Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 592(7853): 195-204, 2021 04.
Article in English | MEDLINE | ID: mdl-33828315

ABSTRACT

The move from reading to writing the human genome offers new opportunities to improve human health. The United States National Institutes of Health (NIH) Somatic Cell Genome Editing (SCGE) Consortium aims to accelerate the development of safer and more-effective methods to edit the genomes of disease-relevant somatic cells in patients, even in tissues that are difficult to reach. Here we discuss the consortium's plans to develop and benchmark approaches to induce and measure genome modifications, and to define downstream functional consequences of genome editing within human cells. Central to this effort is a rigorous and innovative approach that requires validation of the technology through third-party testing in small and large animals. New genome editors, delivery technologies and methods for tracking edited cells in vivo, as well as newly developed animal models and human biological systems, will be assembled-along with validated datasets-into an SCGE Toolkit, which will be disseminated widely to the biomedical research community. We visualize this toolkit-and the knowledge generated by its applications-as a means to accelerate the clinical development of new therapies for a wide range of conditions.


Subject(s)
Cells/metabolism , Gene Editing/methods , Genome, Human/genetics , National Institutes of Health (U.S.)/organization & administration , Animals , Genetic Therapy , Goals , Humans , United States
2.
Hum Mol Genet ; 33(5): 448-464, 2024 Feb 18.
Article in English | MEDLINE | ID: mdl-37975905

ABSTRACT

Biallelic mutations in interphotoreceptor matrix proteoglycan 2 (IMPG2) in humans cause retinitis pigmentosa (RP) with early macular involvement, albeit the disease progression varies widely due to genetic heterogeneity and IMPG2 mutation type. There are currently no treatments for IMPG2-RP. To aid preclinical studies toward eventual treatments, there is a need to better understand the progression of disease pathology in appropriate animal models. Toward this goal, we developed mouse models with patient mimicking homozygous frameshift (T807Ter) or missense (Y250C) Impg2 mutations, as well as mice with a homozygous frameshift mutation (Q244Ter) designed to completely prevent IMPG2 protein expression, and characterized the trajectory of their retinal pathologies across postnatal development until late adulthood. We found that the Impg2T807Ter/T807Ter and Impg2Q244Ter/Q244Ter mice exhibited early onset gliosis, impaired photoreceptor outer segment maintenance, appearance of subretinal deposits near the optic disc, disruption of the outer retina, and neurosensorial detachment, whereas the Impg2Y250C/Y250C mice exhibited minimal retinal pathology. These results demonstrate the importance of mutation type in disease progression in IMPG2-RP and provide a toolkit and preclinical data for advancing therapeutic approaches.


Subject(s)
Proteoglycans , Retinitis Pigmentosa , Humans , Animals , Mice , Adult , Proteoglycans/genetics , Retina , Mutation , Retinitis Pigmentosa/genetics , Disease Progression
3.
Proc Natl Acad Sci U S A ; 120(2): e2213418120, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36598946

ABSTRACT

Human pluripotent stem cell (hPSC)-derived retinal organoids (ROs) can efficiently and reproducibly generate retinal neurons that have potential for use in cell replacement strategies [Capowski et al., Development 146, dev171686 (2019)]. The ability of these lab-grown retinal neurons to form new synaptic connections after dissociation from ROs is key to building confidence in their capacity to restore visual function. However, direct evidence of reestablishment of retinal neuron connectivity via synaptic tracing has not been reported to date. The present study employs an in vitro, rabies virus-based, monosynaptic retrograde tracing assay [Wickersham et al., Neuron 53, 639-647 (2007); Sun et al., Mol. Neurodegener. 14, 8 (2019)] to identify de novo synaptic connections among early retinal cell types following RO dissociation. A reproducible, high-throughput approach for labeling and quantifying traced retinal cell types was developed. Photoreceptors and retinal ganglion cells-the primary neurons of interest for retinal cell replacement-were the two major contributing populations among the traced presynaptic cells. This system provides a platform for assessing synaptic connections in cultured retinal neurons and sets the stage for future cell replacement studies aimed at characterizing or enhancing synaptogenesis. Used in this manner, in vitro synaptic tracing is envisioned to complement traditional preclinical animal model testing, which is limited by evolutionary incompatibilities in synaptic machinery inherent to human xenografts.


