Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
CRISPR J ; 6(6): 502-513, 2023 12.
Article in English | MEDLINE | ID: mdl-38108516

ABSTRACT

Rhodopsin (RHO) mutations such as Pro23His are the leading cause of dominantly inherited retinitis pigmentosa in North America. As with other dominant retinal dystrophies, these mutations lead to production of a toxic protein product, and treatment will require knockdown of the mutant allele. The purpose of this study was to develop a CRISPR-Cas9-mediated transcriptional repression strategy using catalytically inactive Staphylococcus aureus Cas9 (dCas9) fused to the Krüppel-associated box (KRAB) transcriptional repressor domain. Using a reporter construct carrying green fluorescent protein (GFP) cloned downstream of the RHO promoter fragment (nucleotides -1403 to +73), we demonstrate a ∼74-84% reduction in RHO promoter activity in RHOpCRISPRi-treated versus plasmid-only controls. After subretinal transduction of human retinal explants and transgenic Pro23His mutant pigs, significant knockdown of rhodopsin protein was achieved. Suppression of mutant transgene in vivo was associated with a reduction in endoplasmic reticulum (ER) stress and apoptosis markers and preservation of photoreceptor cell layer thickness.


Subject(s)
Retinitis Pigmentosa , Rhodopsin , Humans , Animals , Swine , Rhodopsin/genetics , CRISPR-Cas Systems/genetics , Gene Editing , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Alleles
2.
Gene Ther ; 29(5): 259-288, 2022 05.
Article in English | MEDLINE | ID: mdl-34518651

ABSTRACT

By combining next generation whole exome sequencing and induced pluripotent stem cell (iPSC) technology we found that an Alu repeat inserted in exon 9 of the MAK gene results in a loss of normal MAK transcript and development of human autosomal recessive retinitis pigmentosa (RP). Although a relatively rare cause of disease in the general population, the MAK variant is enriched in individuals of Jewish ancestry. In this population, 1 in 55 individuals are carriers and one third of all cases of recessive RP is caused by this gene. The purpose of this study was to determine if a viral gene augmentation strategy could be used to safely restore functional MAK protein as a step toward a treatment for early stage MAK-associated RP. Patient iPSC-derived photoreceptor precursor cells were generated and transduced with viral vectors containing the MAK transcript. One week after transduction, transcript and protein could be detected via rt-PCR and western blotting respectively. Using patient-derived fibroblast cells and mak knockdown zebra fish we demonstrate that over-expression of the retinal MAK transgene restored the cells ability to regulate primary cilia length. In addition, the visual defect in mak knockdown zebrafish was mitigated via treatment with the retinal MAK transgene. There was no evidence of local or systemic toxicity at 1-month or 3-months following subretinal delivery of clinical grade vector into wild type rats. The findings reported here will help pave the way for initiation of a phase 1 clinical trial for the treatment of patients with MAK-associated RP.


Subject(s)
Retinitis Pigmentosa , Zebrafish , Animals , Exons , Genetic Therapy , Humans , Mutation , Rats , Retina , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Zebrafish/genetics
3.
Stem Cells Transl Med ; 8(8): 797-809, 2019 08.
Article in English | MEDLINE | ID: mdl-31004408

ABSTRACT

Subretinal delivery of stem cell-derived retinal cells as a strategy to treat retinal degenerative blindness holds great promise. Currently, two clinical trials are underway in which human fetal retinal progenitor cells (RPCs) are being delivered to patients by intravitreal or subretinal injection to preserve or restore vision, respectively. With the advent of the induced pluripotent stem cell (iPSC), and in turn three-dimensional derivation of retinal tissue, it is now possible to generate autologous RPCs for cell replacement. The purpose of this study was to evaluate the effect of commonly used cell isolation and surgical manipulation strategies on donor cell viability. iPSC-RPCs were subjected to various conditions, including different dissociation and isolation methods, injection cannula sizes, and preinjection storage temperatures and times. The effects of commonly used surgical techniques on both host and donor cell viability were evaluated in Yucatan mini-pigs (n = 61 eyes). We found a significant increase in cell viability when papain was used for RPC isolation. In addition, a significant decrease in cell viability was detected when using the 41G cannula compared with 31G and at storage times of 4 hours compared with 30 minutes. Although 96.4% of all eyes demonstrated spontaneous retinal reattachment following injection, retinal pigment epithelium (RPE) abnormalities were seen more frequently in eyes receiving injections via a 31G cannula; interestingly, eyes that received cell suspensions were relatively protected against such RPE changes. These findings indicate that optimization of donor cell isolation and delivery parameters should be considered when developing a subretinal cell replacement strategy. Stem Cells Translational Medicine 2019;8:797&809.


