Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Elife ; 92020 02 03.
Article in English | MEDLINE | ID: mdl-32011235

ABSTRACT

Generation of skeletal muscle cells with human pluripotent stem cells (hPSCs) opens new avenues for deciphering essential, but poorly understood aspects of transcriptional regulation in human myogenic specification. In this study, we characterized the transcriptional landscape of distinct human myogenic stages, including OCT4::EGFP+ pluripotent stem cells, MSGN1::EGFP+ presomite cells, PAX7::EGFP+ skeletal muscle progenitor cells, MYOG::EGFP+ myoblasts, and multinucleated myotubes. We defined signature gene expression profiles from each isolated cell population with unbiased clustering analysis, which provided unique insights into the transcriptional dynamics of human myogenesis from undifferentiated hPSCs to fully differentiated myotubes. Using a knock-out strategy, we identified TWIST1 as a critical factor in maintenance of human PAX7::EGFP+ putative skeletal muscle progenitor cells. Our data revealed a new role of TWIST1 in human skeletal muscle progenitors, and we have established a foundation to identify transcriptional regulations of human myogenic ontogeny (online database can be accessed in http://www.myogenesis.net/).


Subject(s)
Gene Expression Regulation, Developmental/genetics , Muscle Development/genetics , Muscle, Skeletal/cytology , Nuclear Proteins , Pluripotent Stem Cells/metabolism , Twist-Related Protein 1 , Cell Line , Gene Knockout Techniques , Humans , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
2.
Cell ; 172(1-2): 191-204.e10, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29224778

ABSTRACT

Hematopoietic stem cell transplantation is a potential curative therapy for malignant and nonmalignant diseases. Improving the efficiency of stem cell collection and the quality of the cells acquired can broaden the donor pool and improve patient outcomes. We developed a rapid stem cell mobilization regimen utilizing a unique CXCR2 agonist, GROß, and the CXCR4 antagonist AMD3100. A single injection of both agents resulted in stem cell mobilization peaking within 15 min that was equivalent in magnitude to a standard multi-day regimen of granulocyte colony-stimulating factor (G-CSF). Mechanistic studies determined that rapid mobilization results from synergistic signaling on neutrophils, resulting in enhanced MMP-9 release, and unexpectedly revealed genetic polymorphisms in MMP-9 that alter activity. This mobilization regimen results in preferential trafficking of stem cells that demonstrate a higher engraftment efficiency than those mobilized by G-CSF. Our studies suggest a potential new strategy for the rapid collection of an improved hematopoietic graft.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Adult , Animals , Benzylamines , Chemokine CXCL2/pharmacology , Cyclams , Female , Hematopoietic Stem Cells/drug effects , Heterocyclic Compounds/pharmacology , Humans , Male , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred ICR , Polymorphism, Genetic
3.
Stem Cells Transl Med ; 4(4): 320-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25742692

ABSTRACT

Reprogramming human adult blood mononuclear cells (MNCs) cells by transient plasmid expression is becoming increasingly popular as an attractive method for generating induced pluripotent stem (iPS) cells without the genomic alteration caused by genome-inserting vectors. However, its efficiency is relatively low with adult MNCs compared with cord blood MNCs and other fetal cells and is highly variable among different adult individuals. We report highly efficient iPS cell derivation under clinically compliant conditions via three major improvements. First, we revised a combination of three EBNA1/OriP episomal vectors expressing five transgenes, which increased reprogramming efficiency by ≥10-50-fold from our previous vectors. Second, human recombinant vitronectin proteins were used as cell culture substrates, alleviating the need for feeder cells or animal-sourced proteins. Finally, we eliminated the previously critical step of manually picking individual iPS cell clones by pooling newly emerged iPS cell colonies. Pooled cultures were then purified based on the presence of the TRA-1-60 pluripotency surface antigen, resulting in the ability to rapidly expand iPS cells for subsequent applications. These new improvements permit a consistent and reliable method to generate human iPS cells with minimal clonal variations from blood MNCs, including previously difficult samples such as those from patients with paroxysmal nocturnal hemoglobinuria. In addition, this method of efficiently generating iPS cells under feeder-free and xeno-free conditions allows for the establishment of clinically compliant iPS cell lines for future therapeutic applications.