Subject(s)
Pluripotent Stem Cells , Retina , Animals , Humans , Reactive Oxygen Species , Retina/physiology , Retinal Ganglion Cells , Organoids , Cell Differentiation
4.
Am J Hum Genet ; 107(2): 278-292, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32707085

ABSTRACT

Dominantly inherited disorders are not typically considered to be therapeutic candidates for gene augmentation. Here, we utilized induced pluripotent stem cell-derived retinal pigment epithelium (iPSC-RPE) to test the potential of gene augmentation to treat Best disease, a dominant macular dystrophy caused by over 200 missense mutations in BEST1. Gene augmentation in iPSC-RPE fully restored BEST1 calcium-activated chloride channel activity and improved rhodopsin degradation in an iPSC-RPE model of recessive bestrophinopathy as well as in two models of dominant Best disease caused by different mutations in regions encoding ion-binding domains. A third dominant Best disease iPSC-RPE model did not respond to gene augmentation, but showed normalization of BEST1 channel activity following CRISPR-Cas9 editing of the mutant allele. We then subjected all three dominant Best disease iPSC-RPE models to gene editing, which produced premature stop codons specifically within the mutant BEST1 alleles. Single-cell profiling demonstrated no adverse perturbation of retinal pigment epithelium (RPE) transcriptional programs in any model, although off-target analysis detected a silent genomic alteration in one model. These results suggest that gene augmentation is a viable first-line approach for some individuals with dominant Best disease and that non-responders are candidates for alternate approaches such as gene editing. However, testing gene editing strategies for on-target efficiency and off-target events using personalized iPSC-RPE model systems is warranted. In summary, personalized iPSC-RPE models can be used to select among a growing list of gene therapy options to maximize safety and efficacy while minimizing time and cost. Similar scenarios likely exist for other genotypically diverse channelopathies, expanding the therapeutic landscape for affected individuals.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Macular Degeneration/genetics , Mutation/genetics , Alleles , Bestrophins/genetics , Calcium/metabolism , Cell Line , Channelopathies/genetics , Eye Proteins/genetics , Gene Editing/methods , Genetic Therapy/methods , Genotype , HEK293 Cells , Humans , Retinal Pigment Epithelium/physiology
5.
Hum Mol Genet ; 29(6): 967-979, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32011687

ABSTRACT

Inherited retinal degenerations (IRDs) are at the focus of current genetic therapeutic advancements. For a genetic treatment such as gene therapy to be successful, an accurate genetic diagnostic is required. Genetic diagnostics relies on the assessment of the probability that a given DNA variant is pathogenic. Non-coding variants present a unique challenge for such assessments as compared to coding variants. For one, non-coding variants are present at much higher number in the genome than coding variants. In addition, our understanding of the rules that govern the non-coding regions of the genome is less complete than our understanding of the coding regions. Methods that allow for both the identification of candidate non-coding pathogenic variants and their functional validation may help overcome these caveats allowing for a greater number of patients to benefit from advancements in genetic therapeutics. We present here an unbiased approach combining whole genome sequencing (WGS) with patient-induced pluripotent stem cell (iPSC)-derived retinal organoids (ROs) transcriptome analysis. With this approach, we identified and functionally validated a novel pathogenic non-coding variant in a small family with a previously unresolved genetic diagnosis.


Subject(s)
Genetic Markers , Genetic Variation , Genome, Human , RNA-Seq/methods , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Whole Genome Sequencing/methods , Child , Female , Gene Expression Profiling , Humans , Male , Pedigree , Exome Sequencing
6.
Am J Hum Genet ; 104(2): 310-318, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30686507