Subject(s)
Cellular Reprogramming Techniques/methods , Neural Stem Cells/transplantation , Primary Cell Culture/methods , Retina/cytology , Retinal Dystrophies/therapy , Stem Cell Transplantation/methods , Animals , Cell Survival , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Neural Stem Cells/cytology , Papain/pharmacology , Primary Cell Culture/standards , Retina/drug effects , Stem Cell Transplantation/adverse effects , Swine , Swine, Miniature
4.
Curr Protoc Stem Cell Biol ; 42: 4A.12.1-4A.12.14, 2017 Aug 14.
Article in English | MEDLINE | ID: mdl-28806854

ABSTRACT

This unit describes protocols for the generation of clinical-grade patient-specific induced pluripotent stem cell (iPSC)-derived retinal cells from patients with inherited retinal degenerative blindness. Specifically, we describe how, using xeno-free reagents in an ISO class 5 environment, one can isolate and culture dermal fibroblasts, generate iPSCs, and derive autologous retinal cells via 3-D differentiation. The universal methods described herein for the isolation of dermal fibroblasts and generation of iPSCs can be employed regardless of disease, tissue, or cell type of interest. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming Techniques/methods , Dermis , Fibroblasts , Induced Pluripotent Stem Cells , Retina , Biopsy , Dermis/metabolism , Dermis/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Retina/metabolism , Retina/pathology
5.
Stem Cells Transl Med ; 6(6): 1533-1546, 2017 06.
Article in English | MEDLINE | ID: mdl-28474838

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of irreversible blindness in the Western world. Although, the majority of stem cell research to date has focused on production of retinal pigment epithelial (RPE) and photoreceptor cells for the purpose of evaluating disease pathophysiology and cell replacement, there is strong evidence that the choroidal endothelial cells (CECs) that form the choriocapillaris vessels are the first to be lost in this disease. As such, to accurately evaluate disease pathophysiology and develop an effective treatment, production of patient-specific, stem cell-derived CECs will be required. In this study, we report for the first time a stepwise differentiation protocol suitable for generating human iPSC-derived CEC-like cells. RNA-seq analysis of the monkey CEC line, RF/6A, combined with two statistical screens allowed us to develop media comprised of various protein combinations. In both screens, connective tissue growth factor (CTGF) was identified as the key component required for driving CEC development. A second factor tumor necrosis factor (TNF)-related weak inducer of apoptosis receptor was also found to promote iPSC to CEC differentiation by inducing endogenous CTGF secretion. CTGF-driven iPSC-derived CEC-like cells formed capillary tube-like vascular networks, and expressed the EC-specific markers CD31, ICAM1, PLVAP, vWF, and the CEC-restricted marker CA4. In combination with RPE and photoreceptor cells, patient-specific iPSC derived CEC-like cells will enable scientists to accurately evaluate AMD pathophysiology and develop effective cell replacement therapies. Stem Cells Translational Medicine 2017;6:1533-1546.


Subject(s)
Choroid/cytology , Endothelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Macular Degeneration/therapy , Animals , Cell Differentiation , Cells, Cultured , Cellular Reprogramming Techniques/methods , Choroid/physiology , Connective Tissue Growth Factor/pharmacology , Endothelial Cells/drug effects , Endothelium/cytology , Endothelium/physiology , Haplorhini , Humans , Induced Pluripotent Stem Cells/drug effects , Regeneration , Stem Cell Transplantation/methods
6.
Stem Cell Res ; 21: 58-70, 2017 05.
Article in English | MEDLINE | ID: mdl-28390992