Subject(s)
Cell Culture Techniques , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Adult , Blood Cells/cytology , Culture Media, Serum-Free , Feeder Cells/cytology , Flow Cytometry , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/transplantation , Leukocytes, Mononuclear/cytology , Vitronectin/administration & dosage
4.
PLoS One ; 10(1): e0116933, 2015.
Article in English | MEDLINE | ID: mdl-25635918

ABSTRACT

Induced pluripotent stem (iPS) cells are at the forefront of research in regenerative medicine and are envisaged as a source for personalized tissue repair and cell replacement therapy. Here, we demonstrate for the first time that oligodendrocyte progenitors (OPs) can be derived from iPS cells generated using either an episomal, non-integrating plasmid approach or standard integrating retroviruses that survive and differentiate into mature oligodendrocytes after early transplantation into the injured spinal cord. The efficiency of OP differentiation in all 3 lines tested ranged from 40% to 60% of total cells, comparable to those derived from human embryonic stem cells. iPS cell lines derived using episomal vectors or retroviruses generated a similar number of early neural progenitors and glial progenitors while the episomal plasmid-derived iPS line generated more OPs expressing late markers O1 and RIP. Moreover, we discovered that iPS-derived OPs (iPS-OPs) engrafted 24 hours following a moderate contusive spinal cord injury (SCI) in rats survived for approximately two months and that more than 70% of the transplanted cells differentiated into mature oligodendrocytes that expressed myelin associated proteins. Transplanted OPs resulted in a significant increase in the number of myelinated axons in animals that received a transplantation 24 h after injury. In addition, nearly a 5-fold reduction in cavity size and reduced glial scarring was seen in iPS-treated groups compared to the control group, which was injected with heat-killed iPS-OPs. Although further investigation is needed to understand the mechanisms involved, these results provide evidence that patient-specific, iPS-derived OPs can survive for three months and improve behavioral assessment (BBB) after acute transplantation into SCI. This is significant as determining the time in which stem cells are injected after SCI may influence their survival and differentiation capacity.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Animals , Axons/physiology , Cell Differentiation , Cell Survival , Cells, Cultured , Early Medical Intervention , Female , Humans , Motor Activity , Myelin Sheath/physiology , Nerve Regeneration , Oligodendroglia/physiology , Rats, Inbred Lew , Recovery of Function , Spinal Cord/pathology , Spinal Cord/physiopathology , Treatment Outcome
5.
Stem Cells ; 32(2): 349-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23765875

ABSTRACT

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) shares much similarity to the cancer initiation process, and the molecular mechanisms underlying both processes remain to be elucidated. Here, we report that a tumor- or embryonic stem cell-specific Ras gene ERas, which encodes a constitutively active form of GTPase, and its downstream Phosphoinositide-3 kinase/Akt signaling pathway are important facilitators for the somatic reprogramming process. We found that overexpression of ERas retrovirally enhanced mouse iPSC induction while ERas knockdown repressed it. Modulation of Akt signaling by genetic or chemical means greatly impacted the reprogramming efficiency. Forced expression of a constitutively active Akt1 gene could rescue the reduced efficiency resulting from ERas knockdown, and point-mutation analyses further revealed that ERas is tightly coupled with Akt signaling to enhance reprogramming. Mechanistically, the forkhead transcription factor FoxO1 can function as a barrier to the iPSC induction, and the inactivation of FoxO1 by Akt-dependent phosphorylation largely accounts for the enhancing effect of ERas-Akt signaling on reprogramming. Collectively, these results unravel the significance of the ERas-Akt-FoxO1 signaling axis in iPSC generation, suggesting a possibly shared molecular basis for both somatic reprogramming and cancer initiation.


Subject(s)
Embryonic Stem Cells/metabolism , Forkhead Transcription Factors/genetics , Oncogene Protein p21(ras)/genetics , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Forkhead Box Protein O1 , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics
6.
Cell Stem Cell ; 13(3): 261-2, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-24012365

ABSTRACT

While most factors used as reprogramming transgenes can be replaced by other means, Oct4 has remained essential until now. Three recent papers have now broken this barrier through the use of opposing lineage specifying transgenes and chemical modulation, thus signifying a milestone in advancing our understanding of pluripotency induction.


Subject(s)
Cell Dedifferentiation , Cell Engineering/methods , Cellular Reprogramming/drug effects , Fibroblasts/drug effects , Fibroblasts/physiology , GATA3 Transcription Factor/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/physiology , Small Molecule Libraries/pharmacology , Transcription Factors/metabolism , Animals , Humans
7.
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
9.
Nat Protoc ; 7(11): 2013-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23080273

ABSTRACT

Several human postnatal somatic cell types have been successfully reprogrammed to induced pluripotent stem cells (iPSCs). Blood mononuclear cells (MNCs) offer several advantages compared with other cell types. They are easily isolated from umbilical cord blood (CB) or adult peripheral blood (PB), and can be used fresh or after freezing. A short culture allows for more efficient reprogramming, with iPSC colonies forming from blood MNCs in 14 d, compared with 28 d for age-matched fibroblastic cells. The advantages of briefly cultured blood MNCs may be due to favorable epigenetic profiles and gene expression patterns. Blood cells from adults, especially nonlymphoid cells that are replenished frequently from intermittently activated blood stem cells, are short-lived in vivo and may contain less somatic mutations than skin fibroblasts, which are more exposed to environmental mutagens over time. We describe here a detailed, validated protocol for effective generation of integration-free human iPSCs from blood MNCs by plasmid vectors.


Subject(s)
Cell Culture Techniques , Induced Pluripotent Stem Cells/cytology , Leukocytes, Mononuclear/cytology , Genetic Vectors , Humans , Plasmids/genetics , Transfection
10.
Int J Hematol ; 95(6): 601-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22619021

ABSTRACT

Postnatal hematopoietic stem cells (HSCs) from umbilical cord blood and adult marrow/blood have been successfully used for treating various human diseases in the past several decades. However, the availability of optimal numbers of HSCs from autologous patients or allogeneic donors with adequate match remains a great barrier to improve and extend HSC and marrow transplantation to more needing patients. In addition, the inability to expand functional human HSCs to sufficient quantity in the laboratory has hindered our research and understanding of human HSCs and hematopoiesis. Recent development in reprogramming technology has provided patient-specific pluripotent stem cells (iPSCs) as a powerful enabling tool for modeling disease and developing therapeutics. Studies have demonstrated the potential of human iPSCs, which can be expanded exponentially and amenable for genome engineering, for using in modeling both inherited and acquired blood diseases. Proof-of-principle studies have also shown the feasibility of iPSCs in gene and cell therapy. Here, we review the recent development in iPSC-based blood disease modeling, and discuss the unsolved issues and challenges in this new and promising field.


Subject(s)
Hematologic Diseases/therapy , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Animals , Cell Differentiation , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/metabolism
11.
Cell Stem Cell ; 10(3): 337-44, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22385660

ABSTRACT

The utility of induced pluripotent stem cells (iPSCs) as models to study diseases and as sources for cell therapy depends on the integrity of their genomes. Despite recent publications of DNA sequence variations in the iPSCs, the true scope of such changes for the entire genome is not clear. Here we report the whole-genome sequencing of three human iPSC lines derived from two cell types of an adult donor by episomal vectors. The vector sequence was undetectable in the deeply sequenced iPSC lines. We identified 1,058-1,808 heterozygous single-nucleotide variants (SNVs), but no copy-number variants, in each iPSC line. Six to twelve of these SNVs were within coding regions in each iPSC line, but ~50% of them are synonymous changes and the remaining are not selectively enriched for known genes associated with cancers. Our data thus suggest that episome-mediated reprogramming is not inherently mutagenic during integration-free iPSC induction.


Subject(s)
Genetic Variation , Induced Pluripotent Stem Cells , Plasmids/genetics , Cell Line , Cellular Reprogramming/genetics , Genetic Vectors/genetics , Humans , Leukocytes, Mononuclear , Microarray Analysis , Sequence Analysis, DNA
12.
Stem Cells Dev ; 21(12): 2298-311, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22225458

ABSTRACT

Derivation of pluripotent stem cells (iPSCs) induced from somatic cell types and the subsequent genetic modifications of disease-specific or patient-specific iPSCs are crucial steps in their applications for disease modeling as well as future cell and gene therapies. Conventional procedures of these processes require co-culture with primary mouse embryonic fibroblasts (MEFs) to support self-renewal and clonal growth of human iPSCs as well as embryonic stem cells (ESCs). However, the variability of MEF quality affects the efficiencies of all these steps. Furthermore, animal sourced feeders may hinder the clinical applications of human stem cells. In order to overcome these hurdles, we established immortalized human feeder cell lines by stably expressing human telomerase reverse transcriptase, Wnt3a, and drug resistance genes in adult mesenchymal stem cells. Here, we show that these immortalized human feeders support efficient derivation of virus-free, integration-free human iPSCs and long-term expansion of human iPSCs and ESCs. Moreover, the drug-resistance feature of these feeders also supports nonviral gene transfer and expression at a high efficiency, mediated by piggyBac DNA transposition. Importantly, these human feeders exhibit superior ability over MEFs in supporting homologous recombination-mediated gene targeting in human iPSCs, allowing us to efficiently target a transgene into the AAVS1 safe harbor locus in recently derived integration-free iPSCs. Our results have great implications in disease modeling and translational applications of human iPSCs, as these engineered human cell lines provide a more efficient tool for genetic modifications and a safer alternative for supporting self-renewal of human iPSCs and ESCs.


Subject(s)
Cell Culture Techniques , Feeder Cells/physiology , Genetic Engineering , Induced Pluripotent Stem Cells/physiology , Alkaline Phosphatase/metabolism , Animals , Antigens, Differentiation/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Embryonic Stem Cells/metabolism , Feeder Cells/metabolism , Female , Genetic Loci , Homologous Recombination , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Karyotype , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Neoplasms, Experimental/pathology , Telomerase/genetics , Teratoma/pathology , Transduction, Genetic , Wnt3A Protein/biosynthesis , Wnt3A Protein/genetics
13.
Blood ; 117(21): 5561-72, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21411759

ABSTRACT

We have developed induced pluripotent stem cells (iPSCs) from a patient with X-linked chronic granulomatous disease (X-CGD), a defect of neutrophil microbicidal reactive oxygen species (ROS) generation resulting from gp91(phox) deficiency. We demonstrated that mature neutrophils differentiated from X-CGD iPSCs lack ROS production, reproducing the pathognomonic CGD cellular phenotype. Targeted gene transfer into iPSCs, with subsequent selection and full characterization to ensure no off-target changes, holds promise for correction of monogenic diseases without the insertional mutagenesis caused by multisite integration of viral or plasmid vectors. Zinc finger nuclease-mediated gene targeting of a single-copy gp91(phox) therapeutic minigene into one allele of the "safe harbor" AAVS1 locus in X-CGD iPSCs without off-target inserts resulted in sustained expression of gp91(phox) and substantially restored neutrophil ROS production. Our findings demonstrate how precise gene targeting may be applied to correction of X-CGD using zinc finger nuclease and patient iPSCs.


Subject(s)
Granulomatous Disease, Chronic/therapy , Induced Pluripotent Stem Cells/metabolism , Membrane Glycoproteins/physiology , NADPH Oxidases/deficiency , Neutrophils/enzymology , Adult , Animals , Blotting, Southern , Blotting, Western , Bone Marrow/metabolism , Cell Differentiation , DNA Fingerprinting , Dependovirus/genetics , Flow Cytometry , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/pathology , Humans , Karyotyping , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, Nude , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/physiology , Phagocytosis , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Zinc Fingers
14.
Cell Res ; 21(3): 518-29, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21243013

ABSTRACT

To identify accessible and permissive human cell types for efficient derivation of induced pluripotent stem cells (iPSCs), we investigated epigenetic and gene expression signatures of multiple postnatal cell types such as fibroblasts and blood cells. Our analysis suggested that newborn cord blood (CB) and adult peripheral blood (PB) mononuclear cells (MNCs) display unique signatures that are closer to iPSCs and human embryonic stem cells (ESCs) than age-matched fibroblasts to iPSCs/ESCs, thus making blood MNCs an attractive cell choice for the generation of integration-free iPSCs. Using an improved EBNA1/OriP plasmid expressing 5 reprogramming factors, we demonstrated highly efficient reprogramming of briefly cultured blood MNCs. Within 14 days of one-time transfection by one plasmid, up to 1000 iPSC-like colonies per 2 million transfected CB MNCs were generated. The efficiency of deriving iPSCs from adult PB MNCs was approximately 50-fold lower, but could be enhanced by inclusion of a second EBNA1/OriP plasmid for transient expression of additional genes such as SV40 T antigen. The duration of obtaining bona fide iPSC colonies from adult PB MNCs was reduced to half (∼14 days) as compared to adult fibroblastic cells (28-30 days). More than 9 human iPSC lines derived from PB or CB blood cells are extensively characterized, including those from PB MNCs of an adult patient with sickle cell disease. They lack V(D)J DNA rearrangements and vector DNA after expansion for 10-12 passages. This facile method of generating integration-free human iPSCs from blood MNCs will accelerate their use in both research and future clinical applications.