ABSTRACT

Pathogenic variants of the KCNJ13 gene are known to cause Leber congenital amaurosis (LCA16), an inherited pediatric blindness. KCNJ13 encodes the Kir7.1 subunit that acts as a tetrameric, inwardly rectifying potassium ion channel in the retinal pigment epithelium (RPE) to maintain ionic homeostasis and allow photoreceptors to encode visual information. We sought to determine whether genetic approaches might be effective in treating blindness arising from pathogenic variants in KCNJ13. We derived human induced pluripotent stem cell (hiPSC)-RPE cells from an individual carrying a homozygous c.158G>A (p.Trp53∗) pathogenic variant of KCNJ13. We performed biochemical and electrophysiology assays to confirm Kir7.1 function. We tested both small-molecule readthrough drug and gene-therapy approaches for this "disease-in-a-dish" approach. We found that the LCA16 hiPSC-RPE cells had normal morphology but did not express a functional Kir7.1 channel and were unable to demonstrate normal physiology. After readthrough drug treatment, the LCA16 hiPSC cells were hyperpolarized by 30 mV, and the Kir7.1 current was restored. Similarly, we rescued Kir7.1 channel function after lentiviral gene delivery to the hiPSC-RPE cells. In both approaches, Kir7.1 was expressed normally, and there was restoration of membrane potential and the Kir7.1 current. Loss-of-function variants of Kir7.1 are one cause of LCA. Using either readthrough therapy or gene augmentation, we rescued Kir7.1 channel function in iPSC-RPE cells derived from an affected individual. This supports the development of precision-medicine approaches for the treatment of clinical LCA16.


Subject(s)
Blindness/congenital , Channelopathies/genetics , Genetic Therapy/methods , Induced Pluripotent Stem Cells/cytology , Leber Congenital Amaurosis/genetics , Models, Biological , Potassium Channels, Inwardly Rectifying/genetics , Retinal Pigment Epithelium/pathology , Base Sequence , Blindness/genetics , Blindness/pathology , Channelopathies/pathology , Child , Humans , Leber Congenital Amaurosis/pathology , Retinal Pigment Epithelium/metabolism
7.
Development ; 146(1)2019 01 09.
Article in English | MEDLINE | ID: mdl-30567931

ABSTRACT

Numerous protocols have been described for producing neural retina from human pluripotent stem cells (hPSCs), many of which are based on the culture of 3D organoids. Although nearly all such methods yield at least partial segments of retinal structure with a mature appearance, variabilities exist within and between organoids that can change over a protracted time course of differentiation. Adding to this complexity are potential differences in the composition and configuration of retinal organoids when viewed across multiple differentiations and hPSC lines. In an effort to understand better the current capabilities and limitations of these cultures, we generated retinal organoids from 16 hPSC lines and monitored their appearance and structural organization over time by light microscopy, immunocytochemistry, metabolic imaging and electron microscopy. We also employed optical coherence tomography and 3D imaging techniques to assess and compare whole or broad regions of organoids to avoid selection bias. Results from this study led to the development of a practical staging system to reduce inconsistencies in retinal organoid cultures and increase rigor when utilizing them in developmental studies, disease modeling and transplantation.


Subject(s)
Organoids/cytology , Pluripotent Stem Cells/cytology , Retina/cytology , Cell Differentiation , Cell Line , Cell Proliferation , Cell Shape , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Humans , Interneurons/cytology , Interneurons/metabolism , Models, Biological , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/ultrastructure , Reproducibility of Results , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Synapses/metabolism , Tomography, Optical Coherence
8.
EMBO Rep ; 21(9): e50000, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32700445

ABSTRACT

PAX6 is essential for neural retina (NR) and forebrain development but how PAX6 instructs NR versus forebrain specification remains unknown. We found that the paired-less PAX6, PAX6D, is expressed in NR cells during human eye development and along human embryonic stem cell (hESC) specification to retinal cells. hESCs deficient for PAX6D failed to enter NR specification. Induced expression of PAX6D but not PAX6A in a PAX6-null background restored the NR specification capacity. ChIP-Seq, confirmed by functional assays, revealed a set of retinal genes and non-retinal neural genes that are potential targets of PAX6D, including WNT8B. Inhibition of WNTs or knocking down of WNT8B restored the NR specification capacity of neuroepithelia with PAX6D knockout, whereas activation of WNTs blocked NR specification even when PAX6D was induced. Thus, PAX6D specifies neuroepithelia to NR cells via the regulation of WNT8B.