ABSTRACT

Retinitis pigmentosa (RP) is a heterogeneous group of monogenic disorders characterized by progressive death of the light-sensing photoreceptor cells of the outer neural retina. We recently identified novel hypomorphic mutations in the tRNA Nucleotidyl Transferase, CCA-Adding 1 (TRNT1) gene that cause early-onset RP. To model this disease in vitro, we generated patient-specific iPSCs and iPSC-derived retinal organoids from dermal fibroblasts of patients with molecularly confirmed TRNT1-associated RP. Pluripotency was confirmed using rt-PCR, immunocytochemistry, and a TaqMan Scorecard Assay. Mutations in TRNT1 caused reduced levels of full-length TRNT1 protein and expression of a truncated smaller protein in both patient-specific iPSCs and iPSC-derived retinal organoids. Patient-specific iPSCs and iPSC-derived retinal organoids exhibited a deficit in autophagy, as evidenced by aberrant accumulation of LC3-II and elevated levels of oxidative stress. Autologous stem cell-based disease modeling will provide a platform for testing multiple avenues of treatment in patients suffering from TRNT1-associated RP.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Nucleotidyltransferases/metabolism , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/physiopathology , Autophagosomes/metabolism , Autophagy , Humans , Induced Pluripotent Stem Cells/metabolism , Lysosomal Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Organoids/metabolism , Oxidative Stress , Retina/metabolism
7.
Hum Gene Ther ; 27(10): 835-846, 2016 10.
Article in English | MEDLINE | ID: mdl-27400765

ABSTRACT

Juvenile neuronal ceroid lipofuscinosis (JNCL) is a childhood neurodegenerative disease with early-onset, severe central vision loss. Affected children develop seizures and CNS degeneration accompanied by severe motor and cognitive deficits. There is no cure for JNCL, and patients usually die during the second or third decade of life. In this study, independent lines of induced pluripotent stem cells (iPSCs) were generated from two patients with molecularly confirmed mutations in CLN3, the gene mutated in JNCL. Clinical-grade adeno-associated adenovirus serotype 2 (AAV2) carrying the full-length coding sequence of human CLN3 was generated in a U.S. Food and Drug Administration-registered cGMP facility. AAV2-CLN3 was efficacious in restoring full-length CLN3 transcript and protein in patient-specific fibroblasts and iPSC-derived retinal neurons. When injected into the subretinal space of wild-type mice, purified AAV2-CLN3 did not show any evidence of retinal toxicity. This study provides proof-of-principle for initiation of a clinical trial using AAV-mediated gene augmentation for the treatment of children with CLN3-associated retinal degeneration.


Subject(s)
Genetic Therapy/methods , Induced Pluripotent Stem Cells/transplantation , Membrane Glycoproteins/genetics , Molecular Chaperones/genetics , Neuronal Ceroid-Lipofuscinoses/therapy , Retinal Degeneration/therapy , Animals , Dependovirus/genetics , Fibroblasts/metabolism , Humans , Membrane Glycoproteins/therapeutic use , Mice , Molecular Chaperones/therapeutic use , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/metabolism , Retina/metabolism , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology
8.
Sci Rep ; 6: 30742, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27471043

ABSTRACT

Immunologically-matched, induced pluripotent stem cell (iPSC)-derived photoreceptor precursor cells have the potential to restore vision to patients with retinal degenerative diseases like retinitis pigmentosa. The purpose of this study was to develop clinically-compatible methods for manufacturing photoreceptor precursor cells from adult skin in a non-profit cGMP environment. Biopsies were obtained from 35 adult patients with inherited retinal degeneration and fibroblast lines were established under ISO class 5 cGMP conditions. Patient-specific iPSCs were then generated, clonally expanded and validated. Post-mitotic photoreceptor precursor cells were generated using a stepwise cGMP-compliant 3D differentiation protocol. The recapitulation of the enhanced S-cone phenotype in retinal organoids generated from a patient with NR2E3 mutations demonstrated the fidelity of these protocols. Transplantation into immune compromised animals revealed no evidence of abnormal proliferation or tumor formation. These studies will enable clinical trials to test the safety and efficiency of patient-specific photoreceptor cell replacement in humans.


Subject(s)
Blindness/pathology , Cyclic GMP/metabolism , Induced Pluripotent Stem Cells/cytology , Organoids/metabolism , Retinal Cone Photoreceptor Cells/cytology , Retinal Degeneration/pathology , Adult , Animals , Blindness/etiology , Blindness/therapy , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mutation , Organoids/transplantation , Orphan Nuclear Receptors/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/congenital , Retinal Degeneration/therapy , Skin/cytology , Skin/metabolism , Transplantation, Autologous
9.
Transl Res ; 166(6): 740-749.e1, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26364624

ABSTRACT

Retinal pigment epithelium-specific 65 kDa (RPE65)-associated Leber congenital amaurosis is an autosomal recessive disease that results in reduced visual acuity and night blindness beginning at birth. It is one of the few retinal degenerative disorders for which promising clinical gene transfer trials are currently underway. However, the ability to enroll patients in a gene augmentation trial is dependent on the identification of 2 bona fide disease-causing mutations, and there are some patients with the phenotype of RPE65-associated disease who might benefit from gene transfer but are ineligible because 2 disease-causing genetic variations have not yet been identified. Some such patients have novel mutations in RPE65 for which pathogenicity is difficult to confirm. The goal of this study was to determine if an intronic mutation identified in a 2-year-old patient with presumed RPE65-associated disease was truly pathogenic and grounds for inclusion in a clinical gene augmentation trial. Sequencing of the RPE65 gene revealed 2 mutations: (1) a previously identified disease-causing exonic leucine-to-proline mutation (L408P) and (2) a novel single point mutation in intron 3 (IVS3-11) resulting in an A>G change. RT-PCR analysis using RNA extracted from control human donor eye-derived primary RPE, control iPSC-RPE cells, and proband iPSC-RPE cells revealed that the identified IVS3-11 variation caused a splicing defect that resulted in a frameshift and insertion of a premature stop codon. In this study, we demonstrate how patient-specific iPSCs can be used to confirm pathogenicity of unknown mutations, which can enable positive clinical outcomes.


Subject(s)
Mutation , Pluripotent Stem Cells/cytology , Retinal Pigment Epithelium/metabolism , cis-trans-Isomerases/genetics , Cell Differentiation , Child, Preschool , Humans
10.
Invest Ophthalmol Vis Sci ; 56(13): 8258-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26720480

ABSTRACT

PURPOSE: Age-related macular degeneration (AMD), the most common cause of incurable blindness in the western world, is characterized by the dysfunction and eventual death of choroidal endothelial (CECs), RPE, and photoreceptor cells. Stem cell-based treatment strategies designed to replace photoreceptor and RPE cells currently are a major scientific focus. However, the success of these approaches likely also will require replacement of the underlying, supportive choroidal vasculature. The purpose of this study was to generate stem cell-derived CECs to develop efficient differentiation and transplantation protocols. METHODS: Dermal fibroblasts from the Tie2-GFP mouse were isolated and reprogrammed into two independent induced pluripotent stem cell (iPSC) lines via viral transduction of the transcription factors Oct4, Sox2, Klf4, and c-Myc. Tie2-GFP iPSCs were differentiated into CECs using a coculture method with either the RF6A CEC line or primary mouse CECs. Induced pluripotent stem cell-derived CECs were characterized via RT-PCR and immunocytochemistry for EC- and CEC-specific markers. RESULTS: Induced pluripotent stem cells generated from mice expressing green fluorescent protein (GFP) under control of the endothelial Tie2 promoter display classic pluripotency markers and stem cell morphology. Induced pluripotent stem cell-derived CECs express carbonic anhydrase IV, eNOS, FOXA2, PLVAP, CD31, CD34, ICAM-1, Tie2, TTR, VE-cadherin, and vWF. CONCLUSIONS: Induced pluripotent stem cell-derived CECs will be a valuable tool for modeling of choriocapillaris-specific insults in AMD and for use in future choroidal endothelial cell replacement approaches.


Subject(s)
Choroid/pathology , Endothelial Cells/pathology , Induced Pluripotent Stem Cells/cytology , Macular Degeneration/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cells, Cultured , Endothelial Cells/metabolism , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Macular Degeneration/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...