Subject(s)
Blood Cells/cytology , Epigenesis, Genetic , Gene Expression Profiling , Induced Pluripotent Stem Cells/metabolism , Plasmids/metabolism , Antigens, CD34/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cellular Reprogramming , Fetal Blood/cytology , Fibroblasts/cytology , Hematopoietic Stem Cells/metabolism , Humans , Plasmids/genetics , RNA-Binding Proteins
15.
Stem Cells ; 28(4): 713-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20201064

ABSTRACT

We report here that butyrate, a naturally occurring fatty acid commonly used as a nutritional supplement and differentiation agent, greatly enhances the efficiency of induced pluripotent stem (iPS) cell derivation from human adult or fetal fibroblasts. After transient butyrate treatment, the iPS cell derivation efficiency is enhanced by 15- to 51-fold using either retroviral or piggyBac transposon vectors expressing 4 to 5 reprogramming genes. Butyrate stimulation is more remarkable (>100- to 200-fold) on reprogramming in the absence of either KLF4 or MYC transgene. Butyrate treatment did not negatively affect properties of iPS cell lines established by either 3 or 4 retroviral vectors or a single piggyBac DNA transposon vector. These characterized iPS cell lines, including those derived from an adult patient with sickle cell disease by either the piggyBac or retroviral vectors, show normal karyotypes and pluripotency. To gain insights into the underlying mechanisms of butyrate stimulation, we conducted genome-wide gene expression and promoter DNA methylation microarrays and other epigenetic analyses on established iPS cells and cells from intermediate stages of the reprogramming process. By days 6 to 12 during reprogramming, butyrate treatment enhanced histone H3 acetylation, promoter DNA demethylation, and the expression of endogenous pluripotency-associated genes, including DPPA2, whose overexpression partially substitutes for butyrate stimulation. Thus, butyrate as a cell permeable small molecule provides a simple tool to further investigate molecular mechanisms of cellular reprogramming. Moreover, butyrate stimulation provides an efficient method for reprogramming various human adult somatic cells, including cells from patients that are more refractory to reprogramming.


Subject(s)
Butyrates/pharmacology , Cell Culture Techniques/methods , Epigenesis, Genetic/drug effects , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Base Sequence , Cells, Cultured , Cellular Senescence , DNA Transposable Elements , Genetic Vectors/genetics , Humans , Kruppel-Like Factor 4 , Sequence Alignment
16.
Methods Mol Biol ; 636: 191-205, 2010.
Article in English | MEDLINE | ID: mdl-20336524

ABSTRACT

This chapter describes the methods we use to derive and characterize human induced pluripotent stem (iPS) cells. We describe in order, first our culture techniques for the starting fibroblast populations and methods for retrovirus preparation and concentration. Subsequently, a detailed iPS derivation protocol suitable for human fibroblast populations is discussed using standard retroviral vectors expressing the classic four or three reprogramming genes. Finally, we elaborate a robust technique for monitoring and identification of potential iPS cells through live staining of reprogrammed cells. We also outline steps for characterization of the resulting iPS cell lines.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/physiology , Animals , Cell Culture Techniques/instrumentation , Cell Separation/methods , Cells, Cultured , Cellular Reprogramming , Fibroblasts/cytology , Fibroblasts/physiology , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Retroviridae/genetics , Retroviridae/metabolism
17.
Cell Stem Cell ; 5(1): 97-110, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19540188

ABSTRACT

We report here homologous recombination (HR)-mediated gene targeting of two different genes in human iPS cells (hiPSCs) and human ES cells (hESCs). HR-mediated correction of a chromosomally integrated mutant GFP reporter gene reaches efficiencies of 0.14%-0.24% in both cell types transfected by donor DNA with plasmids expressing zinc finger nucleases (ZFNs). Engineered ZFNs that induce a sequence-specific double-strand break in the GFP gene enhanced HR-mediated correction by > 1400-fold without detectable alterations in stem cell karyotypes or pluripotency. Efficient HR-mediated insertional mutagenesis was also achieved at the endogenous PIG-A locus, with a > 200-fold enhancement by ZFNs targeted to that gene. Clonal PIG-A null hESCs and iPSCs with normal karyotypes were readily obtained. The phenotypic and biological defects were rescued by PIG-A transgene expression. Our study provides the first demonstration of HR-mediated gene targeting in hiPSCs and shows the power of ZFNs for inducing specific genetic modifications in hiPSCs, as well as hESCs.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Targeting/methods , Membrane Proteins/genetics , Pluripotent Stem Cells/metabolism , Cell Line , Deoxyribonucleases/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/metabolism , Zinc Fingers
18.
Curr Biol ; 14(24): 2208-16, 2004 Dec 29.
Article in English | MEDLINE | ID: mdl-15620647

ABSTRACT

BACKGROUND: Phagocytosis of cells undergoing apoptosis is essential during development, cellular turnover, and wound healing. Failure to promptly clear apoptotic cells has been linked to autoimmune disorders. C. elegans CED-12 and mammalian ELMO are evolutionarily conserved scaffolding proteins that play a critical role in engulfment from worm to human. ELMO functions together with Dock180 (a guanine nucleotide exchange factor for Rac) to mediate Rac-dependent cytoskeletal reorganization during engulfment and cell migration. However, the components upstream of ELMO and Dock180 during engulfment remain elusive. RESULTS: Here, we define a conserved signaling module involving the small GTPase RhoG and its exchange factor TRIO, which functions upstream of ELMO/Dock180/Rac during engulfment. Complementary studies in C. elegans show that MIG-2 (which we identify as the homolog of mammalian RhoG) and UNC-73 (the TRIO homolog) also regulate corpse clearance in vivo, upstream of CED-12. At the molecular level, we identify a novel set of evolutionarily conserved Armadillo (ARM) repeats within CED-12/ELMO that mediate an interaction with activated MIG-2/RhoG; this, in turn, promotes Dock180-mediated Rac activation and cytoskeletal reorganization. CONCLUSIONS: The combination of in vitro and in vivo studies presented here identify two evolutionarily conserved players in engulfment, TRIO/UNC73 and RhoG/MIG-2, and the TRIO --> RhoG signaling module is linked by ELMO/CED-12 to Dock180-dependent Rac activation during engulfment. This work also identifies ARM repeats within CED-12/ELMO and their role in linking RhoG and Rac, two GTPases that function in tandem during engulfment.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cytoskeleton/metabolism , GTP Phosphohydrolases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Nerve Tissue Proteins/metabolism , Phagocytosis/physiology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Caenorhabditis elegans , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Humans , Repetitive Sequences, Nucleic Acid/genetics , rho GTP-Binding Proteins
19.
Science ; 302(5650): 1563-6, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14645848

ABSTRACT

During apoptosis, phosphatidylserine, which is normally restricted to the inner leaflet of the plasma membrane, is exposed on the surface of apoptotic cells and has been suggested to act as an "eat-me" signal to trigger phagocytosis. It is unclear how phagocytes recognize phosphatidylserine. Recently, a putative phosphatidylserine receptor (PSR) was identified and proposed to mediate recognition of phosphatidylserine and phagocytosis. We report that psr-1, the Caenorhabditis elegans homolog of PSR, is important for cell corpse engulfment. In vitro PSR-1 binds preferentially phosphatidylserine or cells with exposed phosphatidylserine. In C. elegans, PSR-1 acts in the same cell corpse engulfment pathway mediated by intracellular signaling molecules CED-2 (homologous to the human CrkII protein), CED-5 (DOCK180), CED-10 (Rac GTPase), and CED-12 (ELMO), possibly through direct interaction with CED-5 and CED-12. Our findings suggest that PSR-1 is likely an upstream receptor for the signaling pathway containing CED-2, CED-5, CED-10, and CED-12 proteins and plays an important role in recognizing phosphatidylserine during phagocytosis.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Carrier Proteins/metabolism , Cytoskeletal Proteins , Membrane Proteins/metabolism , Phagocytosis , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Embryonic Development , Humans , Jumonji Domain-Containing Histone Demethylases , Membrane Proteins/genetics , Molecular Sequence Data , Mutation , Phosphatidylserines/metabolism , Protein Binding , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Signal Transduction , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...