Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cells Transl Med ; 12(6): 365-378, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37221451

RESUMO

Prior to use, newly generated induced pluripotent stem cells (iPSC) should be thoroughly validated. While excellent validation and release testing assays designed to evaluate potency, genetic integrity, and sterility exist, they do not have the ability to predict cell type-specific differentiation capacity. Selection of iPSC lines that have limited capacity to produce high-quality transplantable cells, places significant strain on valuable clinical manufacturing resources. The purpose of this study was to determine the degree and root cause of variability in retinal differentiation capacity between cGMP-derived patient iPSC lines. In turn, our goal was to develop a release testing assay that could be used to augment the widely used ScoreCard panel. IPSCs were generated from 15 patients (14-76 years old), differentiated into retinal organoids, and scored based on their retinal differentiation capacity. Despite significant differences in retinal differentiation propensity, RNA-sequencing revealed remarkable similarity between patient-derived iPSC lines prior to differentiation. At 7 days of differentiation, significant differences in gene expression could be detected. Ingenuity pathway analysis revealed perturbations in pathways associated with pluripotency and early cell fate commitment. For example, good and poor producers had noticeably different expressions of OCT4 and SOX2 effector genes. QPCR assays targeting genes identified via RNA sequencing were developed and validated in a masked fashion using iPSCs from 8 independent patients. A subset of 14 genes, which include the retinal cell fate markers RAX, LHX2, VSX2, and SIX6 (all elevated in the good producers), were found to be predictive of retinal differentiation propensity.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Adolescente , Adulto Jovem , Adulto , Pessoa de Meia-Idade , Idoso , Diferenciação Celular , Retina , Organoides
2.
Prog Retin Eye Res ; 83: 100918, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33130253

RESUMO

Our understanding of inherited retinal disease has benefited immensely from molecular genetic analysis over the past several decades. New technologies that allow for increasingly detailed examination of a patient's DNA have expanded the catalog of genes and specific variants that cause retinal disease. In turn, the identification of pathogenic variants has allowed the development of gene therapies and low-cost, clinically focused genetic testing. Despite this progress, a relatively large fraction (at least 20%) of patients with clinical features suggestive of an inherited retinal disease still do not have a molecular diagnosis today. Variants that are not obviously disruptive to the codon sequence of exons can be difficult to distinguish from the background of benign human genetic variations. Some of these variants exert their pathogenic effect not by altering the primary amino acid sequence, but by modulating gene expression, isoform splicing, or other transcript-level mechanisms. While not discoverable by DNA sequencing methods alone, these variants are excellent targets for studies of the retinal transcriptome. In this review, we present an overview of the current state of pathogenic variant discovery in retinal disease and identify some of the remaining barriers. We also explore the utility of new technologies, specifically patient-derived induced pluripotent stem cell (iPSC)-based modeling, in further expanding the catalog of disease-causing variants using transcriptome-focused methods. Finally, we outline bioinformatic analysis techniques that will allow this new method of variant discovery in retinal disease. As the knowledge gleaned from previous technologies is informing targets for therapies today, we believe that integrating new technologies, such as iPSC-based modeling, into the molecular diagnosis pipeline will enable a new wave of variant discovery and expanded treatment of inherited retinal disease.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças Retinianas , DNA , Éxons , Humanos , Retina , Doenças Retinianas/genética
3.
Exp Eye Res ; 199: 108166, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32771499

RESUMO

Inherited retinal degenerative disorders such as retinitis pigmentosa and Usher syndrome are characterized by progressive death of photoreceptor cells. To restore vision to patients blinded by these diseases, a stem cell-based photoreceptor cell replacement strategy will likely be required. Although retinal stem cell differentiation protocols suitable for generating photoreceptor cells exist, they often yield a rather heterogenous mixture of cell types. To enrich the donor cell population for one or a few cell types, scientists have traditionally relied upon the use of antibody-based selection approaches. However, these strategies are quite labor intensive and require animal derived reagents and equipment that are not well suited to current good manufacturing practices (cGMP). The purpose of this study was to develop and evaluate a microfluidic cell sorting device capable of exploiting the physical and mechanical differences between retinal cell types to enrich specific donor cell populations such as Retinal Pigment Epithelial (RPE) cells and photoreceptor cells. Using this device, we were able to separate a mixture of RPE and iPSC-derived photoreceptor precursor cell lines into two substantially enriched fractions. The enrichment factor of the RPE fraction was 2 and that of the photoreceptor precursor cell fraction was 2.7. Similarly, when human retina, obtained from 3 independent donors, was dissociated and passed through the sorting device, the heterogeneous mixture could be reliably sorted into RPE and photoreceptor cell rich fractions. In summary, microfluidic cell sorting is a promising approach for antibody free enrichment of retinal cell populations.


Assuntos
Microfluídica/métodos , Células Fotorreceptoras/patologia , Degeneração Retiniana/diagnóstico , Epitélio Pigmentado da Retina/patologia , Animais , Diferenciação Celular , Linhagem Celular , Humanos , Microscopia de Força Atômica , Células Fotorreceptoras/metabolismo , Degeneração Retiniana/metabolismo , Epitélio Pigmentado da Retina/metabolismo
4.
Genes (Basel) ; 10(4)2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30959774

RESUMO

Enhanced S-cone syndrome (ESCS) is caused by recessive mutations in the photoreceptor cell transcription factor NR2E3. Loss of NR2E3 is characterized by repression of rod photoreceptor cell gene expression, over-expansion of the S-cone photoreceptor cell population, and varying degrees of M- and L-cone photoreceptor cell development. In this study, we developed a CRISPR-based homology-directed repair strategy and corrected two different disease-causing NR2E3 mutations in patient-derived induced pluripotent stem cells (iPSCs) generated from two affected individuals. In addition, one patient's iPSCs were differentiated into retinal cells and NR2E3 transcription was evaluated in CRISPR corrected and uncorrected clones. The patient's c.119-2A>C mutation caused the inclusion of a portion of intron 1, the creation of a frame shift, and generation of a premature stop codon. In summary, we used a single set of CRISPR reagents to correct different mutations in iPSCs generated from two individuals with ESCS. In doing so we demonstrate the advantage of using retinal cells derived from affected patients over artificial in vitro model systems when attempting to demonstrate pathophysiologic mechanisms of specific mutations.


Assuntos
Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/terapia , Terapia Genética , Receptores Nucleares Órfãos/genética , Degeneração Retiniana/genética , Degeneração Retiniana/terapia , Transtornos da Visão/genética , Transtornos da Visão/terapia , Sistemas CRISPR-Cas/genética , Diferenciação Celular/genética , Códon sem Sentido/genética , Oftalmopatias Hereditárias/patologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Predisposição Genética para Doença , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Mutação , Receptores Nucleares Órfãos/uso terapêutico , Degeneração Retiniana/patologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Transtornos da Visão/patologia
5.
Prog Retin Eye Res ; 65: 28-49, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29578069

RESUMO

Gene correction is a valuable strategy for treating inherited retinal degenerative diseases, a major cause of irreversible blindness worldwide. Single gene defects cause the majority of these retinal dystrophies. Gene augmentation holds great promise if delivered early in the course of the disease, however, many patients carry mutations in genes too large to be packaged into adeno-associated viral vectors and some, when overexpressed via heterologous promoters, induce retinal toxicity. In addition to the aforementioned challenges, some patients have sustained significant photoreceptor cell loss at the time of diagnosis, rendering gene replacement therapy insufficient to treat the disease. These patients will require cell replacement to restore useful vision. Fortunately, the advent of induced pluripotent stem cell and CRISPR-Cas9 gene editing technologies affords researchers and clinicians a powerful means by which to develop strategies to treat patients with inherited retinal dystrophies. In this review we will discuss the current developments in CRISPR-Cas9 gene editing in vivo in animal models and in vitro in patient-derived cells to study and treat inherited retinal degenerative diseases.


Assuntos
Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Terapia Genética/métodos , Degeneração Retiniana/terapia , Transplante de Células-Tronco , Animais , Humanos , Células-Tronco Pluripotentes Induzidas , Degeneração Retiniana/genética
7.
Methods Mol Biol ; 549: 217-30, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19378206

RESUMO

There is incontrovertible evidence that neural progenitor cells (NPC) are found in the adult brain. The ability to identify and track NPC in the adult brain is of considerable importance if the properties of these cells are to be harnessed as potential therapies for degenerative brain disorders. The most commonly used approach of identifying these NPC in experimental studies, bromodeoxyuridine (BrdU) labelling, is outlined in this chapter. Immunohistochemical protocols for detecting endogenous and exogenous (introduced via transplantation) NPC in fresh-frozen and paraffin wax embedded brain tissue are described. Advice on how to label these NPC is also offered and multi-label fluorescence immunochemical staining approaches to determine the differentiation fate of NPC are described.


Assuntos
Encéfalo/citologia , Imuno-Histoquímica/métodos , Neurônios , Células-Tronco , Adulto , Animais , Anticorpos/metabolismo , Antimetabólitos/metabolismo , Bromodesoxiuridina/metabolismo , Humanos , Camundongos , Neurônios/citologia , Neurônios/fisiologia , Fenótipo , Ratos , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/fisiologia , Inclusão do Tecido/métodos , Preservação de Tecido/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...