Subject(s)
Human Embryonic Stem Cells , Cell Differentiation , Eye Proteins/genetics , Homeodomain Proteins/genetics , Humans , Neural Plate , Retina , Wnt Proteins/genetics
9.
Proc Natl Acad Sci U S A ; 114(39): E8214-E8223, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28878022

ABSTRACT

Age-related macular degeneration (AMD) and related macular dystrophies (MDs) are a major cause of vision loss. However, the mechanisms underlying their progression remain ill-defined. This is partly due to the lack of disease models recapitulating the human pathology. Furthermore, in vivo studies have yielded limited understanding of the role of specific cell types in the eye vs. systemic influences (e.g., serum) on the disease pathology. Here, we use human induced pluripotent stem cell-retinal pigment epithelium (hiPSC-RPE) derived from patients with three dominant MDs, Sorsby's fundus dystrophy (SFD), Doyne honeycomb retinal dystrophy/malattia Leventinese (DHRD), and autosomal dominant radial drusen (ADRD), and demonstrate that dysfunction of RPE cells alone is sufficient for the initiation of sub-RPE lipoproteinaceous deposit (drusen) formation and extracellular matrix (ECM) alteration in these diseases. Consistent with clinical studies, sub-RPE basal deposits were present beneath both control (unaffected) and patient hiPSC-RPE cells. Importantly basal deposits in patient hiPSC-RPE cultures were more abundant and displayed a lipid- and protein-rich "drusen-like" composition. Furthermore, increased accumulation of COL4 was observed in ECM isolated from control vs. patient hiPSC-RPE cultures. Interestingly, RPE-specific up-regulation in the expression of several complement genes was also seen in patient hiPSC-RPE cultures of all three MDs (SFD, DHRD, and ADRD). Finally, although serum exposure was not necessary for drusen formation, COL4 accumulation in ECM, and complement pathway gene alteration, it impacted the composition of drusen-like deposits in patient hiPSC-RPE cultures. Together, the drusen model(s) of MDs described here provide fundamental insights into the unique biology of maculopathies affecting the RPE-ECM interface.


Subject(s)
Bruch Membrane/pathology , Eye Diseases, Hereditary/pathology , Induced Pluripotent Stem Cells/cytology , Macular Degeneration/pathology , Retinal Drusen/pathology , Retinal Pigment Epithelium/cytology , Blindness/genetics , Blindness/pathology , Cells, Cultured , Collagen Type IV/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Optic Disk Drusen/congenital , Optic Disk Drusen/pathology , Retinal Pigment Epithelium/pathology , Tissue Inhibitor of Metalloproteinase-3/genetics
10.
Am J Hum Genet ; 99(6): 1388-1394, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27889061

ABSTRACT

Human MITF is, by convention, called the "microphthalmia-associated transcription factor" because of previously published seminal mouse genetic studies; however, mutations in MITF have never been associated with microphthalmia in humans. Here, we describe a syndrome that we term COMMAD, characterized by coloboma, osteopetrosis, microphthalmia, macrocephaly, albinism, and deafness. COMMAD is associated with biallelic MITF mutant alleles and hence suggests a role for MITF in regulating processes such as optic-fissure closure and bone development or homeostasis, which go beyond what is usually seen in individuals carrying monoallelic MITF mutations.


Subject(s)
Albinism/genetics , Alleles , Coloboma/genetics , Deafness/genetics , Megalencephaly/genetics , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmos/genetics , Osteopetrosis/genetics , Animals , Child, Preschool , Female , Homozygote , Humans , Infant , Male , Pedigree , Syndrome , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
11.
Stem Cells ; 36(5): 709-722, 2018 05.
Article in English | MEDLINE | ID: mdl-29327488

ABSTRACT

Loss of photoreceptor cells due to retinal degeneration is one of the main causes of blindness in the developed world. Although there is currently no effective treatment, cell replacement therapy using stem-cell-derived photoreceptor cells may be a feasible future treatment option. In order to ensure safety and efficacy of this approach, robust cell isolation and purification protocols must be developed. To this end, we previously developed a biomarker panel for the isolation of mouse photoreceptor precursors from the developing mouse retina and mouse embryonic stem cell cultures. In the current study we applied this approach to the human pluripotent stem cell (hPSC) system, and identified novel biomarker combinations that can be leveraged for the isolation of human photoreceptors. Human retinal samples and hPSC-derived retinal organoid cultures were screened against 242 human monoclonal antibodies using a high through-put flow cytometry approach. We identified 46 biomarkers with significant expression levels in the human retina and hPSC differentiation cultures. Human retinal cell samples, either from fetal tissue or derived from embryonic and induced pluripotent stem cell cultures, were fluorescence-activated cell sorted (FACS) using selected candidate biomarkers that showed expression in discrete cell populations. Enrichment for photoreceptors and exclusion of mitotically active cells was demonstrated by immunocytochemical analysis with photoreceptor-specific antibodies and Ki-67. We established a biomarker combination, which enables the robust purification of viable human photoreceptors from both human retinae and hPSC-derived organoid cultures. Stem Cells 2018;36:709-722.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/cytology , Photoreceptor Cells/cytology , Retinal Degeneration/therapy , Animals , Biomarkers/analysis , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Photoreceptor Cells, Vertebrate/cytology , Pluripotent Stem Cells/cytology , Stem Cell Transplantation/methods
12.
Stem Cells ; 36(3): 313-324, 2018 03.
Article in English | MEDLINE | ID: mdl-29230913

ABSTRACT

Cell type-specific investigations commonly use gene reporters or single-cell analytical techniques. However, reporter line development is arduous and generally limited to a single gene of interest, while single-cell RNA (scRNA)-sequencing (seq) frequently yields equivocal results that preclude definitive cell identification. To examine gene expression profiles of multiple retinal cell types derived from human pluripotent stem cells (hPSCs), we performed scRNA-seq on optic vesicle (OV)-like structures cultured under cGMP-compatible conditions. However, efforts to apply traditional scRNA-seq analytical methods based on unbiased algorithms were unrevealing. Therefore, we developed a simple, versatile, and universally applicable approach that generates gene expression data akin to those obtained from reporter lines. This method ranks single cells by expression level of a bait gene and searches the transcriptome for genes whose cell-to-cell rank order expression most closely matches that of the bait. Moreover, multiple bait genes can be combined to refine datasets. Using this approach, we provide further evidence for the authenticity of hPSC-derived retinal cell types. Stem Cells 2018;36:313-324.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Retina/cytology , Single-Cell Analysis/methods , Gene Expression Profiling , Humans , Sequence Analysis, RNA/methods
13.
Stem Cells ; 34(11): 2625-2634, 2016 11.
Article in English | MEDLINE | ID: mdl-27301076

ABSTRACT

Few gene targets of Visual System Homeobox 2 (VSX2) have been identified despite its broad and critical role in the maintenance of neural retina (NR) fate during early retinogenesis. We performed VSX2 ChIP-seq and ChIP-PCR assays on early stage optic vesicle-like structures (OVs) derived from human iPS cells (hiPSCs), which highlighted WNT pathway genes as direct regulatory targets of VSX2. Examination of early NR patterning in hiPSC-OVs from a patient with a functional null mutation in VSX2 revealed mis-expression and upregulation of WNT pathway components and retinal pigmented epithelium (RPE) markers in comparison to control hiPSC-OVs. Furthermore, pharmacological inhibition of WNT signaling rescued the early mutant phenotype, whereas augmentation of WNT signaling in control hiPSC-OVs phenocopied the mutant. These findings reveal an important role for VSX2 as a regulator of WNT signaling and suggest that VSX2 may act to maintain NR identity at the expense of RPE in part by direct repression of WNT pathway constituents. Stem Cells 2016;34:2625-2634.


Subject(s)
Body Patterning/genetics , Homeodomain Proteins/genetics , Induced Pluripotent Stem Cells/metabolism , Microphthalmos/genetics , Retinal Pigment Epithelium/metabolism , Transcription Factors/genetics , Wnt1 Protein/genetics , Amino Acid Substitution , Benzothiazoles/pharmacology , Biomarkers/metabolism , Cell Differentiation , Embryoid Bodies/drug effects , Embryoid Bodies/metabolism , Embryoid Bodies/pathology , Gene Expression Profiling , Gene Expression Regulation , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Microphthalmos/metabolism , Microphthalmos/pathology , Mutation , Phenotype , Primary Cell Culture , Pyridines/pharmacology , Pyrimidines/pharmacology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects , Wnt1 Protein/agonists , Wnt1 Protein/antagonists & inhibitors , Wnt1 Protein/metabolism
14.
Hum Mol Genet ; 23(23): 6332-44, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25008112

ABSTRACT

Microphthalmia-associated transcription factor (MITF) is a master regulator of pigmented cell survival and differentiation with direct transcriptional links to cell cycle, apoptosis and pigmentation. In mouse, Mitf is expressed early and uniformly in optic vesicle (OV) cells as they evaginate from the developing neural tube, and null Mitf mutations result in microphthalmia and pigmentation defects. However, homozygous mutations in MITF have not been identified in humans; therefore, little is known about its role in human retinogenesis. We used a human embryonic stem cell (hESC) model that recapitulates numerous aspects of retinal development, including OV specification and formation of retinal pigment epithelium (RPE) and neural retina progenitor cells (NRPCs), to investigate the earliest roles of MITF. During hESC differentiation toward a retinal lineage, a subset of MITF isoforms was expressed in a sequence and tissue distribution similar to that observed in mice. In addition, we found that promoters for the MITF-A, -D and -H isoforms were directly targeted by Visual Systems Homeobox 2 (VSX2), a transcription factor involved in patterning the OV toward a NRPC fate. We then manipulated MITF RNA and protein levels at early developmental stages and observed decreased expression of eye field transcription factors, reduced early OV cell proliferation and disrupted RPE maturation. This work provides a foundation for investigating MITF and other highly complex, multi-purposed transcription factors in a dynamic human developmental model system.


Subject(s)
Embryonic Stem Cells/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Neural Stem Cells/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/cytology , Gene Knockout Techniques , Homeodomain Proteins/metabolism , Humans , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Neural Stem Cells/cytology , Promoter Regions, Genetic , Protein Isoforms/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/embryology , Transcription Factors/metabolism
15.
Mol Ther ; 23(11): 1700-1711, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26300224

ABSTRACT

Degradation of photoreceptor outer segments (POS) by retinal pigment epithelium (RPE) is essential for vision, and studies have implicated altered POS processing in the pathogenesis of some retinal degenerative diseases. Consistent with this concept, a recently established hiPSC-RPE model of inherited macular degeneration, Best disease (BD), displayed reduced rates of POS breakdown. Herein we utilized this model to determine (i) if disturbances in protein degradation pathways are associated with delayed POS digestion and (ii) whether such defect(s) can be pharmacologically targeted. We found that BD hiPSC-RPE cultures possessed increased protein oxidation, decreased free-ubiquitin levels, and altered rates of exosome secretion, consistent with altered POS processing. Application of valproic acid (VPA) with or without rapamycin increased rates of POS degradation in our model, whereas application of bafilomycin-A1 decreased such rates. Importantly, the negative effect of bafilomycin-A1 could be fully reversed by VPA. The utility of hiPSC-RPE for VPA testing was further evident following examination of its efficacy and metabolism in a complementary canine disease model. Our findings suggest that disturbances in protein degradation pathways contribute to the POS processing defect observed in BD hiPSC-RPE, which can be manipulated pharmacologically. These results have therapeutic implications for BD and perhaps other maculopathies.


Subject(s)
Enzyme Inhibitors/therapeutic use , Induced Pluripotent Stem Cells/metabolism , Proteolysis/drug effects , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Pigment Epithelium/metabolism , Sirolimus/therapeutic use , Valproic Acid/therapeutic use , Vitelliform Macular Dystrophy/drug therapy , Animals , Autophagy/drug effects , Cells, Cultured , Disease Models, Animal , Dogs , Humans , Macrolides/pharmacology , Models, Biological , Oxidation-Reduction , Primary Cell Culture , Retinal Pigment Epithelium/drug effects
16.
Hum Mol Genet ; 22(3): 593-607, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23139242

ABSTRACT

Best disease (BD) is an inherited degenerative disease of the human macula that results in progressive and irreversible central vision loss. It is caused by mutations in the retinal pigment epithelium (RPE) gene BESTROPHIN1 (BEST1), which, through mechanism(s) that remain unclear, lead to the accumulation of subretinal fluid and autofluorescent waste products from shed photoreceptor outer segments (POSs). We employed human iPS cell (hiPSC) technology to generate RPE from BD patients and unaffected siblings in order to examine the cellular and molecular processes underlying this disease. Consistent with the clinical phenotype of BD, RPE from mutant hiPSCs displayed disrupted fluid flux and increased accrual of autofluorescent material after long-term POS feeding when compared with hiPSC-RPE from unaffected siblings. On a molecular level, RHODOPSIN degradation after POS feeding was delayed in BD hiPSC-RPE relative to unaffected sibling hiPSC-RPE, directly implicating impaired POS handling in the pathophysiology of the disease. In addition, stimulated calcium responses differed between BD and normal sibling hiPSC-RPE, as did oxidative stress levels after chronic POS feeding. Subcellular localization, fractionation and co-immunoprecipitation experiments in hiPSC-RPE and human prenatal RPE further linked BEST1 to the regulation and release of endoplasmic reticulum calcium stores. Since calcium signaling and oxidative stress are critical regulators of fluid flow and protein degradation, these findings likely contribute to the clinical picture of BD. In a larger context, this report demonstrates the potential to use patient-specific hiPSCs to model and study maculopathies, an important class of blinding disorders in humans.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Vitelliform Macular Dystrophy/genetics , Vitelliform Macular Dystrophy/physiopathology , Animals , Bestrophins , Calcium/metabolism , Cattle , Cell Differentiation , Cell Line , Chloride Channels/genetics , Chloride Channels/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Regulation , Homeostasis , Humans , Immunohistochemistry , Immunoprecipitation , Macula Lutea/pathology , Microscopy, Electron, Transmission , Oxidative Stress , Phagocytosis , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/pathology , Vitelliform Macular Dystrophy/metabolism
17.
Stem Cells ; 32(6): 1480-92, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24532057

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) have been shown to differentiate along the retinal lineage in a manner that mimics normal mammalian development. Under certain culture conditions, hiPSCs form optic vesicle-like structures (OVs), which contain proliferating progenitors capable of yielding all neural retina (NR) cell types over time. Such observations imply conserved roles for regulators of retinogenesis in hiPSC-derived cultures and the developing embryo. However, whether and to what extent this assumption holds true has remained largely uninvestigated. We examined the role of a key NR transcription factor, visual system homeobox 2 (VSX2), using hiPSCs derived from a patient with microphthalmia caused by an R200Q mutation in the VSX2 homeodomain region. No differences were noted between (R200Q)VSX2 and sibling control hiPSCs prior to OV generation. Thereafter, (R200Q)VSX2 hiPSC-OVs displayed a significant growth deficit compared to control hiPSC-OVs, as well as increased production of retinal pigmented epithelium at the expense of NR cell derivatives. Furthermore, (R200Q)VSX2 hiPSC-OVs failed to produce bipolar cells, a distinctive feature previously observed in Vsx2 mutant mice. (R200Q)VSX2 hiPSC-OVs also demonstrated delayed photoreceptor maturation, which could be overcome via exogenous expression of wild-type VSX2 at early stages of retinal differentiation. Finally, RNAseq analysis on isolated hiPSC-OVs implicated key transcription factors and extracellular signaling pathways as potential downstream effectors of VSX2-mediated gene regulation. Our results establish hiPSC-OVs as versatile model systems to study retinal development at stages not previously accessible in humans and support the bona fide nature of hiPSC-OV-derived retinal progeny.


Subject(s)
Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Retina/embryology , Retina/metabolism , Transcription Factors/metabolism , Adult , Amino Acid Substitution , Animals , Body Patterning/genetics , Cell Differentiation , Cell Line , Cell Lineage , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Male , Mice , Mutation/genetics , Phenotype , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Retina/pathology , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/pathology , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/pathology , Sequence Analysis, RNA , Signal Transduction/genetics , Transcription Factors/genetics , Transcriptome/genetics
18.
Exp Eye Res ; 123: 161-72, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24534198

