Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Stem Cells ; 40(4): 397-410, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35385105

ABSTRACT

Somatic cell reprogramming proceeds through a series of events to generate induced pluripotent stem cells (iPSCs). The early stage of reprogramming of mouse embryonic fibroblasts is characterized by rapid cell proliferation and morphological changes, which are accompanied by downregulation of mesenchyme-associated genes. However, the functional relevance of their downregulation to reprogramming remains poorly defined. In this study, we have screened transcriptional regulators that are downregulated immediately upon reprogramming, presumably through direct targeting by reprogramming factors. To test if these transcriptional regulators impact reprogramming when expressed continuously, we generated an expression vector that harbors human cytomegalovirus upstream open reading frame 2 (uORF2), which reduces translation to minimize the detrimental effect of an expressed protein. Screening of transcriptional regulators with this expression vector revealed that downregulation of (odd-skipped related 2 [Osr2]) is crucial for efficient reprogramming. Using a cell-based model for epithelial-mesenchymal transition (EMT), we show that Osr2 is a novel EMT regulator that acts through induction of transforming growth factor-ß (TGF-ß) signaling. During reprogramming, Osr2 downregulation not only diminishes TGF-ß signaling but also allows activation of Wnt signaling, thus promoting mesenchymal-epithelial transition (MET) toward acquisition of pluripotency. Our results illuminate the functional significance of Osr2 downregulation in erasing the mesenchymal phenotype at an early stage of somatic cell reprogramming.


Subject(s)
Epithelial-Mesenchymal Transition , Induced Pluripotent Stem Cells , Animals , Cellular Reprogramming/genetics , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
2.
Mol Ther ; 30(2): 534-549, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34628050

ABSTRACT

We generated dual-antigen receptor (DR) T cells from induced pluripotent stem cells (iPSCs) to mitigate tumor antigen escape. These cells were engineered to express a chimeric antigen receptor (CAR) for the antigen cell surface latent membrane protein 1 (LMP1; LMP1-CAR) and a T cell receptor directed to cell surface latent membrane protein 2 (LMP2), in association with human leucocyte antigen A24, to treat therapy-refractory Epstein-Barr virus-associated lymphomas. We introduced LMP1-CAR into iPSCs derived from LMP2-specific cytotoxic T lymphocytes (CTLs) to generate rejuvenated CTLs (rejTs) active against LMP1 and LMP2, or DRrejTs. All DRrejT-treated mice survived >100 days. Furthermore, DRrejTs rejected follow-up inocula of lymphoma cells, demonstrating that DRrejTs persisted long-term. We also demonstrated that DRrejTs targeting CD19 and LMP2 antigens exhibited a robust tumor suppressive effect and conferred a clear survival advantage. Co-operative antitumor effect and in vivo persistence, with unlimited availability of DRrejT therapy, will provide powerful and sustainable T cell immunotherapy.


Subject(s)
Epstein-Barr Virus Infections , Induced Pluripotent Stem Cells , Lymphoma , Receptors, Chimeric Antigen , Animals , Cell- and Tissue-Based Therapy , Herpesvirus 4, Human/genetics , Immunotherapy, Adoptive , Induced Pluripotent Stem Cells/metabolism , Lymphoma/genetics , Lymphoma/therapy , Mice , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes, Cytotoxic , Viral Matrix Proteins/genetics
3.
Biochem Biophys Res Commun ; 604: 22-29, 2022 05 14.
Article in English | MEDLINE | ID: mdl-35279442

