Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Am J Respir Cell Mol Biol ; 57(6): 711-720, 2017 12.
Article in English | MEDLINE | ID: mdl-28708422

ABSTRACT

Cystic fibrosis (CF) remains the most lethal genetic disease in the Caucasian population. However, there is great variability in clinical phenotypes and survival times, even among patients harboring the same genotype. We identified five patients with CF and a homozygous F508del mutation in the CFTR gene who were in their fifth or sixth decade of life and had shown minimal changes in lung function over a longitudinal period of more than 20 years. Because of the rarity of this long-term nonprogressive phenotype, we hypothesized these individuals may carry rare genetic variants in modifier genes that ameliorate disease severity. Individuals at the extremes of survival time and lung-function trajectory underwent whole-exome sequencing, and the sequencing data were filtered to include rare missense, stopgain, indel, and splicing variants present with a mean allele frequency of <0.2% in general population databases. Epithelial sodium channel (ENaC) mutants were generated via site-directed mutagenesis and expressed for Xenopus oocyte assays. Four of the five individuals carried extremely rare or never reported variants in the SCNN1D and SCNN1B genes of the ENaC. Separately, an independently enriched rare variant in SCNN1D was identified in the Exome Variant Server database associated with a milder pulmonary disease phenotype. Functional analysis using Xenopus oocytes revealed that two of the three variants in δ-ENaC encoded by SCNN1D exhibited hypomorphic channel activity. Our data suggest a potential role for δ-ENaC in controlling sodium reabsorption in the airways, and advance the plausibility of ENaC as a therapeutic target in CF.


Subject(s)
Amino Acid Sequence , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Epithelial Sodium Channels/metabolism , Sequence Deletion , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Sodium Channels/genetics , Female , Humans , Male , Xenopus , Xenopus laevis
2.
Methods ; 101: 27-35, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26525194

ABSTRACT

Precise gene correction using the CRISPR-Cas9 system in human iPS cells holds great promise for various applications, such as the study of gene functions, disease modeling, and gene therapy. In this review article, we summarize methods for effective editing of genomic sequences of iPS cells based on our experiences correcting dystrophin gene mutations with the CRISPR-Cas9 system. Designing specific sgRNAs as well as having efficient transfection methods and proper detection assays to assess genomic cleavage activities are critical for successful genome editing in iPS cells. In addition, because iPS cells are fragile by nature when dissociated into single cells, a step-by-step confirmation during the cell recovery process is recommended to obtain an adequate number of genome-edited iPS cell clones. We hope that the techniques described here will be useful for researchers from diverse backgrounds who would like to perform genome editing in iPS cells.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Base Sequence , CRISPR-Cas Systems , Cell Culture Techniques , Gene Editing , Genome, Human , HEK293 Cells , Humans , Transfection
3.
Dev Growth Differ ; 56(1): 63-77, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24329887

ABSTRACT

Genetic mutations in humans are associated with congenital disorders and phenotypic traits. Gene therapy holds the promise to cure such genetic disorders, although it has suffered from several technical limitations for decades. Recent progress in gene editing technology using tailor-made nucleases, such as meganucleases (MNs), zinc finger nucleases (ZFNs), TAL effector nucleases (TALENs) and, more recently, CRISPR/Cas9, has significantly broadened our ability to precisely modify target sites in the human genome. In this review, we summarize recent progress in gene correction approaches of the human genome, with a particular emphasis on the clinical applications of gene therapy.


Subject(s)
Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Genetic Engineering/methods , Genetic Therapy/methods , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Endodeoxyribonucleases/chemistry , Humans , Zinc Fingers
4.
Stem Cell Reports ; 10(6): 1835-1850, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29731430

ABSTRACT

Nakajo-Nishimura syndrome (NNS) is an immunoproteasome-associated autoinflammatory disorder caused by a mutation of the PSMB8 gene. Although dysfunction of the immunoproteasome causes various cellular stresses attributed to the overproduction of inflammatory cytokines and chemokines in NNS, the underlying mechanisms of the autoinflammation are still largely unknown. To investigate and understand the mechanisms and signal pathways in NNS, we established a panel of isogenic pluripotent stem cell (PSC) lines with PSMB8 mutation. Activity of the immunoproteasome in PSMB8-mutant PSC-derived myeloid cell lines (MT-MLs) was reduced even without stimulation compared with non-mutant-MLs. In addition, MT-MLs showed an overproduction of inflammatory cytokines and chemokines, with elevated reactive oxygen species (ROS) and phosphorylated p38 MAPK levels. Treatment with p38 MAPK inhibitor and antioxidants decreased the abnormal production of cytokines and chemokines. The current PSC model revealed a specific ROS-mediated inflammatory pathway, providing a platform for the discovery of alternative therapeutic options for NNS and related immunoproteasome disorders.


Subject(s)
Erythema Nodosum/etiology , Erythema Nodosum/metabolism , Fingers/abnormalities , Oxidative Stress , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction , Biomarkers , Cell Differentiation/genetics , Erythema Nodosum/pathology , Fingers/pathology , Gene Expression Profiling , Humans , Interferon-gamma/metabolism , Models, Biological , Mutation , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Reactive Oxygen Species/metabolism , Transcriptome , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Cell Rep ; 15(10): 2301-2312, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27239027

ABSTRACT

Duchenne muscular dystrophy (DMD) remains an intractable genetic disease. Althogh there are several animal models of DMD, there is no human cell model that carries patient-specific DYSTROPHIN mutations. Here, we present a human DMD model using human induced pluripotent stem cells (hiPSCs). Our model reveals concordant disease-related phenotypes with patient-dependent variation, which are partially reversed by genetic and pharmacological approaches. Our "chemical-compound-based" strategy successfully directs hiPSCs into expandable myoblasts, which exhibit a myogenic transcriptional program, forming striated contractile myofibers and participating in muscle regeneration in vivo. DMD-hiPSC-derived myoblasts show disease-related phenotypes with patient-to-patient variability, including aberrant expression of inflammation or immune-response genes and collagens, increased BMP/TGFß signaling, and reduced fusion competence. Furthermore, by genetic correction and pharmacological "dual-SMAD" inhibition, the DMD-hiPSC-derived myoblasts and genetically corrected isogenic myoblasts form "rescued" multi-nucleated myotubes. In conclusion, our findings demonstrate the feasibility of establishing a human "DMD-in-a-dish" model using hiPSC-based disease modeling.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Models, Biological , Muscular Dystrophy, Duchenne/pathology , Myoblasts/pathology , Animals , Cell Line , Flow Cytometry , Humans , Mice , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myoblasts/metabolism , Phenotype , Signal Transduction , Smad Proteins/metabolism
6.
Stem Cell Reports ; 4(1): 143-154, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25434822

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe muscle-degenerative disease caused by a mutation in the dystrophin gene. Genetic correction of patient-derived induced pluripotent stem cells (iPSCs) by TALENs or CRISPR-Cas9 holds promise for DMD gene therapy; however, the safety of such nuclease treatment must be determined. Using a unique k-mer database, we systematically identified a unique target region that reduces off-target sites. To restore the dystrophin protein, we performed three correction methods (exon skipping, frameshifting, and exon knockin) in DMD-patient-derived iPSCs, and found that exon knockin was the most effective approach. We further investigated the genomic integrity by karyotyping, copy number variation array, and exome sequencing to identify clones with a minimal mutation load. Finally, we differentiated the corrected iPSCs toward skeletal muscle cells and successfully detected the expression of full-length dystrophin protein. These results provide an important framework for developing iPSC-based gene therapy for genetic disorders using programmable nucleases.


Subject(s)
CRISPR-Cas Systems/genetics , Dystrophin/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Muscular Dystrophy, Duchenne/genetics , Base Sequence , DNA Copy Number Variations , Dystrophin/metabolism , Exons , Gene Order , Gene Targeting , Genetic Loci , Genetic Therapy , Humans , Karyotype , Molecular Sequence Data , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Mutagenesis, Insertional , Mutation , Reading Frames , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL