Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Stem Cells Transl Med ; 7(1): 87-97, 2018 01.
Article in English | MEDLINE | ID: mdl-29164808

ABSTRACT

Beta-thalassemia is one of the most common recessive genetic diseases, caused by mutations in the HBB gene. Over 200 different types of mutations in the HBB gene containing three exons have been identified in patients with ß-thalassemia (ß-thal) whereas a homozygous mutation in exon 1 causes sickle cell disease (SCD). Novel therapeutic strategies to permanently correct the HBB mutation in stem cells that are able to expand and differentiate into erythrocytes producing corrected HBB proteins are highly desirable. Genome editing aided by CRISPR/Cas9 and other site-specific engineered nucleases offers promise to precisely correct a genetic mutation in the native genome without alterations in other parts of the human genome. Although making a sequence-specific nuclease to enhance correction of a specific HBB mutation by homology-directed repair (HDR) is becoming straightforward, targeting various HBB mutations of ß-thal is still challenging because individual guide RNA as well as a donor DNA template for HDR of each type of HBB gene mutation have to be selected and validated. Using human induced pluripotent stem cells (iPSCs) from two ß-thal patients with different HBB gene mutations, we devised and tested a universal strategy to achieve targeted insertion of the HBB cDNA in exon 1 of HBB gene using Cas9 and two validated guide RNAs. We observed that HBB protein production was restored in erythrocytes derived from iPSCs of two patients. This strategy of restoring functional HBB gene expression will be able to correct most types of HBB gene mutations in ß-thal and SCD. Stem Cells Translational Medicine 2018;7:87-97.


Subject(s)
Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Gene Editing/methods , Genetic Therapy/methods , Induced Pluripotent Stem Cells/cytology , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/therapy , CRISPR-Cas Systems/genetics , Cells, Cultured , Cellular Reprogramming Techniques , Erythrocytes/cytology , Female , Humans , Induced Pluripotent Stem Cells/transplantation , Male , Mutation/genetics
2.
Stem Cell Res ; 18: 57-59, 2017 01.
Article in English | MEDLINE | ID: mdl-28395806

ABSTRACT

Activating point mutations in the MPL gene encoding the thrombopoietin receptor are found in 3%-10% of essential thrombocythemia (ET) and myelofibrosis patients. Here, we report the derivation of induced pluripotent stem cells (iPSCs) from an ET patient with a heterozygous MPL V501L mutation. Peripheral blood CD34+ progenitor cells were reprogrammed by transient plasmid expression of OCT4, SOX2, KLF4, c-MYC plus BCL2L1 (BCL-xL) genes. The derived line M494 carries a MPL V501L mutation, displays typical iPSC morphology and characteristics, are pluripotent and karyotypically normal. Upon differentiation, the iPSCs are able to differentiate into cells derived from three germ layers.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Receptors, Thrombopoietin/genetics , Thrombocythemia, Essential/pathology , Antigens, CD34/metabolism , Base Sequence , Cell Differentiation , Cell Line , DNA Mutational Analysis , Embryoid Bodies/metabolism , Embryoid Bodies/pathology , Female , Genotype , Heterozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Karyotype , Kruppel-Like Factor 4 , Microscopy, Fluorescence , Polymorphism, Single Nucleotide , Stem Cells/cytology , Stem Cells/metabolism , Teratoma/metabolism , Teratoma/pathology , Thrombocythemia, Essential/genetics , Thrombocythemia, Essential/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Mol Ther ; 23(3): 570-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25418680

ABSTRACT

Efficient and precise genome editing is crucial for realizing the full research and therapeutic potential of human induced pluripotent stem cells (iPSCs). Engineered nucleases including CRISPR/Cas9 and transcription activator like effector nucleases (TALENs) provide powerful tools for enhancing gene-targeting efficiency. In this study, we investigated the relative efficiencies of CRISPR/Cas9 and TALENs in human iPSC lines for inducing both homologous donor-based precise genome editing and nonhomologous end joining (NHEJ)-mediated gene disruption. Significantly higher frequencies of NHEJ-mediated insertions/deletions were detected at several endogenous loci using CRISPR/Cas9 than using TALENs, especially at nonexpressed targets in iPSCs. In contrast, comparable efficiencies of inducing homologous donor-based genome editing were observed at disease-associated loci in iPSCs. In addition, we investigated the specificity of guide RNAs used in the CRISPR/Cas9 system in targeting disease-associated point mutations in patient-specific iPSCs. Using myeloproliferative neoplasm patient-derived iPSCs that carry an acquired JAK2-V617F point mutation and α1-antitrypsin (AAT) deficiency patient-derived iPSCs that carry an inherited Z-AAT point mutation, we demonstrate that Cas9 can specifically target either the mutant or the wild-type allele with little disruption at the other allele differing by a single nucleotide. Overall, our results demonstrate the advantages of the CRISPR/Cas9 system in allele-specific genome targeting and in NHEJ-mediated gene disruption.


Subject(s)
Alleles , Clustered Regularly Interspaced Short Palindromic Repeats , Genome, Human , Induced Pluripotent Stem Cells/metabolism , Quantitative Trait Loci , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , CRISPR-Associated Protein 9 , Cell Line , DNA End-Joining Repair , Endonucleases/genetics , Endonucleases/metabolism , Gene Expression , Humans , Induced Pluripotent Stem Cells/pathology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Molecular Sequence Data , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Recombinational DNA Repair , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism
5.
Stem Cells ; 32(1): 269-78, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24105986

ABSTRACT

Disease-specific induced pluripotent stem cells (iPSCs) provide an unprecedented opportunity to establish novel disease models and accelerate drug development using distinct tissue target cells generated from isogenic iPSC lines with and without disease-causing mutations. To realize the potential of iPSCs in modeling acquired diseases which are usually heterogeneous, we have generated multiple iPSC lines including two lines that are JAK2-wild-type and four lines homozygous for JAK2-V617F somatic mutation from a single polycythemia vera (PV) patient blood. In vitro differentiation of the same patient-derived iPSC lines have demonstrated the differential contributions of their parental hematopoietic clones to the abnormal erythropoiesis including the formation of endogenous erythroid colonies. This iPSC approach thus may provide unique and valuable insights into the genetic events responsible for disease development. To examine the potential of iPSCs in drug testing, we generated isogenic hematopoietic progenitors and erythroblasts from the same iPSC lines derived from PV patients and normal donors. Their response to three clinical JAK inhibitors, INCB018424 (Ruxolitinib), TG101348 (SAR302503), and the more recent CYT387 was evaluated. All three drugs similarly inhibited erythropoiesis from normal and PV iPSC lines containing the wild-type JAK2 genotype, as well as those containing a homozygous or heterozygous JAK2-V617F activating mutation that showed increased erythropoiesis without a JAK inhibitor. However, the JAK inhibitors had less inhibitory effect on the self-renewal of CD34+ hematopoietic progenitors. The iPSC-mediated disease modeling thus underlies the ineffectiveness of the current JAK inhibitors and provides a modeling system to develop better targeted therapies for the JAK2 mutated hematopoiesis.


Subject(s)
Erythroblasts/drug effects , Hematopoietic Stem Cells/drug effects , Induced Pluripotent Stem Cells/drug effects , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Cell Differentiation/drug effects , Erythroblasts/enzymology , Erythropoiesis/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/enzymology , Janus Kinase 2/genetics
6.
Stem Cell Res ; 11(3): 1103-16, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23973800

ABSTRACT

Large-scale production of human induced pluripotent stem cells (hiPSCs) by robust and economic methods has been one of the major challenges for translational realization of hiPSC technology. Here we demonstrate a scalable culture system for hiPSC expansion using the E8 chemically defined and xeno-free medium under either adherent or suspension conditions. To optimize suspension conditions guided by a computational simulation, we developed a method to efficiently expand hiPSCs as undifferentiated aggregates in spinner flasks. Serial passaging of two different hiPSC lines in the spinner flasks using the E8 medium preserved their normal karyotype and expression of undifferentiated state markers of TRA-1-60, SSEA4, OCT4, and NANOG. The hiPSCs cultured in spinner flasks for more than 10 passages not only could be remained pluripotent as indicated by in vitro and in vivo assays, but also could be efficiently induced toward mesodermal and hematopoietic differentiation. Furthermore, we established a xeno-free protocol of single-cell cryopreservation and recovery for the scalable production of hiPSCs in spinner flasks. This system is the first to enable an efficient scale-up bioprocess in completely xeno-free condition for the expansion and cryopreservation of hiPSCs with the quantity and quality compliant for clinical applications.


Subject(s)
Cell Culture Techniques , Culture Media/chemistry , Induced Pluripotent Stem Cells/cytology , Antigens, Surface/metabolism , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Proteoglycans/metabolism , Stage-Specific Embryonic Antigens/metabolism
7.
Exp Lung Res ; 37(10): 575-84, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22044313

ABSTRACT

To test the hypothesis that interleukin-6 (IL-6) contributes to the development of ventilator-associated lung injury (VALI), IL-6-deficient (IL6(-/-)) and wild-type control (WT) mice received intratracheal hydrochloric acid followed by randomization to mechanical ventilation (MV + IT HCl) or spontaneous ventilation (IT HCl). After 4 hours, injury was assessed by estimation of lung lavage protein concentration and total and differential cell counts, wet/dry lung weight ratio, pulmonary cell death, histologic inflammation score (LIS), and parenchymal myeloperoxidase (MPO) concentration. Vascular endothelial growth factor (VEGF) concentration was measured in lung lavage and homogenate, as IL-6 and stretch both regulate expression of this potent mediator of permeability. MV-induced increases in alveolar barrier dysfunction and lavage VEGF were attenuated in IL6(-/-) mice as compared with WT controls, whereas tissue VEGF concentration increased. The effects of IL-6 deletion on alveolar permeability and VEGF concentration were inflammation independent, as parenchymal MPO concentration, LIS, and lavage total and differential cell counts did not differ between WT and IL6(-/-) mice following MV + IT HCl. These data support a role for IL-6 in promoting VALI in this two-hit model. Strategies to interfere with IL-6 expression or signaling may represent important therapeutic targets to limit the injurious effects of MV in inflamed lungs.


Subject(s)
Capillary Permeability/physiology , Interleukin-6/metabolism , Lung Injury/metabolism , Lung Injury/pathology , Lung/metabolism , Animals , Bronchoalveolar Lavage Fluid , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/deficiency , Interleukin-6/genetics , Lung/pathology , Lung Injury/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Respiration, Artificial/methods , Vascular Endothelial Growth Factor A/metabolism , Ventilators, Mechanical
8.
PLoS One ; 6(11): e27449, 2011.
Article in English | MEDLINE | ID: mdl-22096573

ABSTRACT

RATIONALE: Semaphorin 3A (Sema3A) is a neural guidance cue that also mediates cell migration, proliferation and apoptosis, and inhibits branching morphogenesis. Because we have shown that genetic deletion of neuropilin-1, which encodes an obligatory Sema3A co-receptor, influences airspace remodeling in the smoke-exposed adult lung, we sought to determine whether genetic deletion of Sema3A altered distal lung structure. METHODS: To determine whether loss of Sema3A signaling influenced distal lung morphology, we compared pulmonary histology, distal epithelial cell morphology and maturation, and the balance between lung cell proliferation and death, in lungs from mice with a targeted genetic deletion of Sema3A (Sema3A(-/-)) and wild-type (Sema3A(+/+)) littermate controls. RESULTS: Genetic deletion of Sema3A resulted in significant perinatal lethality. At E17.5, lungs from Sema3A(-/-) mice had thickened septae and reduced airspace size. Distal lung epithelial cells had increased intracellular glycogen pools and small multivesicular and lamellar bodies with atypical ultrastructure, as well as reduced expression of type I alveolar epithelial cell markers. Alveolarization was markedly attenuated in lungs from the rare Sema3A(-/-) mice that survived the immediate perinatal period. Furthermore, Sema3A deletion was linked with enhanced postnatal alveolar septal cell death. CONCLUSIONS: These data suggest that Sema3A modulates distal pulmonary epithelial cell development and alveolar septation. Defining how Sema3A influences structural plasticity of the developing lung is a critical first step for determining if this pathway can be exploited to develop innovative strategies for repair after acute or chronic lung injury.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Lung/cytology , Morphogenesis/physiology , Organogenesis/physiology , Semaphorin-3A/metabolism , Animals , Cell Differentiation/genetics , Immunohistochemistry , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Morphogenesis/genetics , Organogenesis/genetics , Semaphorin-3A/genetics
9.
Am J Respir Crit Care Med ; 180(5): 396-406, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19520907

ABSTRACT

RATIONALE: Cigarette smoke (CS) exposure is an important risk factor for chronic obstructive pulmonary disease; however, not all smokers develop disease, suggesting that other factors influence disease development. OBJECTIVES: We sought to determine whether neuropilin-1 (Nrp1), an integral component of receptor complexes mediating alveolar septation and vascular development, was involved in maintenance of normal alveolar structure, and/or altered susceptibility to the effects of CS. METHODS: Transgenic mice were generated to achieve inducible lung-specific deletion of epithelial Nrp1. We determined whether conditional Nrp1 deletion altered airspace size, then compared the effects of chronic CS or filtered air exposure on airspace size, inflammation, and the balance between cell death and proliferation in conditionally Nrp1-deficient adult mice and littermate controls. Finally, we evaluated the effects of Nrp1 silencing on cell death after acute exposure of A549 cells to cigarette smoke extract or short chain ceramides. MEASUREMENTS AND MAIN RESULTS: Genetic deletion of epithelial Nrp1 in either postnatal or adult lungs resulted in a small increase in airspace size. More notably, both airspace enlargement and apoptosis of type I and type II alveolar epithelial cells were significantly enhanced following chronic CS exposure in conditionally Nrp1-deficient adult mice. Silencing of Nrp1 in A549 cells did not alter cell survival after vehicle treatment but significantly augmented apoptosis after exposure to cigarette smoke extract or ceramide. CONCLUSIONS: These data support a role for epithelial Nrp1 in the maintenance of normal alveolar structure and suggest that dysregulation of Nrp1 expression may promote epithelial cell death in response to CS exposure, thereby enhancing emphysema development.


Subject(s)
Emphysema/chemically induced , Emphysema/genetics , Gene Deletion , Neuropilin-1/genetics , Respiratory Mucosa/metabolism , Smoking/adverse effects , Animals , Apoptosis , Cell Proliferation , Disease Models, Animal , Emphysema/pathology , Mice , Mice, Transgenic , Pulmonary Alveoli/pathology , Random Allocation , Reference Values , Vascular Endothelial Growth Factor A/metabolism
10.
Biochem Biophys Res Commun ; 342(1): 153-63, 2006 Mar 31.
Article in English | MEDLINE | ID: mdl-16472767

ABSTRACT

Extracellular Ca(2+) concentration ([Ca(2+)](o)) regulates the functions of many cell types through a G protein-coupled [Ca(2+)](o)-sensing receptor (CaR). Whether the receptor is functionally expressed in vascular endothelial cells is largely unknown. In cultured human aortic endothelial cells (HAEC), RT-PCR yielded the expected 555-bp product corresponding to the CaR, and CaR protein was demonstrated by fluorescence immunostaining and Western blot. RT-PCR also demonstrated the expression in HAEC of alternatively spliced variants of the CaR lacking exon 5. Although stimulation of fura 2-loaded HAEC by several CaR agonists (high [Ca(2+)](o), neomycin, and gadolinium) failed to increase intracellular Ca(2+) concentration ([Ca(2+)](i)), the CaR agonist spermine stimulated an increase in [Ca(2+)](i) that was diminished in buffer without Ca(2+) and was abolished after depletion of an intracellular Ca(2+) pool with thapsigargin or after blocking IP(3)- and ryanodine receptor-mediated Ca(2+) release with xestospongin C and with high concentration ryanodine, respectively. Spermine stimulated an increase in DAF-FM fluorescence in HAEC, consistent with NO production. Both the increase in [Ca(2+)](i) and in NO production were reduced or absent in HAEC transfected with siRNA specifically targeted to the CaR. HAEC express a functional CaR that responds to the endogenous polyamine spermine with an increase in [Ca(2+)](i), primarily due to release of IP(3)- and ryanodine-sensitive intracellular Ca(2+) stores, leading to the production of NO. Expression of alternatively spliced variants of the CaR may result in the absence of a functional response to other known CaR agonists in HAEC.


Subject(s)
Aorta/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Receptors, Calcium-Sensing/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Nitric Oxide/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/genetics
11.
Biochem Biophys Res Commun ; 322(1): 68-73, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15313174

ABSTRACT

Increased endothelial ICAM-1 expression is found in normal aging and in atherosclerosis and is related to the chronic effects of oxidative stress. We examined the Ca(2+)-dependence of ICAM-1 mRNA expression in human aortic endothelial cells (HAEC) exposed to hypoxia/reoxygenation (H/R) as a model of oxidative stress. HAEC were exposed to glucose-free hypoxia (95% N(2)/5% CO(2)) for 60 min and were then reoxygenated (21% O(2)/5% CO(2)) and observed for up to 6h. Reactive oxygen species (ROS) generation was measured by dichlorofluorescein fluorescence and ICAM-1 mRNA was assessed by Northern blot. Upon reoxygenation after hypoxia, ROS production occurred in HAEC and was inhibited by diphenyleneiodonium and by polyethylene glycol-catalase, suggesting the involvement of NADPH oxidase-derived hydrogen peroxide. Hypoxia alone did not increase either ROS production or ICAM-1 mRNA levels, but a 2.5-fold increase in ICAM-1 mRNA was noted by 30 min of reoxygenation. This was not observed in Ca(2+)-free buffer or in cells treated with diphenyleneiodonium. Thus, H/R upregulates ICAM-1 mRNA in HAEC by a Ca(2+)- and ROS-dependent mechanism. Characterizing the signaling pathways involved in H/R-induced adhesion molecule expression may result in a better understanding of the vascular biology of normal aging and the pathobiology of atherosclerosis.


Subject(s)
Calcium/metabolism , Cell Hypoxia/physiology , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Oxidative Stress/physiology , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Aorta/metabolism , Cells, Cultured , Gene Expression Regulation/physiology , Humans , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...