ABSTRACT

OBJECTIVE: Cluster of differentiation 81 (CD81) is a tetraspanin membrane protein consisting of 4 transmembrane domains and 2 outer membrane loops. CD81 inhibition is a potential treatment for rheumatoid arthritis (RA). Here, we investigated the therapeutic effects of the cytoplasmic RNA vector expressing anti-CD81 antibodies (the anti-CD81 vector) on the ankle joint synovium in collagen-induced arthritis (CIA) rats. METHODS: Body weight, paw volume, and clinical scores were measured on days 0, 7, and 10 and daily thereafter. On day 28, the ankle joints of the rats were removed and stained with haematoxylin, eosin, and Safranin O. Arthritic changes such as inflammatory cell infiltration, synovial proliferation, articular cartilage destruction, and bone erosion were evaluated by histological scoring. RESULTS: Symptom onset was delayed in the right lower limbs of the rats administered the cytoplasmic RNA vector (CIA + anti-CD81) compared with that in the control group (CIA + control). The CIA + anti-CD81 rats were heavier than the CIA + control rats. The paw volume and clinical scores were significantly lower in the CIA + anti-CD81 than in the CIA + control. The histological scores indicated significantly milder manifestations of RA in the CIA + anti-CD81 than in the CIA + control. CONCLUSIONS: Administration of the cytoplasmic RNA vector expressing anti-CD81 antibodies suppressed arthritis and joint destruction in CIA rats. Our findings suggest that the cytoplasmic RNA vector can be used to treat RA.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Cartilage, Articular , Animals , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/pathology , Cartilage, Articular/metabolism , RNA/metabolism , Rats , Synovial Membrane/pathology
4.
Genes Cells ; 26(11): 891-904, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34480399

ABSTRACT

The Sendai virus vector has received a lot of attention due to its broad tropism for mammalian cells. As a result of efforts for genetic studies based on a mutant virus, we can now express more than 10 genes of up to 13.5 kilo nucleotides in a single vector with high protein expression efficiency. To prove this benefit, we examined the efficacy of the novel ribonucleic acid (RNA) virus vector harboring the human iduronate-2-sulfatase (IDS) gene with 1,653 base pairs, a causative gene for mucopolysaccharidosis type II, also known as a disorder of lysosomal storage disorders. As expected, this novel RNA vector with the human IDS gene exhibited its marked expression as determined by the expression of enhanced green fluorescent protein and IDS enzyme activity. While these cells exhibited a normal growth rate, the BHK-21 transformant cells stably expressing the human IDS gene persistently generated an active human IDS enzyme extracellularly. The human IDS protein produced failed to be incorporated into the lysosome when cells were pretreated with mannose-6-phosphate, demonstrating that this human IDS enzyme has potential for therapeutic use by cross-correction. These results suggest that our novel RNA vector may be applicable for further clinical settings.


Subject(s)
Iduronate Sulfatase , Mucopolysaccharidosis II , RNA Viruses , Animals , Humans , Iduronic Acid , Lysosomes
5.
Int J Mol Sci ; 23(15)2022 Jul 24.
Article in English | MEDLINE | ID: mdl-35897729

ABSTRACT

Sulfatases are enzymes that catalyze the removal of sulfate from biological substances, an essential process for the homeostasis of the body. They are commonly activated by the unusual amino acid formylglycine, which is formed from cysteine at the catalytic center, mediated by a formylglycine-generating enzyme as a post-translational modification. Sulfatases are expressed in various cellular compartments such as the lysosome, the endoplasmic reticulum, and the Golgi apparatus. The substrates of mammalian sulfatases are sulfolipids, glycosaminoglycans, and steroid hormones. These enzymes maintain neuronal function in both the central and the peripheral nervous system, chondrogenesis and cartilage in the connective tissue, detoxification from xenobiotics and pharmacological compounds in the liver, steroid hormone inactivation in the placenta, and the proper regulation of skin humidification. Human sulfatases comprise 17 genes, 10 of which are involved in congenital disorders, including lysosomal storage disorders, while the function of the remaining seven is still unclear. As for the genes responsible for pathogenesis, therapeutic strategies have been developed. Enzyme replacement therapy with recombinant enzyme agents and gene therapy with therapeutic transgenes delivered by viral vectors are administered to patients. In this review, the biochemical substrates, disease manifestation, and therapy for sulfatases are summarized.


