neovascularization/drug therapy , Drug Delivery Systems , Indazoles/administration & dosage , Pyrimidines/administration & dosage , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Sulfonamides/administration & dosage , Sulfones/administration & dosage , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Administration, Oral , Administration, Topical , Angiogenesis Inhibitors/pharmacokinetics , Animals , Autoradiography , Choroidal Neovascularization/diagnosis , Choroidal Neovascularization/metabolism , Female , Fluorescein Angiography , Half-Life , Indazoles/pharmacokinetics , Melanins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Pyrimidines/pharmacokinetics , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Sulfonamides/pharmacokinetics , Sulfones/pharmacokinetics , Uvea/metabolism
15.
Biotechnol Lett ; 34(12): 2307-15, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22983716

ABSTRACT

The commercialisation of human embryonic stem cell derived cell therapies for large patient populations is reliant on both minimising expensive and variable manual-handling methods whilst realising economies of scale. The Quantum Cell Expansion System, a hollow fibre bioreactor (Terumo BCT), was used in a pilot study to expand 60 million human embryonic stem cells to 708 million cells. Further improvements can be expected with optimisation of media flow rates throughout the run to better control the cellular microenvironment. High levels of pluripotency marker expression were maintained on the bioreactor, with 97.7 % of cells expressing SSEA-4 when harvested.


Subject(s)
Bioreactors , Cell Culture Techniques/methods , Embryonic Stem Cells/physiology , Gene Expression , Humans , Stage-Specific Embryonic Antigens/biosynthesis
16.
Cell Stem Cell ; 10(1): 1-2, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22226345

ABSTRACT

The innate capacity of adult somatic cells has many potential applications in regenerative medicine. In this issue of Cell Stem Cell, Salero et al. (2012) describe an adult retinal stem cell population capable of generating neural and mesenchymal cell lineages.

17.
Mol Vis ; 17: 1701-15, 2011.
Article in English | MEDLINE | ID: mdl-21738400

ABSTRACT

PURPOSE: In several species the retinal pigment epithelium (RPE) has the potential to transdifferentiate into retinal cells to regenerate functional retinal tissue after injury. However, this capacity for regeneration is lost in mammals. The synthetic retinoic acid derivative, fenretinide [N(4-hydroxyphenyl) retinamide], induces a neuronal-like phenotype in the human adult retinal pigment epithelial cell line (ARPE-19). These changes are characterized by the appearance of neural-like processes and the expression of neuronal markers not normally associated with RPE cells. Here we assess whether fenretinide can induce a neuroretinal cell phenotype in ARPE-19 cells, by examining retinal cell marker expression. METHODS: ARPE-19 cells were treated daily with culture medium containing either 3 µM fenretinide or dimethyl sulfoxide as a control for 7 days. Cells were processed for immunocytochemistry, western blotting, and for analysis by PCR to examine the expression of a panel of RPE, neural, and retinal-associated cellular markers, including classical and non-canonical opsins. RESULTS: Treatment with fenretinide for 7 days induced the formation of neuronal-like processes in ARPE-19 cells. Fenretinide induced the expression of the cone long wavelength sensitive opsin (OPN1lw) but not rhodopsin (RHO), while decreasing the expression of RPE cell markers. Many of the neuronal and retinal specific markers examined were expressed in both control and fenretinide treated cells, including those involved in photoreceptor cell development and the multipotency of neural retinal progenitor cells. Interestingly, ARPE-19 cells also expressed both photoreceptor specific and non-specific canonical opsins. CONCLUSIONS: The expression of retinal-associated markers and loss of RPE cell markers in control ARPE-19 cells suggests that these cells might have dedifferentiated from an RPE cell phenotype under standard culture conditions. The expression of molecules, such as the transcription factors paired box 6 gene (PAX6), sex determining region Y-box 2 (SOX2), cone-rod homeobox (CRX), and neural retina leucine zipper (NRL), further implies that in culture these cells are predisposed toward a retinal progenitor-like state. The fenretinide-induced increase in photoreceptor cell markers, accompanied by a decrease in RPE cell markers, suggests that retinoids may play a role in the transdifferentiation of RPE cells. Importantly, our data show for the first time the expression of a vertebrate ciliary opsin (OPN1lw) and rhabdomeric-like opsin, opsin 4 (OPN4 also known as melanopsin) in a clonal cell line. Together these data suggest that ARPE-19 cells are primed for and possess the capacity to differentiate toward a retinal cell-like lineage.


Subject(s)
Biomarkers/metabolism , Cell Transdifferentiation/drug effects , Epithelial Cells/drug effects , Fenretinide/pharmacology , Retina/drug effects , Retinal Pigment Epithelium/drug effects , Adult , Blotting, Western , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression/drug effects , Humans , Immunohistochemistry , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Polymerase Chain Reaction , Retina/cytology , Retina/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Retinoids/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
19.
PLoS One ; 4(12): e8152, 2009 Dec 03.
Article in English | MEDLINE | ID: mdl-19997644

ABSTRACT

Transformation of somatic cells with a set of embryonic transcription factors produces cells with the pluripotent properties of embryonic stem cells (ESCs). These induced pluripotent stem (iPS) cells have the potential to differentiate into any cell type, making them a potential source from which to produce cells as a therapeutic platform for the treatment of a wide range of diseases. In many forms of human retinal disease, including age-related macular degeneration (AMD), the underlying pathogenesis resides within the support cells of the retina, the retinal pigment epithelium (RPE). As a monolayer of cells critical to photoreceptor function and survival, the RPE is an ideally accessible target for cellular therapy. Here we report the differentiation of human iPS cells into RPE. We found that differentiated iPS-RPE cells were morphologically similar to, and expressed numerous markers of developing and mature RPE cells. iPS-RPE are capable of phagocytosing photoreceptor material, in vitro and in vivo following transplantation into the Royal College of Surgeons (RCS) dystrophic rat. Our results demonstrate that iPS cells can be differentiated into functional iPS-RPE and that transplantation of these cells can facilitate the short-term maintenance of photoreceptors through phagocytosis of photoreceptor outer segments. Long-term visual function is maintained in this model of retinal disease even though the xenografted cells are eventually lost, suggesting a secondary protective host cellular response. These findings have identified an alternative source of replacement tissue for use in human retinal cellular therapies, and provide a new in vitro cellular model system in which to study RPE diseases affecting human patients.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Retinal Diseases/therapy , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/transplantation , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Polarity , Cell Shape , Cell Survival , Epithelial Cells/cytology , Epithelial Cells/transplantation , Humans , Immunohistochemistry , Macrophages/cytology , Phagocytosis , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/ultrastructure , Proto-Oncogene Proteins c-fos/metabolism , Rats , Retinal Diseases/pathology , Retinal Diseases/physiopathology , Retinal Pigment Epithelium/ultrastructure , Vision, Ocular/physiology
20.
Regen Med ; 4(4): 505-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19580400

ABSTRACT

Professor Coffey joined the faculty at the University of Oxford in 1987 on completing his doctoral work. In 1989, he was awarded a personal Royal Society Research Fellowship, at the same time moving to the University of Sheffield to establish a new laboratory for retinal transplantation. After 14 years at the University of Sheffield, Prof. Coffey was appointed Professor in the newly built Henry Wellcome building for translational eye research at the Institute of Ophthalmology in London. Prof. Coffey has many years experience in cellular therapies as applied to retinal transplantation and was recently the principal author and co-author of two landmark papers demonstrating that grafting human cells could prevent visual loss. As Professor and head of Ocular Biology and Therapeutics, Prof. Coffey has established the London Project to Cure Blindness. This project aims to deliver a stem cell therapy for age-related macular degeneration by 2011.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Macular Degeneration/therapy , Stem Cell Transplantation/methods , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...