Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Eye Res ; 240: 109789, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38242423

ABSTRACT

Age-related macular degeneration (AMD), a leading cause of vision loss, primarily arises from the degeneration of retinal pigment epithelium (RPE) and photoreceptors. Current therapeutic options for dry AMD are limited. Encouragingly, cultured RPE cells on parylene-based biomimetic Bruch's membrane demonstrate characteristics akin to the native RPE layer. In this study, we cultivated human embryonic stem cell-derived polarized RPE (hESC-PRPE) cells on parylene membranes at both small- and large-scale settings, collecting conditioned supernatant, denoted as PRPE-SF. We conducted a comprehensive analysis of the morphology of the cultured hESC-RPE cells and the secreted growth factors in PRPE-SF. To evaluate the in vivo efficacy of these products, the product was administered via intravitreal injections of PRPE-SF in immunodeficient Royal College of Surgeons (iRCS) rats, a model for retinal degeneration. Our study not only demonstrated the scalability of PRPE-SF production while maintaining RPE cell phenotype but also showed consistent protein concentrations between small- and large-scale batches. We consistently identified 10 key factors in PRPE-SF, including BMP-7, IGFBP-2, IGFBP-3, IGFBP-4, IGFBP-6, MANF, PEDF, PDGF-AA, TGFß1, and VEGF. Following intravitreal administration of PRPE-SF, we observed a significant increase in the thickness of the outer nuclear layer (ONL) and photoreceptor preservation in iRCS rats. Furthermore, correlation analysis revealed that IGFBP-3, IGFBP-4, MANF, PEDF, and TGFß1 displayed positive associations with in vivo bioactivity, while GDF-15 exhibited a negative correlation. Overall, this study highlights the feasibility of scaling up PRPE-SF production on parylene membranes without compromising its essential constituents. The outcomes of PRPE-SF administration in an animal model of retinal degeneration present substantial potential for photoreceptor preservation. Moreover, the identification of candidate surrogate potency markers, showing strong positive associations with in vivo bioactivity, lays a solid foundation for the development of a promising therapeutic intervention for retinal degenerative diseases.


Subject(s)
Polymers , Retinal Degeneration , Retinal Pigment Epithelium , Xylenes , Humans , Animals , Rats , Retinal Pigment Epithelium/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 4 , Retinal Degeneration/metabolism
2.
Ophthalmology ; 131(6): 682-691, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38160882

ABSTRACT

PURPOSE: To report long-term results from a phase 1/2a clinical trial assessment of a scaffold-based human embryonic stem cell-derived retinal pigmented epithelium (RPE) implant in patients with advanced geographic atrophy (GA). DESIGN: A single-arm, open-label phase 1/2a clinical trial approved by the United States Food and Drug Administration. PARTICIPANTS: Patients were 69-85 years of age at the time of enrollment and were legally blind in the treated eye (best-corrected visual acuity [BCVA], ≤ 20/200) as a result of GA involving the fovea. METHODS: The clinical trial enrolled 16 patients, 15 of whom underwent implantation successfully. The implant was administered to the worse-seeing eye with the use of a custom subretinal insertion device. The companion nonimplanted eye served as the control. The primary endpoint was at 1 year; thereafter, patients were followed up at least yearly. MAIN OUTCOME MEASURES: Safety was the primary endpoint of the study. The occurrence and frequency of adverse events (AEs) were determined by scheduled eye examinations, including measurement of BCVA and intraocular pressure and multimodal imaging. Serum antibody titers were collected to monitor systemic humoral immune responses to the implanted cells. RESULTS: At a median follow-up of 3 years, fundus photography revealed no migration of the implant. No unanticipated, severe, implant-related AEs occurred, and the most common anticipated severe AE (severe retinal hemorrhage) was eliminated in the second cohort (9 patients) through improved intraoperative hemostasis. Nonsevere, transient retinal hemorrhages were noted either during or after surgery in all patients as anticipated for a subretinal surgical procedure. Throughout the median 3-year follow-up, results show that implanted eyes were more likely to improve by > 5 letters of BCVA and were less likely to worsen by > 5 letters compared with nonimplanted eyes. CONCLUSIONS: This report details the long-term follow-up of patients with GA to receive a scaffold-based stem cell-derived bioengineered RPE implant. Results show that the implant, at a median 3-year follow-up, is safe and well tolerated in patients with advanced dry age-related macular degeneration. The safety profile, along with the early indication of efficacy, warrants further clinical evaluation of this novel approach for the treatment of GA. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.


Subject(s)
Geographic Atrophy , Retinal Pigment Epithelium , Visual Acuity , Humans , Geographic Atrophy/surgery , Geographic Atrophy/physiopathology , Retinal Pigment Epithelium/transplantation , Retinal Pigment Epithelium/pathology , Aged , Visual Acuity/physiology , Female , Aged, 80 and over , Male , Follow-Up Studies , Tomography, Optical Coherence , Human Embryonic Stem Cells/transplantation , Human Embryonic Stem Cells/cytology , Stem Cell Transplantation , Treatment Outcome
3.
Transl Vis Sci Technol ; 10(10): 13, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34613357

ABSTRACT

Purpose: To report 1-year follow-up of a phase 1/2a clinical trial testing a composite subretinal implant having polarized human embryonic stem cell (hESC)-derived retinal pigment epithelium (RPE) cells on an ultrathin parylene substrate in subjects with advanced non-neovascular age-related macular degeneration (NNAMD). Methods: The phase 1/2a clinical trial included 16 subjects in two cohorts. The main endpoint was safety assessed at 365 days using ophthalmic and systemic exams. Pseudophakic subjects with geographic atrophy (GA) and severe vision loss were eligible. Low-dose tacrolimus immunosuppression was utilized for 68 days in the peri-implantation period. The implant was delivered to the worst seeing eye with a custom subretinal insertion device in an outpatient setting. A data safety monitoring committee reviewed all results. Results: The treated eyes of all subjects were legally blind with a baseline best-corrected visual acuity (BCVA) of ≤ 20/200. There were no unexpected serious adverse events. Four subjects in cohort 1 had serious ocular adverse events, including retinal hemorrhage, edema, focal retinal detachment, or RPE detachment, which was mitigated in cohort 2 using improved hemostasis during surgery. Although this study was not powered to assess efficacy, treated eyes from four subjects showed an increased BCVA of >5 letters (6-13 letters). A larger proportion of treated eyes experienced a >5-letter gain when compared with the untreated eye (27% vs. 7%; P = not significant) and a larger proportion of nonimplanted eyes demonstrated a >5-letter loss (47% vs. 33%; P = not significant). Conclusions: Outpatient delivery of the implant can be performed routinely. At 1 year, the implant is safe and well tolerated in subjects with advanced dry AMD. Translational Relevance: This work describes the first clinical trial, to our knowledge, of a novel implant for advanced dry AMD.


Subject(s)
Geographic Atrophy , Hematopoietic Stem Cell Transplantation , Macular Degeneration , Follow-Up Studies , Geographic Atrophy/therapy , Humans , Macular Degeneration/therapy , Visual Acuity
4.
Sci Rep ; 11(1): 6286, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737600

ABSTRACT

Age-related macular degeneration (AMD) is the primary cause of blindness in adults over 60 years of age, and clinical trials are currently assessing the therapeutic potential of retinal pigmented epithelial (RPE) cell monolayers on implantable scaffolds to treat this disease. However, challenges related to the culture, long-term storage, and long-distance transport of such implants currently limit the widespread use of adherent RPE cells as therapeutics. Here we report a xeno-free protocol to cryopreserve a confluent monolayer of clinical-grade, human embryonic stem cell-derived RPE cells on a parylene scaffold (REPS) that yields viable, polarized, and functional RPE cells post-thaw. Thawed cells exhibit ≥ 95% viability, have morphology, pigmentation, and gene expression characteristic of mature RPE cells, and secrete the neuroprotective protein, pigment epithelium-derived factor (PEDF). Stability under liquid nitrogen (LN2) storage has been confirmed through one year. REPS were administered immediately post-thaw into the subretinal space of a mammalian model, the Royal College of Surgeons (RCS)/nude rat. Implanted REPS were assessed at 30, 60, and 90 days post-implantation, and thawed cells demonstrate survival as an intact monolayer on the parylene scaffold. Furthermore, immunoreactivity for the maturation marker, RPE65, significantly increased over the post-implantation period in vivo, and cells demonstrated functional attributes similar to non-cryopreserved controls. The capacity to cryopreserve adherent cellular therapeutics permits extended storage and stable transport to surgical sites, enabling broad distribution for the treatment of prevalent diseases such as AMD.


Subject(s)
Cryopreservation/methods , Epithelial Cells/transplantation , Macular Degeneration/therapy , Retinal Pigment Epithelium/transplantation , Specimen Handling/methods , Stem Cell Transplantation/methods , Animals , Cell Differentiation , Cell Line , Cell Survival , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Eye Proteins/metabolism , Human Embryonic Stem Cells/cytology , Humans , Nerve Growth Factors/metabolism , Polymers , Rats , Rats, Nude , Regenerative Medicine/methods , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Serpins/metabolism , Tissue Scaffolds , Treatment Outcome , Xylenes
5.
Sci Transl Med ; 11(485)2019 03 27.
Article in English | MEDLINE | ID: mdl-30918111

ABSTRACT

Tau inclusions are a shared feature of many neurodegenerative diseases, among them frontotemporal dementia caused by tau mutations. Treatment approaches for these conditions include targeting posttranslational modifications of tau proteins, maintaining a steady-state amount of tau, and preventing its tendency to aggregate. We discovered a new regulatory pathway for tau degradation that operates through the farnesylated protein, Rhes, a GTPase in the Ras family. Here, we show that treatment with the farnesyltransferase inhibitor lonafarnib reduced Rhes and decreased brain atrophy, tau inclusions, tau sumoylation, and tau ubiquitination in the rTg4510 mouse model of tauopathy. In addition, lonafarnib treatment attenuated behavioral abnormalities in rTg4510 mice and reduced microgliosis in mouse brain. Direct reduction of Rhes in the rTg4510 mouse by siRNA reproduced the results observed with lonafarnib treatment. The mechanism of lonafarnib action mediated by Rhes to reduce tau pathology was shown to operate through activation of lysosomes. We finally showed in mouse brain and in human induced pluripotent stem cell-derived neurons a normal developmental increase in Rhes that was initially suppressed by tau mutations. The known safety of lonafarnib revealed in human clinical trials for cancer suggests that this drug could be repurposed for treating tauopathies.


Subject(s)
Farnesyltranstransferase/antagonists & inhibitors , Tauopathies/drug therapy , Tauopathies/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice , Mice, Transgenic , Mutation , Neurons/drug effects , Neurons/metabolism , Piperidines/pharmacology , Proteolysis/drug effects , Pyridines/pharmacology , RNA, Small Interfering/genetics , Tauopathies/pathology , Translational Research, Biomedical , tau Proteins/genetics , tau Proteins/metabolism
6.
Tissue Eng Part A ; 22(7-8): 597-609, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26956095

ABSTRACT

Soft tissue defects are relatively common, yet currently used reconstructive treatments have varying success rates, and serious potential complications such as unpredictable volume loss and reabsorption. Human adipose-derived stem cells (ASCs), isolated from liposuction aspirate have great potential for use in soft tissue regeneration, especially when combined with a supportive scaffold. To design scaffolds that promote differentiation of these cells down an adipogenic lineage, we characterized changes in the surrounding extracellular environment during adipogenic differentiation. We found expression changes in both extracellular matrix proteins, including increases in expression of collagen-IV and vitronectin, as well as changes in the integrin expression profile, with an increase in expression of integrins such as αVß5 and α1ß1. These integrins are known to specifically interact with vitronectin and collagen-IV, respectively, through binding to an Arg-Gly-Asp (RGD) sequence. When three different short RGD-containing peptides were incorporated into three-dimensional (3D) hydrogel cultures, it was found that an RGD-containing peptide derived from vitronectin provided strong initial attachment, maintained the desired morphology, and created optimal conditions for in vitro 3D adipogenic differentiation of ASCs. These results describe a simple, nontoxic encapsulating scaffold, capable of supporting the survival and desired differentiation of ASCs for the treatment of soft tissue defects.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue/cytology , Biomimetic Materials/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Stem Cells/cytology , Tissue Scaffolds/chemistry , Vitronectin/pharmacology , Amino Acid Sequence , Cell Adhesion/drug effects , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Integrins/metabolism , Oligopeptides/chemistry , Oligopeptides/pharmacology , Polyethylene Glycols/chemistry , Stem Cells/drug effects
7.
Neuron ; 83(1): 69-86, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-24991955

ABSTRACT

Neural stem cells have been adopted to model a wide range of neuropsychiatric conditions in vitro. However, how well such models correspond to in vivo brain has not been evaluated in an unbiased, comprehensive manner. We used transcriptomic analyses to compare in vitro systems to developing human fetal brain and observed strong conservation of in vivo gene expression and network architecture in differentiating primary human neural progenitor cells (phNPCs). Conserved modules are enriched in genes associated with ASD, supporting the utility of phNPCs for studying neuropsychiatric disease. We also developed and validated a machine learning approach called CoNTExT that identifies the developmental maturity and regional identity of in vitro models. We observed strong differences between in vitro models, including hiPSC-derived neural progenitors from multiple laboratories. This work provides a systems biology framework for evaluating in vitro systems and supports their value in studying the molecular mechanisms of human neurodevelopmental disease.


Subject(s)
Artificial Intelligence , Cerebral Cortex/embryology , Embryonic Stem Cells/physiology , Gene Regulatory Networks/genetics , Models, Neurological , Neural Stem Cells/physiology , Artificial Intelligence/trends , Cells, Cultured , Cerebral Cortex/cytology , Female , Humans , Male
8.
Stem Cells Transl Med ; 2(5): 384-93, 2013 May.
Article in English | MEDLINE | ID: mdl-23599499

ABSTRACT

Controlling the differentiation of human pluripotent stem cells is the goal of many laboratories, both to study normal human development and to generate cells for transplantation. One important cell type under investigation is the retinal pigmented epithelium (RPE). Age-related macular degeneration (AMD), the leading cause of blindness in the Western world, is caused by dysfunction and death of the RPE. Currently, RPE derived from human embryonic stem cells are in clinical trials for the treatment of AMD. Although protocols to generate RPE from human pluripotent stem cells have become more efficient since the first report in 2004, they are still time-consuming and relatively inefficient. We have found that the addition of defined factors at specific times leads to conversion of approximately 80% of the cells to an RPE phenotype in only 14 days. This protocol should be useful for rapidly generating RPE for transplantation as well as for studying RPE development in vitro.


Subject(s)
Cell Differentiation , Pluripotent Stem Cells/cytology , Retinal Pigment Epithelium/cytology , Activins/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Niacinamide/pharmacology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Pyrroles/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Time Factors , Vasoactive Intestinal Peptide/pharmacology , Visual Fields/drug effects
9.
Stem Cells ; 27(10): 2427-34, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19658190

ABSTRACT

Human induced pluripotent stem cells (iPSCs) have great promise for cellular therapy, but it is unclear if they have the same potential as human embryonic stem cells (hESCs) to differentiate into specialized cell types. Ocular cells such as the retinal pigmented epithelium (RPE) are of particular interest because they could be used to treat degenerative eye diseases, including age-related macular degeneration and retinitis pigmentosa. We show here that iPSCs generated using Oct4, Sox2, Nanog, and Lin28 can spontaneously differentiate into RPE cells, which can then be isolated and cultured to form highly differentiated RPE monolayers. RPE derived from iPSCs (iPS-RPE) were analyzed with respect to gene expression, protein expression, and rod outer segment phagocytosis, and compared with cultured fetal human RPE (fRPE) and RPE derived from hESCs (hESC-RPE). iPS-RPE expression of marker mRNAs was quantitatively similar to that of fRPE and hESC-RPE, and marker proteins were appropriately expressed and localized in polarized monolayers. Levels of rod outer segment phagocytosis by iPS-RPE, fRPE, and hESC-RPE were likewise similar and dependent on integrin alpha v beta 5. This work shows that iPSCs can differentiate into functional RPE that are quantitatively similar to fRPE and hESC-RPE and further supports the finding that iPSCs are similar to hESCs in their differentiation potential.


Subject(s)
Cell Differentiation/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Brain Tissue Transplantation/methods , Cell Differentiation/drug effects , Cell Line , Cell Polarity/physiology , Cell Proliferation , Homeodomain Proteins/metabolism , Homeodomain Proteins/pharmacology , Humans , Integrin alphaV/metabolism , Nanog Homeobox Protein , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/pharmacology , Phagocytosis/physiology , Phenotype , Pluripotent Stem Cells/drug effects , RNA, Messenger/analysis , RNA, Messenger/metabolism , Regeneration/drug effects , Regeneration/physiology , Retinal Diseases/therapy , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/metabolism , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...