Subject(s)
Lysosomal Storage Diseases , Sulfatases , Animals , Cysteine/metabolism , Female , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Mammals/metabolism , Pregnancy , Protein Processing, Post-Translational , Proteins/metabolism , Sulfatases/genetics , Sulfatases/metabolism
6.
Med Mol Morphol ; 55(3): 174-186, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35461467

ABSTRACT

Cleidocranial dysplasia (CCD) is a hereditary disorder associated with skeletal dysplasia and dental abnormalities. CCD arises from heterozygous loss of function mutations in the Runt-related transcription factor 2 (RUNX2) gene. Osteoporosis is often observed in CCD patients and conventional vitamin D supplementation is recommended. However, sufficient evidences have not been presented yet. This study investigated the role of RUNX2 in osteoblastic differentiation and sought to identify potential target genes for the treatment of osteoporosis associated with CCD, using induced pluripotent stem cell (iPSC) technology. We successfully established Runx2-/-, Runx2+/- and wild-type miPSCs from litter-matched mice and found poor Vdr expression in Runx2-/-cells. Significant down-regulation of osteoblastic differentiation in Runx2-/- miPSCs was observed. Gene expression array revealed unexpected results such as remarkable increase of Rankl expression and decrease of Vdr in Runx2-/- cells. Insufficient response to vitamin D in Runx2-/- cells was also observed. Our results suggest that RUNX2 functions as a regulator of Rankl and Vdr and thereby controls bone density. These findings also suggest that conventional vitamin D supplementation may not be as effective as previously expected, in the treatment of osteoporosis associated with CCD, and that inhibiting RANKL function might be worth considering as an alternative treatment strategy.


Subject(s)
Cleidocranial Dysplasia , Core Binding Factor Alpha 1 Subunit , Induced Pluripotent Stem Cells , Osteoporosis , Vitamin D , Animals , Cell Differentiation , Cleidocranial Dysplasia/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoporosis/drug therapy , Osteoporosis/genetics , Vitamin D/pharmacology
7.
Mol Reprod Dev ; 88(6): 395-404, 2021 06.
Article in English | MEDLINE | ID: mdl-34010985

ABSTRACT

Canine induced pluripotent stem cells (ciPSCs) provide a platform for regenerative veterinary medicine, disease modeling, and drug discovery. However, in the conventional method, ciPSCs are maintained using chemically-undefined media containing unknown animal components under on-murine embryonic fibroblast feeder conditions, which were reported to modify cell surface of iPSCs and increases the risk of immune rejection when the cells are transplanted into patients. Moreover, in the conventional method, ciPSCs are mechanically passaged, which requires much time and effort. Therefore, the large-scale expansion of ciPSCs is difficult, which should be resolved for using ciPSCs in clinical application and research. Here, it was shown that StemFit® AK02N and iMatrix-511 could maintain the pluripotency of ciPSCs using conventional culture method. Furthermore, it was demonstrated that the feeder-free and chemically-defined ciPSC culture systems using StemFit® AK02N and iMatrix-511 could stably maintain and allow the easy expansion of ciPSCs generated using N2B27 and StemFit® AK02N, without causing karyotype abnormalities. ciPSCs expressed several pluripotency markers and formed teratomas, including cells derived from three germ layers.


Subject(s)
Cell Culture Techniques , Culture Media/pharmacology , Dogs/anatomy & histology , Induced Pluripotent Stem Cells/cytology , Primary Cell Culture/methods , Animals , Biomarkers , Cell Adhesion , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Clone Cells , Coculture Techniques , Culture Media/analysis , Germ Layers/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/transplantation , Karyotyping , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Recombinant Proteins/pharmacology , Teratoma/etiology , Teratoma/pathology
8.
Mol Ther ; 28(11): 2394-2405, 2020 11 04.
Article in English | MEDLINE | ID: mdl-32710827

ABSTRACT

Immunotherapy utilizing induced pluripotent stem cell (iPSC) technology has great potential. Functionally rejuvenated cytotoxic T lymphocytes (CTLs) can survive long-term as young memory T cells in vivo, with continuous tumor eradication. Banking of iPSCs as an unlimited "off-the-shelf" source of therapeutic T cells may be feasible. To generate safer iPSCs, we reprogrammed human papilloma virus type 16 (HPV16) E6-specific CTLs by Sendai virus vector without cotransduction of SV40 large T antigen. The iPSCs efficiently differentiated into HPV16-specific rejuvenated CTLs that demonstrated robust cytotoxicity against cervical cancer. The tumor-suppressive effect of rejuvenated CTLs was stronger and more persistent than that of original peripheral blood CTLs. These rejuvenated HPV16-specific CTLs provide a sustained tumor-suppressive effect even for epithelial cancers and constitute promising immunotherapy for cervical cancer.