ABSTRACT

Human pluripotent stem cells have made a remarkable impact on science, technology and medicine by providing a potentially unlimited source of human cells for basic research and clinical applications. In recent years, knowledge gained from the study of human embryonic stem cells and mammalian somatic cell reprogramming has led to the routine production of human induced pluripotent stem cells (hiPSCs) in laboratories worldwide. hiPSCs show promise for use in transplantation, high throughput drug screening, "disease-in-a-dish" modeling, disease gene discovery, and gene therapy testing. This review will focus on the first application, beginning with a discussion of methods for producing retinal lineage cells that are lost in inherited and acquired forms of retinal degenerative disease. The selection of appropriate hiPSC-derived donor cell type(s) for transplantation will be discussed, as will the caveats and prerequisite steps to formulating a clinical Good Manufacturing Practice (cGMP) product for clinical trials.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Retinal Degeneration/therapy , Stem Cell Transplantation , Cell Lineage , Humans
19.
Proc Natl Acad Sci U S A ; 108(16): 6537-42, 2011 Apr 19.
Article in English | MEDLINE | ID: mdl-21464322

ABSTRACT

Gene-corrected patient-specific induced pluripotent stem (iPS) cells offer a unique approach to gene therapy. Here, we begin to assess whether the mutational load acquired during gene correction of iPS cells is compatible with use in the treatment of genetic causes of retinal degenerative disease. We isolated iPS cells free of transgene sequences from a patient with gyrate atrophy caused by a point mutation in the gene encoding ornithine-δ-aminotransferase (OAT) and used homologous recombination to correct the genetic defect. Cytogenetic analysis, array comparative genomic hybridization (aCGH), and exome sequencing were performed to assess the genomic integrity of an iPS cell line after three sequential clonal events: initial reprogramming, gene targeting, and subsequent removal of a selection cassette. No abnormalities were detected after standard G-band metaphase analysis. However, aCGH and exome sequencing identified two deletions, one amplification, and nine mutations in protein coding regions in the initial iPS cell clone. Except for the targeted correction of the single nucleotide in the OAT locus and a single synonymous base-pair change, no additional mutations or copy number variation were identified in iPS cells after the two subsequent clonal events. These findings confirm that iPS cells themselves may carry a significant mutational load at initial isolation, but that the clonal events and prolonged cultured required for correction of a genetic defect can be accomplished without a substantial increase in mutational burden.


Subject(s)
Gyrate Atrophy/enzymology , Gyrate Atrophy/genetics , Ornithine-Oxo-Acid Transaminase/genetics , Ornithine-Oxo-Acid Transaminase/metabolism , Pluripotent Stem Cells/enzymology , Cells, Cultured , Gene Targeting/methods , Genome-Wide Association Study , Genomic Instability/genetics , Gyrate Atrophy/pathology , Gyrate Atrophy/therapy , Humans , Pluripotent Stem Cells/pathology , Recombination, Genetic
20.
JCI Insight ; 8(11)2023 06 08.
Article in English | MEDLINE | ID: mdl-37288665

ABSTRACT

Over 30 million people worldwide suffer from untreatable vision loss and blindness associated with childhood-onset and age-related eye diseases caused by photoreceptor (PR), retinal pigment epithelium (RPE), and choriocapillaris (CC) degeneration. Recent work suggests that RPE-based cell therapy may slow down vision loss in late stages of age-related macular degeneration (AMD), a polygenic disease induced by RPE atrophy. However, accelerated development of effective cell therapies is hampered by the lack of large-animal models that allow testing safety and efficacy of clinical doses covering the human macula (20 mm2). We developed a versatile pig model to mimic different types and stages of retinal degeneration. Using an adjustable power micropulse laser, we generated varying degrees of RPE, PR, and CC damage and confirmed the damage by longitudinal analysis of clinically relevant outcomes, including analyses by adaptive optics and optical coherence tomography/angiography, along with automated image analysis. By imparting a tunable yet targeted damage to the porcine CC and visual streak - with a structure similar to the human macula - this model is optimal for testing cell and gene therapies for outer retinal diseases including AMD, retinitis pigmentosa, Stargardt, and choroideremia. The amenability of this model to clinically relevant imaging outcomes will facilitate faster translation to patients.


Subject(s)
Macular Degeneration , Retinal Degeneration , Humans , Animals , Swine , Child , Retina/diagnostic imaging , Retinal Degeneration/etiology , Retinal Degeneration/therapy , Retinal Pigment Epithelium , Macular Degeneration/therapy
SELECTION OF CITATIONS
SEARCH DETAIL