Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Cell Stem Cell ; 31(1): 52-70.e8, 2024 01 04.
Article En | MEDLINE | ID: mdl-38181751

Human pluripotent stem cell-derived kidney organoids offer unprecedented opportunities for studying polycystic kidney disease (PKD), which still has no effective cure. Here, we developed both in vitro and in vivo organoid models of PKD that manifested tubular injury and aberrant upregulation of renin-angiotensin aldosterone system. Single-cell analysis revealed that a myriad of metabolic changes occurred during cystogenesis, including defective autophagy. Experimental activation of autophagy via ATG5 overexpression or primary cilia ablation significantly inhibited cystogenesis in PKD kidney organoids. Employing the organoid xenograft model of PKD, which spontaneously developed tubular cysts, we demonstrate that minoxidil, a potent autophagy activator and an FDA-approved drug, effectively attenuated cyst formation in vivo. This in vivo organoid model of PKD will enhance our capability to discover novel disease mechanisms and validate candidate drugs for clinical translation.


Cilia , Polycystic Kidney Diseases , Humans , Kidney , Polycystic Kidney Diseases/drug therapy , Autophagy , Organoids
2.
Biochem Biophys Res Commun ; 686: 149158, 2023 12 17.
Article En | MEDLINE | ID: mdl-37922574

Caspase-11 is an inflammatory caspase that triggers an inflammatory response by regulating non-canonical NLRP3 inflammasome activation. Although the deficiency of both caspase-11 and caspase-1, another inflammatory caspase that functions as an executor of the inflammasome, prevents the development of atherosclerosis, the effect of caspase-11 deficiency alone on the development of atherosclerosis has not been fully evaluated. In the present study, we found that caspase-11 deficiency prevented the formation of the necrotic core, whereas it did not affect the development of atherosclerosis in Apoe-deficient mice. Notably, the infiltration of neutrophils into atherosclerotic lesions was attenuated by caspase-11 deficiency. RNA-seq analysis of stage-dependent expression of atherosclerotic lesions revealed that both upregulations of caspase-11 and neutrophil migration are common features of advanced atherosclerotic lesions. Furthermore, similar expression profiles were observed in unstable human plaque. These data suggest that caspase-11 regulates neutrophil recruitment and plaque destabilization in advanced atherosclerotic lesions.


Atherosclerosis , Plaque, Atherosclerotic , Animals , Humans , Mice , Inflammasomes/metabolism , Caspases , Neutrophil Infiltration , Mice, Knockout , Atherosclerosis/metabolism , Plaque, Atherosclerotic/pathology , Apolipoproteins E/genetics , Apolipoproteins/pharmacology , Mice, Inbred C57BL
4.
Nat Commun ; 13(1): 3646, 2022 06 25.
Article En | MEDLINE | ID: mdl-35752626

The diverse functions of WASP, the deficiency of which causes Wiskott-Aldrich syndrome (WAS), remain poorly defined. We generated three isogenic WAS models using patient induced pluripotent stem cells and genome editing. These models recapitulated WAS phenotypes and revealed that WASP deficiency causes an upregulation of numerous RNA splicing factors and widespread altered splicing. Loss of WASP binding to splicing factor gene promoters frequently leads to aberrant epigenetic activation. WASP interacts with dozens of nuclear speckle constituents and constrains SRSF2 mobility. Using an optogenetic system, we showed that WASP forms phase-separated condensates that encompasses SRSF2, nascent RNA and active Pol II. The role of WASP in gene body condensates is corroborated by ChIPseq and RIPseq. Together our data reveal that WASP is a nexus regulator of RNA splicing that controls the transcription of splicing factors epigenetically and the dynamics of the splicing machinery through liquid-liquid phase separation.


Wiskott-Aldrich Syndrome Protein , Wiskott-Aldrich Syndrome , Alternative Splicing , Cell Nucleus/metabolism , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA Splicing Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism
5.
Nat Commun ; 13(1): 3107, 2022 06 03.
Article En | MEDLINE | ID: mdl-35661110

Inherited glycosylphosphatidylinositol (GPI) deficiency (IGD) is caused by mutations in GPI biosynthesis genes. The mechanisms of its systemic, especially neurological, symptoms are not clarified and fundamental therapy has not been established. Here, we report establishment of mouse models of IGD caused by PIGO mutations as well as development of effective gene therapy. As the clinical manifestations of IGD are systemic and lifelong lasting, we treated the mice with adeno-associated virus for homology-independent knock-in as well as extra-chromosomal expression of Pigo cDNA. Significant amelioration of neuronal phenotypes and growth defect was achieved, opening a new avenue for curing IGDs.


Glycosylphosphatidylinositols , Seizures , Animals , Disease Models, Animal , Genetic Therapy , Glycosylphosphatidylinositols/deficiency , Glycosylphosphatidylinositols/genetics , Immunoglobulin D/genetics , Mice , Seizures/genetics
6.
Elife ; 112022 05 26.
Article En | MEDLINE | ID: mdl-35616535

Cryopyrin-associated periodic syndrome (CAPS) is an autoinflammatory syndrome caused by mutations of NLRP3 gene encoding cryopyrin. Familial cold autoinflammatory syndrome, the mildest form of CAPS, is characterized by cold-induced inflammation induced by the overproduction of IL-1ß. However, the molecular mechanism of how mutated NLRP3 causes inflammasome activation in CAPS remains unclear. Here, we found that CAPS-associated NLRP3 mutants form cryo-sensitive aggregates that function as a scaffold for inflammasome activation. Cold exposure promoted inflammasome assembly and subsequent IL-1ß release triggered by mutated NLRP3. While K+ efflux was dispensable, Ca2+ was necessary for mutated NLRP3-mediated inflammasome assembly. Notably, Ca2+ influx was induced during mutated NLRP3-mediated inflammasome assembly. Furthermore, caspase-1 inhibition prevented Ca2+ influx and inflammasome assembly induced by the mutated NLRP3, suggesting a feed-forward Ca2+ influx loop triggered by mutated NLRP3. Thus, the mutated NLRP3 forms cryo-sensitive aggregates to promote inflammasome assembly distinct from canonical NLRP3 inflammasome activation.


Cryopyrin-Associated Periodic Syndromes , Carrier Proteins/genetics , Caspase 1/genetics , Cryopyrin-Associated Periodic Syndromes/genetics , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
7.
J Immunol ; 205(5): 1393-1405, 2020 09 01.
Article En | MEDLINE | ID: mdl-32727891

Intestinal ischemia/reperfusion (I/R) injury is a life-threatening complication that leads to inflammation and remote organ damage. The NLRP3 inflammasome regulates the caspase-1-dependent release of IL-1ß, an early mediator of inflammation after I/R injury. In this study, we investigated the role of the NLRP3 inflammasome in mice with intestinal I/R injury. Deficiency of NLRP3, ASC, caspase-1/11, or IL-1ß prolonged survival after intestinal I/R injury, but neither NLRP3 nor caspase-1/11 deficiency affected intestinal inflammation. Intestinal I/R injury caused acute lung injury (ALI) characterized by inflammation, reactive oxygen species generation, and vascular permeability, which was markedly improved by NLRP3 deficiency. Bone marrow chimeric experiments showed that NLRP3 in non-bone marrow-derived cells was the main contributor to development of intestinal I/R-induced ALI. The NLRP3 inflammasome in lung vascular endothelial cells is thought to be important to lung vascular permeability. Using mass spectrometry, we identified intestinal I/R-derived lipid mediators that enhanced NLRP3 inflammasome activation in lung vascular endothelial cells. Finally, we confirmed that serum levels of these lipid mediators were elevated in patients with intestinal ischemia. To our knowledge, these findings provide new insights into the mechanism underlying intestinal I/R-induced ALI and suggest that endothelial NLRP3 inflammasome-driven IL-1ß is a novel potential target for treating and preventing this disorder.


Acute Lung Injury/metabolism , Endothelial Cells/metabolism , Inflammasomes/metabolism , Lung/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/metabolism , Animals , Caspase 1/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL
8.
iScience ; 23(5): 101070, 2020 May 22.
Article En | MEDLINE | ID: mdl-32361594

Pyroptosis is a form of regulated cell death that is characterized by gasdermin processing and increased membrane permeability. Caspase-1 and caspase-11 have been considered to be essential for gasdermin D processing associated with inflammasome activation. In the present study, we found that NLRP3 inflammasome activation induces delayed necrotic cell death via ASC in caspase-1/11-deficient macrophages. Furthermore, ASC-mediated caspase-8 activation and subsequent gasdermin E processing are necessary for caspase-1-independent necrotic cell death. We define this necrotic cell death as incomplete pyroptosis because IL-1ß release, a key feature of pyroptosis, is absent, whereas IL-1α release is induced. Notably, unprocessed pro-IL-1ß forms a molecular complex to be retained inside pyroptotic cells. Moreover, incomplete pyroptosis accompanied by IL-1α release is observed under the pharmacological inhibition of caspase-1 with VX765. These findings suggest that caspase-1 inhibition during NLRP3 inflammasome activation modulates forms of cell death and permits the release of IL-1α from dying cells.

9.
Biochem Biophys Res Commun ; 519(1): 15-22, 2019 10 29.
Article En | MEDLINE | ID: mdl-31472954

BACKGROUND: Intestinal ischemia/reperfusion (I/R) injury is a life-threatening complication that leads to inflammation and remote organ damage. However, the underlying mechanism is not yet fully understood. Toll-like receptor 5 (TLR5) is highly expressed in mucosa and recognizes flagellin, the main component of the bacterial flagella. Here, we investigated the role of TLR5 in inflammation and tissue damage after intestinal I/R injury using TLR5-deficient mice. METHODS AND RESULTS: Intestinal levels of TLR5 mRNA and flagellin protein were elevated in wild-type mice subjected to intestinal I/R. Although TLR5 deficiency had no effect on intestinal flagellin levels, it significantly attenuated intestinal injury and inflammatory responses after intestinal I/R. TLR5 deficiency also markedly improved survival in mice after intestinal I/R injury. In wild-type mice, intestinal I/R injury induced remote organ damage, particularly in the lung, which was attenuated by TLR5 deficiency. Furthermore, TLR5 deficiency prevented lung inflammatory responses and vascular permeability after intestinal I/R injury. CONCLUSION: These findings demonstrate a novel role of TLR5 and provide new insights into the mechanism underlying inflammation and tissue damage after intestinal I/R injury.


Inflammation/metabolism , Intestinal Mucosa/metabolism , Reperfusion Injury/metabolism , Toll-Like Receptor 5/metabolism , Animals , Inflammation/pathology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/pathology
10.
Cell Res ; 29(10): 804-819, 2019 Oct.
Article En | MEDLINE | ID: mdl-31444470

In vivo genome editing represents a powerful strategy for both understanding basic biology and treating inherited diseases. However, it remains a challenge to develop universal and efficient in vivo genome-editing tools for tissues that comprise diverse cell types in either a dividing or non-dividing state. Here, we describe a versatile in vivo gene knock-in methodology that enables the targeting of a broad range of mutations and cell types through the insertion of a minigene at an intron of the target gene locus using an intracellularly linearized single homology arm donor. As a proof-of-concept, we focused on a mouse model of premature-aging caused by a dominant point mutation, which is difficult to repair using existing in vivo genome-editing tools. Systemic treatment using our new method ameliorated aging-associated phenotypes and extended animal lifespan, thus highlighting the potential of this methodology for a broad range of in vivo genome-editing applications.


Gene Editing/methods , Animals , CRISPR-Cas Systems/genetics , DNA Repair , Dependovirus/genetics , GATA3 Transcription Factor/genetics , Gene Knock-In Techniques , Genetic Therapy/methods , Genetic Vectors/metabolism , Human Embryonic Stem Cells , Humans , Introns , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/cytology , Neurons/metabolism , RNA, Guide, Kinetoplastida/metabolism , Rats , Tubulin/genetics
11.
Cell Stem Cell ; 25(3): 373-387.e9, 2019 09 05.
Article En | MEDLINE | ID: mdl-31303547

Human pluripotent stem cell-derived kidney organoids recapitulate developmental processes and tissue architecture, but intrinsic limitations, such as lack of vasculature and functionality, have greatly hampered their application. Here we establish a versatile protocol for generating vascularized three-dimensional (3D) kidney organoids. We employ dynamic modulation of WNT signaling to control the relative proportion of proximal versus distal nephron segments, producing a correlative level of vascular endothelial growth factor A (VEGFA) to define a resident vascular network. Single-cell RNA sequencing identifies a subset of nephron progenitor cells as a potential source of renal vasculature. These kidney organoids undergo further structural and functional maturation upon implantation. Using this kidney organoid platform, we establish an in vitro model of autosomal recessive polycystic kidney disease (ARPKD), the cystic phenotype of which can be effectively prevented by gene correction or drug treatment. Our studies provide new avenues for studying human kidney development, modeling disease pathogenesis, and performing patient-specific drug validation.


Kidney/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Polycystic Kidney, Autosomal Recessive/pathology , Cell Differentiation , Cells, Cultured , Drug Discovery , Genetic Therapy , Humans , Kidney/blood supply , Neovascularization, Physiologic , Organ Culture Techniques , Organogenesis , Organoids/blood supply , Polycystic Kidney, Autosomal Recessive/metabolism , Polycystic Kidney, Autosomal Recessive/therapy , Precision Medicine , Vascular Endothelial Growth Factor A/metabolism , Wnt Signaling Pathway
12.
Sci Rep ; 9(1): 10363, 2019 07 17.
Article En | MEDLINE | ID: mdl-31316105

Long-term peritoneal dialysis (PD) therapy leads to peritoneal inflammation and fibrosis. However, the mechanism underlying PD-related peritoneal inflammation and fibrosis remains unclear. NLRP3 inflammasome regulates the caspase-1-dependent release of interleukin-1ß and mediates inflammation in various diseases. Here, we investigated the role of NLRP3 inflammasome in a murine model of PD-related peritoneal fibrosis induced by methylglyoxal (MGO). Inflammasome-related proteins were upregulated in the peritoneum of MGO-treated mice. MGO induced parietal and visceral peritoneal fibrosis in wild-type mice, which was significantly reduced in mice deficient in NLRP3, ASC, and interleukin-1ß (IL-1ß). ASC deficiency reduced the expression of inflammatory cytokines and fibrotic factors, and the infiltration of macrophages. However, myeloid cell-specific ASC deficiency failed to inhibit MGO-induced peritoneal fibrosis. MGO caused hemorrhagic ascites, fibrin deposition, and plasminogen activator inhibitor-1 upregulation, but all of these manifestations were inhibited by ASC deficiency. Furthermore, in vitro experiments showed that MGO induced cell death via the generation of reactive oxygen species in vascular endothelial cells, which was inhibited by ASC deficiency. Our results showed that endothelial NLRP3 inflammasome contributes to PD-related peritoneal inflammation and fibrosis, and provide new insights into the mechanisms underlying the pathogenesis of this disorder.


CARD Signaling Adaptor Proteins/physiology , Inflammasomes/physiology , Interleukin-1beta/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/etiology , Animals , CARD Signaling Adaptor Proteins/deficiency , CARD Signaling Adaptor Proteins/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-1beta/deficiency , Interleukin-1beta/genetics , Leukocytes/pathology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/pathology , Pyruvaldehyde/toxicity , Reactive Oxygen Species
13.
Cell ; 168(3): 473-486.e15, 2017 01 26.
Article En | MEDLINE | ID: mdl-28129541

Interspecies blastocyst complementation enables organ-specific enrichment of xenogenic pluripotent stem cell (PSC) derivatives. Here, we establish a versatile blastocyst complementation platform based on CRISPR-Cas9-mediated zygote genome editing and show enrichment of rat PSC-derivatives in several tissues of gene-edited organogenesis-disabled mice. Besides gaining insights into species evolution, embryogenesis, and human disease, interspecies blastocyst complementation might allow human organ generation in animals whose organ size, anatomy, and physiology are closer to humans. To date, however, whether human PSCs (hPSCs) can contribute to chimera formation in non-rodent species remains unknown. We systematically evaluate the chimeric competency of several types of hPSCs using a more diversified clade of mammals, the ungulates. We find that naïve hPSCs robustly engraft in both pig and cattle pre-implantation blastocysts but show limited contribution to post-implantation pig embryos. Instead, an intermediate hPSC type exhibits higher degree of chimerism and is able to generate differentiated progenies in post-implantation pig embryos.


Chimerism , Gene Editing , Mammals/embryology , Animals , Blastocyst , CRISPR-Cas Systems , Cattle , Embryo, Mammalian/cytology , Female , Humans , Male , Mammals/classification , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Pluripotent Stem Cells , Rats , Rats, Sprague-Dawley , Sus scrofa
14.
Nature ; 540(7631): 144-149, 2016 12 01.
Article En | MEDLINE | ID: mdl-27851729

Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.


CRISPR-Cas Systems/genetics , Gene Editing/methods , Gene Targeting/methods , Genome/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Animals , Cell Division , Disease Models, Animal , Gene Knock-In Techniques , Genetic Therapy/methods , Neurons/cytology , Neurons/metabolism , Rats , Sequence Homology
15.
Science ; 348(6239): 1160-3, 2015 Jun 05.
Article En | MEDLINE | ID: mdl-25931448

Werner syndrome (WS) is a premature aging disorder caused by WRN protein deficiency. Here, we report on the generation of a human WS model in human embryonic stem cells (ESCs). Differentiation of WRN-null ESCs to mesenchymal stem cells (MSCs) recapitulates features of premature cellular aging, a global loss of H3K9me3, and changes in heterochromatin architecture. We show that WRN associates with heterochromatin proteins SUV39H1 and HP1α and nuclear lamina-heterochromatin anchoring protein LAP2ß. Targeted knock-in of catalytically inactive SUV39H1 in wild-type MSCs recapitulates accelerated cellular senescence, resembling WRN-deficient MSCs. Moreover, decrease in WRN and heterochromatin marks are detected in MSCs from older individuals. Our observations uncover a role for WRN in maintaining heterochromatin stability and highlight heterochromatin disorganization as a potential determinant of human aging.


Aging/metabolism , Cellular Senescence , Exodeoxyribonucleases/metabolism , Heterochromatin/metabolism , Mesenchymal Stem Cells/metabolism , RecQ Helicases/metabolism , Werner Syndrome/metabolism , Aging/genetics , Animals , Cell Differentiation , Centromere/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Exodeoxyribonucleases/genetics , Gene Knockout Techniques , HEK293 Cells , Heterochromatin/chemistry , Humans , Membrane Proteins/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Models, Biological , RecQ Helicases/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Werner Syndrome/genetics , Werner Syndrome Helicase
16.
Nature ; 521(7552): 316-21, 2015 May 21.
Article En | MEDLINE | ID: mdl-25945737

Pluripotency, the ability to generate any cell type of the body, is an evanescent attribute of embryonic cells. Transitory pluripotent cells can be captured at different time points during embryogenesis and maintained as embryonic stem cells or epiblast stem cells in culture. Since ontogenesis is a dynamic process in both space and time, it seems counterintuitive that these two temporal states represent the full spectrum of organismal pluripotency. Here we show that by modulating culture parameters, a stem-cell type with unique spatial characteristics and distinct molecular and functional features, designated as region-selective pluripotent stem cells (rsPSCs), can be efficiently obtained from mouse embryos and primate pluripotent stem cells, including humans. The ease of culturing and editing the genome of human rsPSCs offers advantages for regenerative medicine applications. The unique ability of human rsPSCs to generate post-implantation interspecies chimaeric embryos may facilitate our understanding of early human development and evolution.


Chimera , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques/methods , Cell Line , Embryonic Stem Cells/cytology , Female , Germ Layers/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Male , Mice , Pan troglodytes , Pluripotent Stem Cells/metabolism , Regenerative Medicine , Species Specificity
17.
Cell Stem Cell ; 15(1): 31-6, 2014 Jul 03.
Article En | MEDLINE | ID: mdl-24996168

The utility of genome editing technologies for disease modeling and developing cellular therapies has been extensively documented, but the impact of these technologies on mutational load at the whole-genome level remains unclear. We performed whole-genome sequencing to evaluate the mutational load at single-base resolution in individual gene-corrected human induced pluripotent stem cell (hiPSC) clones in three different disease models. In single-cell clones, gene correction by helper-dependent adenoviral vector (HDAdV) or Transcription Activator-Like Effector Nuclease (TALEN) exhibited few off-target effects and a low level of sequence variation, comparable to that accumulated in routine hiPSC culture. The sequence variants were randomly distributed and unique to individual clones. We also combined both technologies and developed a TALEN-HDAdV hybrid vector, which significantly increased gene-correction efficiency in hiPSCs. Therefore, with careful monitoring via whole-genome sequencing it is possible to apply genome editing to human pluripotent cells with minimal impact on genomic mutational load.


Adenoviridae/genetics , Endonucleases/metabolism , Genetic Therapy , Genetic Vectors/metabolism , Induced Pluripotent Stem Cells/physiology , CRISPR-Cas Systems/genetics , Clone Cells , DNA Repair/genetics , Endonucleases/genetics , Genetic Vectors/genetics , Genome/genetics , HEK293 Cells , Humans , Mutation/genetics , Regenerative Medicine , Sequence Analysis, DNA
18.
Nat Commun ; 5: 4330, 2014 Jul 07.
Article En | MEDLINE | ID: mdl-24999918

Fanconi anaemia (FA) is a recessive disorder characterized by genomic instability, congenital abnormalities, cancer predisposition and bone marrow (BM) failure. However, the pathogenesis of FA is not fully understood partly due to the limitations of current disease models. Here, we derive integration free-induced pluripotent stem cells (iPSCs) from an FA patient without genetic complementation and report in situ gene correction in FA-iPSCs as well as the generation of isogenic FANCA-deficient human embryonic stem cell (ESC) lines. FA cellular phenotypes are recapitulated in iPSCs/ESCs and their adult stem/progenitor cell derivatives. By using isogenic pathogenic mutation-free controls as well as cellular and genomic tools, our model serves to facilitate the discovery of novel disease features. We validate our model as a drug-screening platform by identifying several compounds that improve hematopoietic differentiation of FA-iPSCs. These compounds are also able to rescue the hematopoietic phenotype of FA patient BM cells.


Drug Evaluation, Preclinical/methods , Fanconi Anemia/etiology , Fanconi Anemia/pathology , Models, Biological , Stem Cells/pathology , Cell Differentiation , Epigenesis, Genetic , Fanconi Anemia/drug therapy , Fanconi Anemia Complementation Group A Protein/genetics , Humans , Induced Pluripotent Stem Cells , Male , Young Adult
19.
Mol Ther ; 20(2): 424-31, 2012 Feb.
Article En | MEDLINE | ID: mdl-22146343

Low efficiencies of gene targeting via homologous recombination (HR) have limited basic research and applications using human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Here, we show highly and equally efficient gene knockout and knock-in at both transcriptionally active (HPRT1, KU80, LIG1, LIG3) and inactive (HB9) loci in these cells using high-capacity helper-dependent adenoviral vectors (HDAdVs). Without the necessity of introducing artificial DNA double-strand breaks, 7-81% of drug-resistant colonies were gene-targeted by accurate HR, which were not accompanied with additional ectopic integrations. Even at the motor neuron-specific HB9 locus, the enhanced green fluorescent protein (EGFP) gene was accurately knocked in in 23-57% of drug-resistant colonies. In these clones, induced differentiation into the HB9-positive motor neuron correlated with EGFP expression. Furthermore, HDAdV infection had no detectable adverse effects on the undifferentiated state and pluripotency of hESCs and hiPSCs. These results suggest that HDAdV is one of the best methods for efficient and accurate gene targeting in hESCs and hiPSCs and might be especially useful for therapeutic applications.


Adenoviridae/genetics , Embryonic Stem Cells/metabolism , Genetic Vectors/genetics , Homologous Recombination , Induced Pluripotent Stem Cells/metabolism , Antigens, Nuclear/genetics , Cell Line , DNA Ligase ATP , DNA Ligases/genetics , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Gene Knock-In Techniques , Gene Knockout Techniques , Gene Order , Gene Targeting , Heterozygote , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Induced Pluripotent Stem Cells/cytology , Ku Autoantigen , Mutation , Poly-ADP-Ribose Binding Proteins , Xenopus Proteins
20.
Biochem Biophys Res Commun ; 388(4): 711-7, 2009 Oct 30.
Article En | MEDLINE | ID: mdl-19695233

Human pluripotent stem cells, such as embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs), have the ability to differentiate into various cell types, and will become a potential source of cellular materials for regenerative medicine. To make full use of hESCs or hiPSCs for both basic and clinical research, genetic modification, especially gene targeting via homologous recombination (HR), would be an essential technique. This report describes the successful gene targeting of the hypoxanthine phosphoribosyl transferase 1 (HPRT1) and the NANOG loci in human pluripotent stem cells with adeno-associated virus (AAV) vectors. At the HPRT1 locus, up to 1% of stable transformants were targeted via HR with an AAV-HPRT1 targeting vector, without loss of pluripotency. On the other hand, 20-87% of stable transformants were targeted using an AAV-NANOG-targeting vector designed for the promoter-trap strategy. In the KhES-3 cell line, which shows particularly high fragility to experimental manipulation, gene targeting was successful only by using an AAV vector but not by electroporation. In addition to hESC, gene targeting was achieved in hiPSC lines at similar frequencies. These data indicate that AAV vectors may therefore be a useful tool to introduce genetic modifications in hESCs and hiPSCs.


Dependovirus , Gene Targeting/methods , Genetic Vectors , Pluripotent Stem Cells/metabolism , Homeodomain Proteins/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Nanog Homeobox Protein
...