Subject(s)
Cytotoxicity, Immunologic , Immunomodulation , Induced Pluripotent Stem Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Uterine Cervical Neoplasms/immunology , Cell Differentiation/immunology , Female , Humans , Immunotherapy , Induced Pluripotent Stem Cells/cytology , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/complications , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Repressor Proteins/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocytes/cytology , T-Lymphocytes, Cytotoxic/immunology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/therapy , Uterine Cervical Neoplasms/virology
9.
Mol Reprod Dev ; 87(6): 663-665, 2020 06.
Article in English | MEDLINE | ID: mdl-32424848

ABSTRACT

Using auto-erasable Sendai virus vector, we generated ciPSC line. After several passages, virus was not present in ciPSCs by RT-PCR. ciPSCs from canine PBMCs had pluripotent state, differentiated all three germ layers in vitro, and had normal 78 XX karyotype. These results proved that PBMCs were one of the good cell sources to generate ciPSC lines from companion and patient dogs.


Subject(s)
Dogs , Induced Pluripotent Stem Cells/physiology , Leukocytes, Mononuclear/physiology , Primary Cell Culture , Sendai virus/physiology , Animals , Cell Differentiation/genetics , Cell Line, Transformed , Cell Transformation, Viral/genetics , Cellular Reprogramming/genetics , Female , Genetic Vectors/genetics , Induced Pluripotent Stem Cells/cytology , Karyotype , Leukocytes, Mononuclear/cytology , Primary Cell Culture/methods , Primary Cell Culture/veterinary , Sendai virus/genetics
10.
Int J Mol Sci ; 21(15)2020 Jul 27.
Article in English | MEDLINE | ID: mdl-32727073

ABSTRACT

Frontotemporal dementia (FTD) is caused by the progressive degeneration of the frontal and temporal lobes of the brain. Behavioral variant FTD (bvFTD) is the most common clinical subtype of FTD and pathological subtypes of bvFTD are known as FTD-tau, transactive response (TAR) DNA-binding protein 43 (TDP-43), and fused in sarcoma (FUS). Pathological mechanisms of bvFTD are largely unknown. In this study, we investigated the expression of pathological markers, such as p-Tau, TDP-43, and FUS, in the induced pluripotent stem-cell-derived neurons (iPSN) from two sporadic bvFTD patients and one normal subject. We also used an FTD-patient-derived iPSC-line-carrying microtubule-associated protein tau (MAPT) P301L point mutation as positive control for p-Tau expression. Staurosporine (STS) was used to induce cellular stress in order to investigate dynamic cellular responses related to the cell death pathway. As a result, the expression of active caspase-3 was highly increased in the bvFTD-iPSNs compared with control iPSNs in the STS-treated conditions. Other cell-death-related proteins, including Bcl-2-associated X protein (Bax)/Bcl-2 and cytochrome C, were also increased in the bvFTD-iPSNs. Moreover, we observed abnormal expression patterns of TDP-43 and FUS in the bvFTD-iPSNs compared with control iPSNs. We suggest that the iPSC technology might serve as a potential tool to demonstrate neurodegenerative phenotypes of bvFTD, which will be useful for studying pathological mechanisms for FTD as well as related drug screening in the future.


Subject(s)
Frontotemporal Dementia , Induced Pluripotent Stem Cells , Models, Neurological , Caspase 3/genetics , Caspase 3/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , tau Proteins/genetics , tau Proteins/metabolism
11.
Gene Ther ; 27(10-11): 525-534, 2019 11.
Article in English | MEDLINE | ID: mdl-32704085

ABSTRACT

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have promising potential for opening new avenues in regenerative medicine. However, since the tumorigenic potential of undifferentiated pluripotent stem cells (PSCs) is a major safety concern for clinical transplantation, inducible Caspase-9 (iC9) is under consideration for use as a fail-safe system. Here, we used targeted gene editing to introduce the iC9 system into human iPSCs, and then interrogated the efficiency of inducible apoptosis with normal iPSCs as well as diseased iPSCs derived from patients with acute myeloid leukemia (AML-iPSCs). The iC9 system induced quick and efficient apoptosis to iPSCs in vitro. More importantly, complete eradication of malignant cells without AML recurrence was shown in disease mouse models by using AML-iPSCs. In parallel, it shed light on several limitations of the iC9 system usage. Our results suggest that careful use of the iC9 system will serve as an important countermeasure against posttransplantation adverse events in stem cell transplantation therapies.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Apoptosis , Caspase 9/genetics , Caspase 9/metabolism , Cell Differentiation , Cell Line , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Pluripotent Stem Cells/metabolism
12.
Stem Cells ; 36(10): 1552-1566, 2018 10.
Article in English | MEDLINE | ID: mdl-30004605

ABSTRACT

Although autologous induced pluripotent stem cells (iPSCs) can potentially be useful for treating patients without immune rejection, in reality it will be extremely expensive and labor-intensive to make iPSCs to realize personalized medicine. An alternative approach is to make use of human leukocyte antigen (HLA) haplotype homozygous donors to provide HLA matched iPSC products to significant numbers of patients. To establish a haplobank of iPSCs, we repurposed the cord blood bank by screening ∼4,200 high resolution HLA typed cord blood samples, and selected those homozygous for the 10 most frequent HLA-A,-B,-DRB1 haplotypes in the Korean population. Following the generation of 10 iPSC lines, we conducted a comprehensive characterization, including morphology, expression of pluripotent markers and cell surface antigens, three-germ layer formation, vector clearance, mycoplasma/microbiological/viral contamination, endotoxin, and short tandem repeat (STR) assays. Various genomic analyses using microarray and comparative genomic hybridization (aCGH)-based single nucleotide polymorphism (SNP) and copy number variation (CNV) were also conducted. These 10 HLA-homozygous iPSC lines match 41.07% of the Korean population. Comparative analysis of HLA population data shows that they are also of use in other Asian populations, such as Japan, with some limited utility in ethnically diverse populations, such as the UK. Taken together, the generation of the 10 most frequent Korean HLA-homozygous iPSC lines serves as a useful pointer for the development of optimal methods for iPSC generation and quality control and indicates the benefits and limitations of collaborative HLA driven selection of donors for future stocking of worldwide iPSC haplobanks. Stem Cells 2018;36:1552-1566.


Subject(s)
Blood Banking/methods , Genomic Instability/genetics , HLA Antigens/metabolism , Induced Pluripotent Stem Cells/metabolism , Haplotypes , Histocompatibility Antigens Class II , Humans
13.
Hum Mol Genet ; 25(5): 989-1000, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26740550

ABSTRACT

Induced pluripotent stem cell (iPSC)-derived cortical neurons potentially present a powerful new model to understand corticogenesis and neurological disease. Previous work has established that differentiation protocols can produce cortical neurons, but little has been done to characterize these at cellular resolution. In particular, it is unclear to what extent in vitro two-dimensional, relatively disordered culture conditions recapitulate the development of in vivo cortical layer identity. Single-cell multiplex reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) was used to interrogate the expression of genes previously implicated in cortical layer or phenotypic identity in individual cells. Totally, 93.6% of single cells derived from iPSCs expressed genes indicative of neuronal identity. High proportions of single neurons derived from iPSCs expressed glutamatergic receptors and synaptic genes. And, 68.4% of iPSC-derived neurons expressing at least one layer marker could be assigned to a laminar identity using canonical cortical layer marker genes. We compared single-cell RNA-seq of our iPSC-derived neurons to available single-cell RNA-seq data from human fetal and adult brain and found that iPSC-derived cortical neurons closely resembled primary fetal brain cells. Unexpectedly, a subpopulation of iPSC-derived neurons co-expressed canonical fetal deep and upper cortical layer markers. However, this appeared to be concordant with data from primary cells. Our results therefore provide reassurance that iPSC-derived cortical neurons are highly similar to primary cortical neurons at the level of single cells but suggest that current layer markers, although effective, may not be able to disambiguate cortical layer identity in all cells.


Subject(s)
Cerebral Cortex/metabolism , Induced Pluripotent Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neurons/metabolism , Transcriptome , Adult , Aged , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cerebral Cortex/cytology , Female , Fetus , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/cytology , Nerve Tissue Proteins/genetics , Neurons/cytology , Real-Time Polymerase Chain Reaction , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Sequence Analysis, RNA , Single-Cell Analysis
14.
Development ; 142(18): 3222-30, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26023098

ABSTRACT

Functional assay limitations are an emerging issue in characterizing human pluripotent stem cells (PSCs). With rodent PSCs, chimera formation using pre-implantation embryos is the gold-standard assay of pluripotency (competence of progeny to differentiate into all three germ layers). In human PSCs (hPSCs), however, this can only be monitored via teratoma formation or in vitro differentiation, as ethical concerns preclude generation of human-human or human-animal chimeras. To circumvent this issue, we developed a functional assay utilizing interspecific blastocyst injection and in vitro culture (interspecies in vitro chimera assay) that enables the development and observation of embryos up to headfold stage. The assay uses mouse pre-implantation embryos and rat, monkey and human PSCs to create interspecies chimeras cultured in vitro to the early egg-cylinder stage. Intra- and interspecific chimera assays with rodent PSC lines were performed to confirm the consistency of results in vitro and in vivo. The behavior of chimeras developed in vitro appeared to recapitulate that of chimeras developed in vivo; that is, PSC-derived cells survived and were integrated into the epiblast of egg-cylinder-stage embryos. This indicates that the interspecific in vitro chimera assay is useful in evaluating the chimera-forming ability of rodent PSCs. However, when human induced PSCs (both conventional and naïve-like types) were injected into mouse embryos and cultured, some human cells survived but were segregated; unlike epiblast-stage rodent PSCs, they never integrated into the epiblast of egg-cylinder-stage embryos. These data suggest that the mouse-human interspecies in vitro chimera assay does not accurately reflect the early developmental potential/process of hPSCs. The use of evolutionarily more closely related species as host embryos might be necessary to evaluate the developmental potency of hPSCs.


Subject(s)
Cell Culture Techniques/methods , Chimera/embryology , Embryonic Development/physiology , In Vitro Techniques/methods , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Animals , Cell Line , Haplorhini , Humans , Mice , Microarray Analysis , Microinjections , Rats , Species Specificity
15.
IUBMB Life ; 70(2): 129-142, 2018 02.
Article in English | MEDLINE | ID: mdl-29316264

ABSTRACT

Cellular stress can influence efficiency of iPSCs generation and their differentiation. However, the role of intracellular cytoprotective factors in these processes is still not well known. Therefore, we investigated the effect of HO-1 (Hmox1) or Nrf2 (Nfe2l2), two major cytoprotective genes. Hmox1-/- fibroblasts demonstrated decreased reprogramming efficiency in comparison to Hmox1+/+ cells. Reversely, pharmacological enhancement of HO-1 resulted in higher number of iPSCs colonies. Importantly, elevated level of both p53 and p53-regulated miR-34a and 14-3-3σ was observed in HO-1-deficient fibroblasts whereas downregulation of p53 in these cells markedly increased their reprogramming efficiency. In human fibroblasts HO-1 silencing also induced p53 expression and affected reprogramming outcome. Hmox1+/+ and Hmox1-/- iPSCs similarly differentiated in vitro to cells originating from three germ layers, however, lower number of contracting cells was observed during this process in HO-1-deficient cells indicating attenuated cardiac differentiation. Importantly, silencing of Hmox1 in murine ESC using CRISPR/Cas-9 editing also impaired their spontaneous cardiac differentiation. Decreased reprogramming efficiency was also observed in Nrf2-lacking fibroblasts. Reversely, sulforaphane, a Nrf2 activator, increased the number of iPSCs colonies. However, both Nfe2l2+/+ and Nfe2l2-/- iPSCs showed similar pluripotency and differentiation capacity. These results indicate that regulation of HO-1 expression can further optimize generation and cardiac differentiation of iPSCs. © 2018 IUBMB Life, 70(2):129-142, 2018.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming Techniques/methods , Heme Oxygenase-1/metabolism , Induced Pluripotent Stem Cells/cytology , Membrane Proteins/metabolism , Animals , Cell Cycle/physiology , Fibroblasts , Heme Oxygenase-1/genetics , Humans , Induced Pluripotent Stem Cells/physiology , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Stem Cells ; 34(3): 581-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26850912

ABSTRACT

Although B cells have been shown to be refractory to reprogramming into pluripotency, induced pluripotent stem cells (iPSCs) have been very recently generated, at very low efficiency, from human cord blood (CB)- and peripheral blood (PB)-derived CD19+CD20 + B cells using nonintegrative tetracistronic OSKM-expressing Sendai Virus (SeV). Here, we addressed whether cell ontogeny and hierarchy influence the reprogramming efficiency of the B-cell compartment. We demonstrate that human fetal liver (FL)-derived CD19 + B cells are 110-fold easier to reprogram into iPSCs than those from CB/PB. Similarly, FL-derived CD34+CD19 + B progenitors are reprogrammed much easier than mature B cells (0.46% vs. 0.11%). All FL B-cell iPSCs carry complete VDJH rearrangements while 55% and 45% of the FL B-progenitor iPSCs carry incomplete and complete VDJH rearrangements, respectively, reflecting the reprogramming of developmentally different B progenitors (pro-B vs. pre-B) within a continuous differentiation process. Finally, our data suggest that successful B-cell reprogramming relies on active cell proliferation, and it is MYC-dependent as identical nonintegrative polycistronic SeV lacking MYC (OSKL or OSKLN) fail to reprogram B cells. The ability to efficiently reprogram human fetal primary B cells and B precursors offers an unprecedented opportunity for studying developmental B-lymphopoiesis and modeling B-cell malignances.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells , Animals , Cell Culture Techniques , Cell Proliferation/genetics , Fetal Blood/cytology , Genetic Vectors , Histone-Lysine N-Methyltransferase/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics
17.
Stem Cells ; 34(8): 2063-78, 2016 08.
Article in English | MEDLINE | ID: mdl-27097283

ABSTRACT

An expanded hexanucleotide repeat in a noncoding region of the C9orf72 gene is a major cause of amyotrophic lateral sclerosis (ALS), accounting for up to 40% of familial cases and 7% of sporadic ALS in European populations. We have generated induced pluripotent stem cells (iPSCs) from fibroblasts of patients carrying C9orf72 hexanucleotide expansions, differentiated these to functional motor and cortical neurons, and performed an extensive phenotypic characterization. In C9orf72 iPSC-derived motor neurons, decreased cell survival is correlated with dysfunction in Ca(2+) homeostasis, reduced levels of the antiapoptotic protein Bcl-2, increased endoplasmic reticulum (ER) stress, and reduced mitochondrial membrane potential. Furthermore, C9orf72 motor neurons, and also cortical neurons, show evidence of abnormal protein aggregation and stress granule formation. This study is an extensive characterization of iPSC-derived motor neurons as cellular models of ALS carrying C9orf72 hexanucleotide repeats, which describes a novel pathogenic link between C9orf72 mutations, dysregulation of calcium signaling, and altered proteostasis and provides a potential pharmacological target for the treatment of ALS and the related neurodegenerative disease frontotemporal dementia. Stem Cells 2016;34:2063-2078.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/genetics , Calcium/metabolism , DNA Repeat Expansion/genetics , Endoplasmic Reticulum/metabolism , Frontotemporal Dementia/pathology , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/metabolism , Amyotrophic Lateral Sclerosis/genetics , Apoptosis , Caspase 3/metabolism , Cell Differentiation , Cellular Reprogramming , Cerebral Cortex/pathology , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Endoplasmic Reticulum/ultrastructure , Fibroblasts/metabolism , Fibroblasts/pathology , Frontotemporal Dementia/genetics , Homeostasis/genetics , Humans , Mitochondria/metabolism , Mitochondria/ultrastructure , Peptides/metabolism , Protein Aggregates , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA/genetics
18.
Proc Natl Acad Sci U S A ; 111(47): 16772-7, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385620

ABSTRACT

Interindividual differences in hepatic metabolism, which are mainly due to genetic polymorphism in its gene, have a large influence on individual drug efficacy and adverse reaction. Hepatocyte-like cells (HLCs) differentiated from human induced pluripotent stem (iPS) cells have the potential to predict interindividual differences in drug metabolism capacity and drug response. However, it remains uncertain whether human iPSC-derived HLCs can reproduce the interindividual difference in hepatic metabolism and drug response. We found that cytochrome P450 (CYP) metabolism capacity and drug responsiveness of the primary human hepatocytes (PHH)-iPS-HLCs were highly correlated with those of PHHs, suggesting that the PHH-iPS-HLCs retained donor-specific CYP metabolism capacity and drug responsiveness. We also demonstrated that the interindividual differences, which are due to the diversity of individual SNPs in the CYP gene, could also be reproduced in PHH-iPS-HLCs. We succeeded in establishing, to our knowledge, the first PHH-iPS-HLC panel that reflects the interindividual differences of hepatic drug-metabolizing capacity and drug responsiveness.


