Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Stem Cell Res ; 61: 102779, 2022 05.
Article in English | MEDLINE | ID: mdl-35427904

ABSTRACT

The cardiac muscle-specific protein, α-myosin heavy chain (αMHC), is a major component of cardiac muscle filaments involved in cardiac muscle contraction. Here, we established an αMHC-enhanced fluorescent protein (EGFP) knock-in human pluripotent stem cell (hPSC) line by linking the EGFP gene to the C-terminal region of αMHC via a 2A non-joining peptide using CRISPR/Cas9 nuclease. The EGFP reporter precisely reflected the endogenous level of αMHC upon the induction of cardiac differentiation. This reporter cell line will be a valuable platform for cardiotoxicity tests, drug screening, and investigating the pathological mechanisms of cardiomyocytes.


Subject(s)
CRISPR-Cas Systems , Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Cell Line , Gene Targeting , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Myosin Heavy Chains/genetics , Pluripotent Stem Cells/metabolism
2.
Stem Cell Res ; 53: 102321, 2021 05.
Article in English | MEDLINE | ID: mdl-33878708

ABSTRACT

Brachyury is an embryonic nuclear transcription factor required for mesoderm formation and differentiation. Here, we introduced an mCherry reporter into the C-terminus of Brachyury in the human pluripotent stem cell line SNUhES3 using the CRISPR/Cas9 nuclease approach. Successful gene editing was verified by DNA sequencing. SNUhES3-Brachyury-mCherry cells expressed pluripotent stem cell markers, exhibited a normal karyotype, and could generate all three germ layers. This cell line expressed the red fluorescence protein mCherry upon the induction of mesoderm differentiation. This reporter cell line could be used to monitor mesodermal population enrichment during mesodermal differentiation.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Line , Fetal Proteins , Humans , T-Box Domain Proteins
3.
Stem Cell Res ; 48: 101948, 2020 10.
Article in English | MEDLINE | ID: mdl-32798916

ABSTRACT

Hemophilia A is an ideal target for cell or gene therapy because a mild increase in coagulation factor VIII (FVIII) improves symptoms in patients with severe hemophilia A. In this study, we used CRISPR/Cas9 to insert FVIII cDNA into exon 1 of the mutant FVIII locus in induced pluripotent stem cells (iPSCs) from a hemophilia A patient. This gene-modified YCMi001-B-1 line maintained its pluripotency, formed all three germ layers, and had a normal karyotype. In addition, FVIII expression was confirmed in YCMi001-B-1-derived endothelial cells.


Subject(s)
Factor VIII , Hemophilia A , Induced Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Cell Line , Endothelial Cells/metabolism , Factor VIII/genetics , Factor VIII/metabolism , Gene Editing , Hemophilia A/genetics , Hemophilia A/therapy , Humans , Induced Pluripotent Stem Cells/metabolism
4.
Stem Cell Reports ; 12(6): 1242-1249, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31105049

ABSTRACT

Hemophilia A (HA) is caused by genetic mutations in the blood coagulation factor VIII (FVIII) gene. Genome-editing approaches can be used to target the mutated site itself in patient-derived induced pluripotent stem cells (iPSCs). However, these approaches can be hampered by difficulty in preparing thousands of editing platforms for each corresponding variant found in HA patients. Here, we report a universal approach to correct the various mutations in HA patient iPSCs by the targeted insertion of the FVIII gene into the human H11 site via CRISPR/Cas9. We derived corrected clones from two types of patient iPSCs with frequencies of up to 64% and 66%, respectively, without detectable unwanted off-target mutations. Moreover, we demonstrated that endothelial cells differentiated from the corrected iPSCs successfully secreted functional protein. This strategy may provide a universal therapeutic method for correcting all genetic variants found in HA patients.


Subject(s)
CRISPR-Cas Systems , Factor VIII , Gene Editing , Hemophilia A , Induced Pluripotent Stem Cells , Mutation , Factor VIII/biosynthesis , Factor VIII/genetics , Hemophilia A/genetics , Hemophilia A/metabolism , Hemophilia A/pathology , Hemophilia A/therapy , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology
5.
Exp Mol Med ; 51(4): 1-9, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30996250

ABSTRACT

Target-specific genome editing, using engineered nucleases zinc finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), and type II clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), is considered a promising approach to correct disease-causing mutations in various human diseases. In particular, hemophilia A can be considered an ideal target for gene modification via engineered nucleases because it is a monogenic disease caused by a mutation in coagulation factor VIII (FVIII), and a mild restoration of FVIII levels in plasma can prevent disease symptoms in patients with severe hemophilia A. In this study, we describe a universal genome correction strategy to restore FVIII expression in induced pluripotent stem cells (iPSCs) derived from a patient with hemophilia A by the human elongation factor 1 alpha (EF1α)-mediated normal FVIII gene expression in the FVIII locus of the patient. We used the CRISPR/Cas9-mediated homology-directed repair (HDR) system to insert the B-domain deleted from the FVIII gene with the human EF1α promoter. After gene targeting, the FVIII gene was correctly inserted into iPSC lines at a high frequency (81.81%), and these cell lines retained pluripotency after knock-in and neomycin resistance cassette removal. More importantly, we confirmed that endothelial cells from the gene-corrected iPSCs could generate functionally active FVIII protein from the inserted FVIII gene. This is the first demonstration that the FVIII locus is a suitable site for integration of the normal FVIII gene and can restore FVIII expression by the EF1α promoter in endothelial cells differentiated from the hemophilia A patient-derived gene-corrected iPSCs.


Subject(s)
Factor VIII/metabolism , Hemophilia A/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Cells, Cultured , Exons/genetics , Factor VIII/genetics , Gene Editing , Genetic Engineering/methods , HEK293 Cells , Hemophilia A/genetics , Humans , Promoter Regions, Genetic/genetics , Streptococcus pyogenes/genetics , Streptococcus pyogenes/metabolism
6.
Invest Ophthalmol Vis Sci ; 59(8): 3531-3542, 2018 07 02.
Article in English | MEDLINE | ID: mdl-30025074

ABSTRACT

Purpose: We aimed to establish an efficient method for retinal ganglion cell (RGC) differentiation from human pluripotent stem cells (hPSCs) using defined factors. Methods: To define the contribution of specific signal pathways to RGC development and optimize the differentiation of hPSCs toward RGCs, we examined RGC differentiation in three stages: (1) eye field progenitors expressing the eye field transcription factors (EFTFs), (2) RGC progenitors expressing MATH5, and (3) RGCs expressing BRN3B and ISLET1. By monitoring the condition that elicited the highest yield of cells expressing stage-specific markers, we determined the optimal concentrations and combinations of signaling pathways required for efficient generation of RGCs from hPSCs. Results: Precise modulation of signaling pathways, including Wnt, insulin growth factor-1, and fibroblast growth factor, in combination with mechanical isolation of neural rosette cell clusters significantly enriched RX and PAX6 double-positive eye field progenitors from hPSCs by day 12. Furthermore, Notch signal inhibition facilitated differentiation into MATH5-positive progenitors at 90% efficiency by day 20, and these cells further differentiated to BRN3B and ISLET1 double-positive RGCs at 45% efficiency by day 40. RGCs differentiated via this method were functional as exemplified by their ability to generate action potentials, express microfilament components on neuronal processes, and exhibit axonal transportation of mitochondria. Conclusions: This protocol offers highly defined culture conditions for RGC differentiation from hPSCs and in vitro disease model and cell source for transplantation for diseases related to RGCs.


Subject(s)
Cell Differentiation/physiology , Pluripotent Stem Cells/cytology , Retinal Ganglion Cells/cytology , Signal Transduction/physiology , Axonal Transport/physiology , Biomarkers/metabolism , Cell Line , Cells, Cultured , Electrophysiology , Humans , Immunohistochemistry , Mitochondria/metabolism , Pluripotent Stem Cells/metabolism , Real-Time Polymerase Chain Reaction , Retinal Ganglion Cells/metabolism , Transcription Factors/metabolism
7.
Nat Protoc ; 11(11): 2154-2169, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27711053

ABSTRACT

Genome engineering technology using engineered nucleases has been rapidly developing, enabling the efficient correction of simple mutations. However, the precise correction of structural variations (SVs) such as large inversions remains limited. Here we describe a detailed procedure for the modeling or correction of large chromosomal rearrangements and short nucleotide repeat expansions using engineered nucleases in human induced pluripotent stem cells (hiPSCs) from a healthy donor and patients with SVs. This protocol includes the delivery of engineered nucleases with no donor template to hiPSCs, and genotyping and derivation/characterization of gene-manipulated hiPSC clones. With engineered nucleases, genomic inversions, reversions, and deletions of short nucleotide expansions can be identified in 2 weeks, and desired clones can be generated in as little as 3-4 weeks. This protocol enables the correction of large inverted segments and short nucleotide repeat expansions in diseases such as hemophilia A, fragile X syndrome, Hunter syndrome, and Friedreich's ataxia.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Engineering/methods , Genetic Variation , Induced Pluripotent Stem Cells/metabolism , Models, Genetic , Base Sequence , DNA End-Joining Repair/genetics , Gene Rearrangement/genetics , Genome, Human/genetics , Humans , Repetitive Sequences, Nucleic Acid/genetics
8.
Hum Genet ; 135(9): 977-81, 2016 09.
Article in English | MEDLINE | ID: mdl-27357631

ABSTRACT

Hemophilia is caused by various mutations in blood coagulation factor genes, including factor VIII (FVIII) and factor IX (FIX), that encode key proteins in the blood clotting pathway. Although the addition of therapeutic genes or infusion of clotting factors may be used to remedy hemophilia's symptoms, no permanent cure for the disease exists. Moreover, patients often develop neutralizing antibodies or experience adverse effects that limit the therapy's benefits. However, targeted gene therapy involving the precise correction of these mutated genes at the genome level using programmable nucleases is a promising strategy. These nucleases can induce double-strand breaks (DSBs) on genomes, and repairs of such induced DSBs by the two cellular repair systems enable a targeted gene correction. Going beyond cultured cell systems, we are now entering the age of direct gene correction in vivo using various delivery tools. Here, we describe the current status of in vivo and ex vivo genome-editing technology related to potential hemophilia gene correction and the prominent issues surrounding its application in patients with monogenic diseases.


Subject(s)
Gene Editing , Genetic Therapy , Hemophilia A/therapy , Humans
9.
Trends Biotechnol ; 34(7): 548-561, 2016 07.
Article in English | MEDLINE | ID: mdl-27016031

ABSTRACT

The analysis of chromosomal structural variations (SVs), such as inversions and translocations, was made possible by the completion of the human genome project and the development of genome-wide sequencing technologies. SVs contribute to genetic diversity and evolution, although some SVs can cause diseases such as hemophilia A in humans. Genome engineering technology using programmable nucleases (e.g., ZFNs, TALENs, and CRISPR/Cas9) has been rapidly developed, enabling precise and efficient genome editing for SV research. Here, we review advances in modeling and gene correction of SVs, focusing on inversion, translocation, and nucleotide repeat expansion.


Subject(s)
Chromosome Aberrations , Chromosomes, Human , Gene Editing/methods , Genetic Therapy/methods , Genetic Variation , Genome, Human , Humans
10.
Cell Rep ; 13(2): 234-41, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26440889

ABSTRACT

Fragile X syndrome (FXS) is the most common form of inherited intellectual disability, resulting from a CGG repeat expansion in the fragile X mental retardation 1 (FMR1) gene. Here, we report a strategy for CGG repeat correction using CRISPR/Cas9 for targeted deletion in both embryonic stem cells and induced pluripotent stem cells derived from FXS patients. Following gene correction in FXS induced pluripotent stem cells, FMR1 expression was restored and sustained in neural precursor cells and mature neurons. Strikingly, after removal of the CGG repeats, the upstream CpG island of the FMR1 promoter showed extensive demethylation, an open chromatin state, and transcription initiation. These results suggest a silencing maintenance mechanism for the FMR1 promoter that is dependent on the existence of the CGG repeat expansion. Our strategy for deletion of trinucleotide repeats provides further insights into the molecular mechanisms of FXS and future therapies of trinucleotide repeat disorders.


Subject(s)
DNA Methylation , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Gene Silencing , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Trinucleotide Repeats , CRISPR-Cas Systems , Cells, Cultured , CpG Islands , Fragile X Mental Retardation Protein/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Promoter Regions, Genetic
11.
Cell Stem Cell ; 17(2): 213-20, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26212079

ABSTRACT

Hemophilia A is an X-linked genetic disorder caused by mutations in the F8 gene, which encodes the blood coagulation factor VIII. Almost half of all severe hemophilia A cases result from two gross (140-kbp or 600-kbp) chromosomal inversions that involve introns 1 and 22 of the F8 gene, respectively. We derived induced pluripotent stem cells (iPSCs) from patients with these inversion genotypes and used CRISPR-Cas9 nucleases to revert these chromosomal segments back to the WT situation. We isolated inversion-corrected iPSCs with frequencies of up to 6.7% without detectable off-target mutations based on whole-genome sequencing or targeted deep sequencing. Endothelial cells differentiated from corrected iPSCs expressed the F8 gene and functionally rescued factor VIII deficiency in an otherwise lethal mouse model of hemophilia. Our results therefore provide a proof of principle for functional correction of large chromosomal rearrangements in patient-derived iPSCs and suggest potential therapeutic applications.


Subject(s)
CRISPR-Cas Systems/genetics , Chromosome Inversion/genetics , Factor VII/genetics , Hemophilia A/genetics , Induced Pluripotent Stem Cells/metabolism , Animals , Base Sequence , Clone Cells , HeLa Cells , Humans , Mice , Molecular Sequence Data
12.
Biochem Biophys Res Commun ; 378(3): 629-33, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19059211

ABSTRACT

We examined the role of zebrafish (Danio rerio) Jak2a, a homolog of mammalian Jak2, in the developing embryo by injecting in vitro synthesized Jak2a shRNA into zebrafish zygotes. Blood circulation was suppressed in Jak2a shRNA-injected embryos from 24hours post fertilization (hpf) and all embryos died with enlarged pericardium, shortened body lengths, and defects in some vasculature within 8 days post fertilization. O-dianisidine staining of red blood cells revealed normal blood island formation with no circulating red blood cells. As in Jak2(-/-) transgenic mice, expression of definitive Ba1 globin was significantly reduced in Jak2a knockdown embryos at 36hpf, whereas expression of other hematopoietic markers, primitive be1 globin, gata-1, and scl, were unaffected. More importantly, blood vessel formation was disturbed in Jak2a knockdown embryos as revealed by alkaline phosphatase staining at 72hpf. Thus, our data indicate that zebrafish Jak2a is important in both definitive hematopoiesis and blood vessel formation.


Subject(s)
Blood Vessels/embryology , Hematopoiesis , Neovascularization, Physiologic , Protein-Tyrosine Kinases/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Blood Circulation/genetics , Embryo, Nonmammalian/enzymology , Gene Knockdown Techniques , Gene Silencing , Hematopoiesis/genetics , Mice , Mice, Transgenic , Neovascularization, Physiologic/genetics , Protein-Tyrosine Kinases/genetics , RNA, Small Interfering/genetics , Transgenes , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...