Subject(s)
Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Liver Function Tests , Liver/drug effects , Cell Differentiation , Cytochrome P-450 Enzyme System/metabolism , Flow Cytometry , Hepatocytes/enzymology , Humans , Liver/enzymology
19.
RNA ; 19(6): 803-10, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23611983

ABSTRACT

piRNA (PIWI-interacting RNA) is a germ cell-specific small RNA in which biogenesis PIWI (P-element wimpy testis) family proteins play crucial roles. MILI (mouse Piwi-like), one of the three mouse PIWI family members, is indispensable for piRNA production, DNA methylation of retrotransposons presumably through the piRNA, and spermatogenesis. The biogenesis of piRNA has been divided into primary and secondary processing pathways; in both of these MILI is involved in mice. To analyze the molecular function of MILI in piRNA biogenesis, we utilized germline stem (GS) cells, which are derived from testicular stem cells and possess a spermatogonial phenotype. We established MILI-null GS cell lines and their revertant, MILI-rescued GS cells, by introducing the Mili gene with Sendai virus vector. Comparison of wild-type, MILI-null, and MILI-rescued GS cells revealed that GS cells were quite useful for analyzing the molecular mechanisms of piRNA production, especially the primary processing pathway. We found that glycerol-3-phosphate acyltransferase 2 (GPAT2), a mitochondrial outer membrane protein for lysophosphatidic acid, bound to MILI using the cells and that gene knockdown of GPAT2 brought about impaired piRNA production in GS cells. GPAT2 is not only one of the MILI bound proteins but also a protein essential for primary piRNA biogenesis.


Subject(s)
Glycerol-3-Phosphate O-Acyltransferase/metabolism , RNA, Small Interfering/metabolism , Stem Cells/metabolism , Testis/metabolism , Animals , Animals, Newborn , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Blotting, Western , Cell Cycle Proteins , Cells, Cultured , Gene Knockdown Techniques , Genetic Vectors/metabolism , Glycerol-3-Phosphate O-Acyltransferase/genetics , Immunoprecipitation , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred DBA , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Protein Binding , RNA, Small Interfering/genetics , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Sendai virus/genetics , Sendai virus/metabolism , Stem Cells/cytology , Testis/cytology
20.
Stem Cells ; 32(12): 3099-111, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25187421

ABSTRACT

Pluripotent stem cells have been shown to have unique nuclear properties, for example, hyperdynamic chromatin and large, condensed nucleoli. However, the contribution of the latter unique nucleolar character to pluripotency has not been well understood. Here, we show that fibrillarin (FBL), a critical methyltransferase for ribosomal RNA (rRNA) processing in nucleoli, is one of the proteins highly expressed in pluripotent embryonic stem (ES) cells. Stable expression of FBL in ES cells prolonged the pluripotent state of mouse ES cells cultured in the absence of leukemia inhibitory factor (LIF). Analyses using deletion mutants and a point mutant revealed that the methyltransferase activity of FBL regulates stem cell pluripotency. Knockdown of this gene led to significant delays in rRNA processing, growth inhibition, and apoptosis in mouse ES cells. Interestingly, both partial knockdown of FBL and treatment with actinomycin D, an inhibitor of rRNA synthesis, induced the expression of differentiation markers in the presence of LIF and promoted stem cell differentiation into neuronal lineages. Moreover, we identified p53 signaling as the regulatory pathway for pluripotency and differentiation of ES cells. These results suggest that proper activity of rRNA production in nucleoli is a novel factor for the regulation of pluripotency and differentiation ability of ES cells.


Subject(s)
Apoptosis/physiology , Cell Differentiation/physiology , Cell Nucleolus/metabolism , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , RNA, Ribosomal/biosynthesis , Animals , Cell Differentiation/genetics , Cells, Cultured , Leukemia Inhibitory Factor/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL