Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Stem Cell Res Ther ; 15(1): 106, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627844

ABSTRACT

BACKGROUND: Although oncogenic RAS mutants are thought to exert mutagenic effects upon blood cells, it remains uncertain how a single oncogenic RAS impacts non-transformed multipotent hematopoietic stem or progenitor cells (HPCs). Such potential pre-malignant status may characterize HPCs in patients with RAS-associated autoimmune lymphoproliferative syndrome-like disease (RALD). This study sought to elucidate the biological and molecular alterations in human HPCs carrying monoallelic mutant KRAS (G13C) with no other oncogene mutations. METHODS: We utilized induced pluripotent stem cells (iPSCs) derived from two unrelated RALD patients. Isogenic HPC pairs harboring either wild-type KRAS or monoallelic KRAS (G13C) alone obtained following differentiation enabled reliable comparative analyses. The compound screening was conducted with an established platform using KRAS (G13C) iPSCs and differentiated HPCs. RESULTS: Cell culture assays revealed that monoallelic KRAS (G13C) impacted both myeloid differentiation and expansion characteristics of iPSC-derived HPCs. Comprehensive RNA-sequencing analysis depicted close clustering of HPC samples within the isogenic group, warranting that comparative studies should be performed within the same genetic background. When compared with no stimulation, iPSC-derived KRAS (G13C)-HPCs showed marked similarity with the wild-type isogenic control in transcriptomic profiles. After stimulation with cytokines, however, KRAS (G13C)-HPCs exhibited obvious aberrant cell-cycle and apoptosis responses, compatible with "dysregulated expansion," demonstrated by molecular and biological assessment. Increased BCL-xL expression was identified amongst other molecular changes unique to mutant HPCs. With screening platforms established for therapeutic intervention, we observed selective activity against KRAS (G13C)-HPC expansion in several candidate compounds, most notably in a MEK- and a BCL-2/BCL-xL-inhibitor. These two compounds demonstrated selective inhibitory effects on KRAS (G13C)-HPCs even with primary patient samples when combined. CONCLUSIONS: Our findings indicate that a monoallelic oncogenic KRAS can confer dysregulated expansion characteristics to non-transformed HPCs, which may constitute a pathological condition in RALD hematopoiesis. The use of iPSC-based screening platforms will lead to discovering treatments that enable selective inhibition of RAS-mutated HPC clones.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
2.
Cancer Sci ; 114(8): 3433-3445, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37302818

ABSTRACT

Classic Hodgkin lymphoma (cHL) is characterized by multinucleated cells called Reed-Sternberg (RS) cells and genetic complexity. Although CD30 also characterizes cHL cells, its biological roles are not fully understood. In this report, we examined the link between CD30 and these characteristics of cHL cells. CD30 stimulation increased multinucleated cells resembling RS cells. We found chromatin bridges, a cause of mitotic errors, among the nuclei of multinucleated cells. CD30 stimulation induced DNA double-strand breaks (DSBs) and chromosomal imbalances. RNA sequencing showed significant changes in the gene expression by CD30 stimulation. We found that CD30 stimulation increased intracellular reactive oxygen species (ROS), which induced DSBs and multinucleated cells with chromatin bridges. The PI3K pathway was responsible for CD30-mediated generation of multinucleated cells by ROS. These results suggest that CD30 involves generation of RS cell-like multinucleated cells and chromosomal instability through induction of DSBs by ROS, which subsequently induces chromatin bridges and mitotic error. The results link CD30 not only to the morphological features of cHL cells, but also to the genetic complexity, both of which are characteristic of cHL cells.


Subject(s)
Hodgkin Disease , Reed-Sternberg Cells , Humans , Reed-Sternberg Cells/metabolism , Reed-Sternberg Cells/pathology , Hodgkin Disease/metabolism , Reactive Oxygen Species/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Line , Chromosomal Instability/genetics , Chromatin/genetics , Chromatin/metabolism , Ki-1 Antigen/genetics , Ki-1 Antigen/metabolism
3.
J Autoimmun ; 139: 103085, 2023 09.
Article in English | MEDLINE | ID: mdl-37354689

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disease characterized by genetic heterogeneity and an interferon (IFN) signature. The overall landscapes of the heritability of SLE remains unclear. OBJECTIVES: To identify and elucidate the biological functions of rare variants underlying SLE, we conducted analyses of patient-derived induced pluripotent stem cells (iPSCs) in combination with genetic analysis. METHODS: Two familial SLE patient- and two healthy donor (HD)-derived iPSCs were established. Type 1 IFN-secreting dendritic cells (DCs) were differentiated from iPSCs. Genetic analyses of SLE-iPSCs, and 117 SLE patients and 107 HDs in the ImmuNexUT database were performed independently. Genome editing of the variants on iPSCs was performed with the CRISPR/Cas9 system. RESULTS: Type 1 IFN secretion was significantly increased in DCs differentiated from SLE-iPSCs compared to HD-iPSCs. Genetic analyses revealed a rare variant in the 2'-5'-Oligoadenylate Synthetase Like (OASL) shared between SLE-iPSCs and another independent SLE patient, and significant accumulation of OASL variants among SLE patients (HD 0.93%, SLE 6.84%, OR 8.387) in the database. Genome editing of mutated OASL 202Q to wild-type 202 R or wild-type OASL 202 R to mutated 202Q resulted in reduced or enhanced Type 1 IFN secretion of DCs. Three other OASL variants (R60W, T261S and A447V) accumulated in SLE patients had also capacities to enhance Type 1 IFN secretion in response to dsRNA. CONCLUSIONS: We established a patient-derived iPSC-based strategy to investigate the linkage of genotype and phenotype in autoimmune diseases. Detailed case-based investigations using patient-derived iPSCs provide information to unveil the heritability of the pathogenesis of autoimmune diseases.


Subject(s)
Induced Pluripotent Stem Cells , Lupus Erythematosus, Systemic , Humans , Interferons , Adenine Nucleotides , Lupus Erythematosus, Systemic/genetics
4.
Transfusion ; 63(3): 463-469, 2023 03.
Article in English | MEDLINE | ID: mdl-36597800

ABSTRACT

BACKGROUND: Flow cytometry (FC) has proven its utility in scrutinizing AB antigen expression in red blood cells (RBCs), cooperating with serological tests for accurate blood group typing. However, technical difficulties may impair the characterization of weak ABO subtypes when background noises appear at non-negligible levels. STUDY DESIGN AND METHODS: We sought to establish an FC method that could prevent antibody-induced hemagglutination and an increase in cellular autofluorescence, two major issues inherent to RBC-FC analysis of AB expression. We optimized fixatives, multicolor-staining protocols, and sequential gating strategies. Blood samples from weak ABO subtype cases, Bm and Ael , were analyzed with the established protocol. RESULTS: The optimized mixture of glutaraldehyde and formaldehyde successfully generated fixed RBCs resistant to agglutination while maintaining low autofluorescence. These features allowed co-staining of leukocyte- and erythrocyte-markers, which enabled sequential gating strategies facilitating the precise AB antigen analysis in purely single RBCs with minimum background noises. By the established FC analysis, we could detect in the Bm sample a small RBC population exhibiting weak B antigen expression. The assay also proved it feasible to identify a small population (0.04%) of RBCs weakly expressing the A antigen in the Ael sample confirmed as harboring a rare c.816dupG ABO variant allele. CONCLUSION: The RBC-FC analysis described here allows the detection of AB antigens weakly expressed in RBCs while achieving minimum background noise levels in negative control samples. Overall, the modified protocol provides a quick and reliable assay valuable in transfusion medicine and is potentially applicable to the characterization of rare weak ABO variants.


Subject(s)
ABO Blood-Group System , Erythrocytes , Humans , Flow Cytometry/methods , Erythrocytes/metabolism , Antibodies/metabolism , Blood Grouping and Crossmatching , Antigens/metabolism
5.
Mol Ther Methods Clin Dev ; 23: 424-433, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34786435

ABSTRACT

Two patients with adenosine deaminase (ADA)-deficient severe combined immunodeficiency (ADA-SCID) received stem cell-based gene therapy (SCGT) using GCsapM-ADA retroviral vectors without preconditioning in 2003 and 2004. The first patient (Pt1) was treated at 4.7 years old, and the second patient (Pt2), who had previously received T cell gene therapy (TCGT), was treated at 13 years old. More than 10 years after SCGT, T cells showed a higher vector copy number (VCN) than other lineages. Moreover, the VCN increased with differentiation toward memory T and B cells. The distribution of vector-marked cells reflected variable levels of ADA requirements in hematopoietic subpopulations. Although neither patient developed leukemia, clonal expansion of SCGT-derived clones was observed in both patients. The use of retroviral vectors yielded clonal dominance of vector-marked clones, irrespective of the lack of leukemic changes. Vector integration sites common to all hematopoietic lineages suggested the engraftment of gene-marked progenitors in Pt1, who showed severe osteoblast (OB) insufficiency compared to Pt2, which might cause a reduction in the stem/progenitor cells in the bone marrow (BM). The impaired BM microenvironment due to metabolic abnormalities may create space for the engraftment of vector-marked cells in ADA-SCID, despite the lack of preconditioning.

6.
Stem Cell Reports ; 13(3): 530-544, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31402337

ABSTRACT

A simple induction protocol to differentiate chondrocytes from pluripotent stem cells (PSCs) using small-molecule compounds is beneficial for cartilage regenerative medicine and mechanistic studies of chondrogenesis. Here, we demonstrate that chondrocytes are robustly induced from human PSCs by simple combination of two compounds, CHIR99021, a glycogen synthase kinase 3 inhibitor, and TTNPB, a retinoic acid receptor (RAR) agonist, under serum- and feeder-free conditions within 5-9 days. An excellent differentiation efficiency and potential to form hyaline cartilaginous tissues in vivo were demonstrated. Comprehensive gene expression and open chromatin analyses at each protocol stage revealed step-by-step differentiation toward chondrocytes. Genome-wide analysis of RAR and ß-catenin association with DNA showed that retinoic acid and Wnt/ß-catenin signaling collaboratively regulated the key marker genes at each differentiation stage. This method provides a promising cell source for regenerative medicine and, as an in vitro model, may facilitate elucidation of the molecular mechanisms underlying chondrocyte differentiation.


Subject(s)
Benzoates/pharmacology , Cell Differentiation/drug effects , Chondrocytes/metabolism , Pluripotent Stem Cells/cytology , Pyridines/pharmacology , Pyrimidines/pharmacology , Retinoids/pharmacology , Animals , Cartilage/metabolism , Cartilage/pathology , Chondrocytes/cytology , Chondrocytes/transplantation , Chondrogenesis , Chromatin/metabolism , Collagen Type X/genetics , Collagen Type X/metabolism , Gene Expression , Humans , Mice , Mice, Inbred NOD , Pluripotent Stem Cells/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism
7.
Stem Cell Res ; 39: 101485, 2019 08.
Article in English | MEDLINE | ID: mdl-31255830

ABSTRACT

We established three iPSC lines from postmortem-cultured fibroblasts derived following the sudden unexpected death of an 8-year-old girl with Lennox-Gastaut syndrome, who turned out to have the R551H-mutant STXBP1 gene. These iPSC clones showed pluripotent characteristics while retaining the genotype and demonstrated trilineage differentiation capability, indicating their utility in disease-modeling studies, i.e., STXBP1-encephalopathy. This is the first report on the establishment of iPSCs from a sudden death child, suggesting the possible use of postmortem-iPSC technologies as an epoch-making approach for precise identification of the cause of sudden death.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Munc18 Proteins/genetics , Adolescent , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Karyotype , Leukocytes, Mononuclear/metabolism , Microsatellite Repeats/genetics , Mutation/genetics
8.
Stem Cell Res Ther ; 10(1): 185, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31234949

ABSTRACT

BACKGROUND: Disease modeling with patient-derived induced pluripotent stem cells (iPSCs) is a powerful tool for elucidating the mechanisms underlying disease pathogenesis and developing safe and effective treatments. Patient peripheral blood (PB) cells are used for iPSC generation in many cases since they can be collected with minimum invasiveness. To derive iPSCs that lack immunoreceptor gene rearrangements, hematopoietic stem and progenitor cells (HSPCs) are often targeted as the reprogramming source. However, the current protocols generally require HSPC mobilization and/or ex vivo expansion owing to their sparsity at the steady state and low reprogramming efficiencies, making the overall procedure costly, laborious, and time-consuming. METHODS: We have established a highly efficient method for generating iPSCs from non-mobilized PB-derived CD34+ HSPCs. The source PB mononuclear cells were obtained from 1 healthy donor and 15 patients and were kept frozen until the scheduled iPSC generation. CD34+ HSPC enrichment was done using immunomagnetic beads, with no ex vivo expansion culture. To reprogram the CD34+-rich cells to pluripotency, the Sendai virus vector SeVdp-302L was used to transfer four transcription factors: KLF4, OCT4, SOX2, and c-MYC. In this iPSC generation series, the reprogramming efficiencies, success rates of iPSC line establishment, and progression time were recorded. After generating the iPSC frozen stocks, the cell recovery and their residual transgenes, karyotypes, T cell receptor gene rearrangement, pluripotency markers, and differentiation capability were examined. RESULTS: We succeeded in establishing 223 iPSC lines with high reprogramming efficiencies from 15 patients with 8 different disease types. Our method allowed the rapid appearance of primary colonies (~ 8 days), all of which were expandable under feeder-free conditions, enabling robust establishment steps with less workload. After thawing, the established iPSC lines were verified to be pluripotency marker-positive and of non-T cell origin. A majority of the iPSC lines were confirmed to be transgene-free, with normal karyotypes. Their trilineage differentiation capability was also verified in a defined in vitro assay. CONCLUSION: This robust and highly efficient method enables the rapid and cost-effective establishment of transgene-free iPSC lines from a small volume of PB, thus facilitating the biobanking of patient-derived iPSCs and their use for the modeling of various diseases.


Subject(s)
Antigens, CD34/metabolism , Cellular Reprogramming/physiology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Sendai virus/genetics , Adolescent , Adult , Aged , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cellular Reprogramming/genetics , Female , Flow Cytometry , Humans , Karyotyping , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Middle Aged , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Young Adult
9.
Sci Rep ; 8(1): 15855, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30367142

ABSTRACT

Chronic myelomonocytic leukemia (CMML) is an entity of myelodysplastic syndrome/myeloproliferative neoplasm. Although CMML can be cured with allogeneic stem cell transplantation, its prognosis is generally very poor due to the limited efficacy of chemotherapy and to the patient's age, which is usually not eligible for transplantation. Comprehensive analysis of CMML pathophysiology and the development of therapeutic agents have been limited partly due to the lack of cell lines in CMML and the limited developments of mouse models. After successfully establishing patient's derived disease-specific induced pluripotent stem cells (iPSCs) derived from a patient with CMML, we utilized these CMML-iPSCs to achieve hematopoietic re-differentiation in vitro, created a humanized CMML mouse model via teratomas, and developed a drug-testing system. The clinical characteristics of CMML were recapitulated following hematopoietic re-differentiation in vitro and a humanized CMML mouse model in vivo. The drug-testing system using CMML-iPSCs identified a MEK inhibitor, a Ras inhibitor, and liposomal clodronate as potential drugs for treating CMML. Clodronate is a drug commonly used as a bisphosphonate for osteoporosis. In this study, the liposomalization of clodronate enhanced its effectiveness in these assays, suggesting that this variation of clodronate may be adopted as a repositioned drug for CMML therapy.


Subject(s)
Clodronic Acid/therapeutic use , Leukemia, Myelomonocytic, Chronic/drug therapy , Liposomes/chemistry , Protein Kinase Inhibitors/therapeutic use , Aged , Animals , Cell Differentiation , Disease Models, Animal , Drug Repositioning , Enhancer of Zeste Homolog 2 Protein/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Leukemia, Myelomonocytic, Chronic/pathology , Leukemia, Myelomonocytic, Chronic/therapy , Male , Mice , Neurofibromin 1/metabolism , Phosphorylation , STAT5 Transcription Factor/metabolism , Teratoma/metabolism , Teratoma/pathology , Transplantation, Heterologous
10.
Stem Cell Reports ; 11(2): 380-394, 2018 08 14.
Article in English | MEDLINE | ID: mdl-29983389

ABSTRACT

Oncogenic KRAS mutations in hematopoietic stem cells cause RAS-associated autoimmune lymphoproliferative syndrome-like disease (RALD). KRAS plays essential roles in stemness maintenance in some types of stem cells. However, its roles in pluripotent stem cells (PSCs) are poorly understood. Here, we investigated the roles of KRAS on stemness in the context of induced PSCs (iPSCs). We used KRAS mutant (G13C/WT) and wild-type isogenic (WT/WT) iPSCs from the same RALD patients, as well as wild-type (WTed/WT) and heterozygous knockout (Δed/WT) iPSCs, both obtained by genome editing from the same G13C/WT clone. Compared with WT iPSCs, G13C/WT iPSCs displayed enforced retention of self-renewal and suppressed capacity for neuronal differentiation, while Δed/WT iPSCs showed normalized cellular characteristics similar to those of isogenic WTed/WT cells. The KRAS-ERK pathway, but not the KRAS-PI3K pathway, was shown to govern these G13C/WT-specific phenotypes, indicating the strong impact of the KRAS-ERK signaling upon self-renewal and differentiation propensity in human iPSCs.


Subject(s)
Cell Differentiation , Cell Self Renewal , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Alleles , Autoimmune Lymphoproliferative Syndrome , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , Cells, Cultured , Chromosome Aberrations , DNA Mutational Analysis , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Editing , Gene Expression Profiling , Genotype , Humans , Induced Pluripotent Stem Cells/drug effects , Karyotype , Molecular Imaging , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects
11.
ACS Synth Biol ; 7(7): 1709-1714, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29920201

ABSTRACT

The understanding of signaling events is critical for attaining long-term expansion of hematopoietic stem cells ex vivo. In this study, we aim to analyze the contribution of multiple signaling molecules in proliferation of hematopoietic stem cells. To this end, we design a bottom-up engineered receptor with multiple tyrosine motifs, which can recruit multiple signaling molecules of interest. This is followed by a top-down approach, where one of the multiple tyrosine motifs in the bottom-up engineered receptor is functionally knocked out by tyrosine-to-phenylalanine mutation. The combination of these two approaches demonstrates the importance of Shc in cooperation with STAT3 or STAT5 in the proliferation of hematopoietic stem cells. The platform developed herein may be applied for analyzing other cells and/or other cell fate regulation systems.


Subject(s)
Cell Proliferation/physiology , Hematopoietic Stem Cells/metabolism , Signal Transduction/physiology , Animals , Cell Proliferation/genetics , Hematopoietic Stem Cells/cytology , Humans , Mutation/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/genetics
12.
Stem Cell Rev Rep ; 14(1): 101-109, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28948469

ABSTRACT

Hematopoietic stem cells (HSCs) are a valuable resource in transplantation medicine. Cytokines are often used to culture HSCs aiming at better clinical outcomes through enhancement of HSC reconstitution capability. Roles for each signal molecule downstream of receptors in HSCs, however, remain puzzling due to complexity of the cytokine-signaling network. Engineered receptors that are non-responsive to endogenous cytokines represent an attractive tool for dissection of signaling events. We here tested a previously developed chimeric receptor (CR) system in primary murine HSCs, target cells that are indispensable for analysis of stem cell activity. Each CR contains tyrosine motifs that enable selective activation of signal molecules located downstream of the c-Mpl receptor upon stimulation by an artificial ligand. Signaling through a control CR with a wild-type c-Mpl cytoplasmic tail sufficed to enhance HSC proliferation and colony formation in cooperation with stem cell factor (SCF). Among a series of CRs, only one compatible with selective Stat5 activation showed similar positive effects. The HSCs maintained ex vivo in these environments retained long-term reconstitution ability following transplantation. This ability was also demonstrated in secondary recipients, indicating effective transmission of stem cell-supportive signals into HSCs via these artificial CRs during culture. Selective activation of Stat5 through CR ex vivo favored preservation of lymphoid potential in long-term reconstituting HSCs, but not of myeloid potential, exemplifying possible dissection of signals downstream of c-Mpl. These CR systems therefore offer a useful tool to scrutinize complex signaling pathways in HSCs.


Subject(s)
Hematopoietic Stem Cells/metabolism , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cell Line , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Stem Cell Factor/genetics , Stem Cell Factor/metabolism
13.
Stem Cell Reports ; 10(1): 314-328, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29233552

ABSTRACT

Gut epithelial organoids are routinely used to investigate intestinal biology; however, current culture methods are not amenable to genetic manipulation, and it is difficult to generate sufficient numbers for high-throughput studies. Here, we present an improved culture system of human induced pluripotent stem cell (iPSC)-derived intestinal organoids involving four methodological advances. (1) We adopted a lentiviral vector to readily establish and optimize conditioned medium for human intestinal organoid culture. (2) We obtained intestinal organoids from human iPSCs more efficiently by supplementing WNT3A and fibroblast growth factor 2 to induce differentiation into definitive endoderm. (3) Using 2D culture, followed by re-establishment of organoids, we achieved an efficient transduction of exogenous genes in organoids. (4) We investigated suspension organoid culture without scaffolds for easier harvesting and assays. These techniques enable us to develop, maintain, and expand intestinal organoids readily and quickly at low cost, facilitating high-throughput screening of pathogenic factors and candidate treatments for gastrointestinal diseases.


Subject(s)
Cell Culture Techniques/methods , Epithelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Intestinal Mucosa/metabolism , Organoids/metabolism , Animals , Epithelial Cells/cytology , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/cytology , Intestinal Mucosa/cytology , Lentivirus , Mice , Organoids/cytology , Wnt3A Protein/genetics , Wnt3A Protein/metabolism
14.
EBioMedicine ; 23: 34-45, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28789943

ABSTRACT

Visceral fat accumulation as observed in Crohn's disease and obesity is linked to chronic gut inflammation, suggesting that accumulation of gut adipocytes can trigger local inflammatory signaling. However, direct interactions between intestinal epithelial cells (IECs) and adipocytes have not been investigated, in part because IEC physiology is difficult to replicate in culture. In this study, we originally prepared intact, polarized, and cytokine responsive IEC monolayers from primary or induced pluripotent stem cell-derived intestinal organoids by simple and repeatable methods. When these physiological IECs were co-cultured with differentiated adipocytes in Transwell, pro-inflammatory genes were induced in both cell types, suggesting reciprocal inflammatory activation in the absence of immunocompetent cells. These inflammatory responses were blocked by nuclear factor-κB or signal transducer and activator of transcription 3 inhibition and by anti-tumor necrosis factor- or anti-interleukin-6-neutralizing antibodies. Our results highlight the utility of these monolayers for investigating IEC biology. Furthermore, this system recapitulates the intestinal epithelium-mesenteric fat signals that potentially trigger or worsen inflammatory disorders such as Crohn's disease and obesity-related enterocolitis.


Subject(s)
Adipocytes/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Signal Transduction , Adipocytes/cytology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Line , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Male , Mice , NF-kappa B/metabolism , STAT3 Transcription Factor
15.
Article in Japanese | MEDLINE | ID: mdl-28539554

ABSTRACT

The pluripotent stem cells have a self-renewal ability and can be differentiated into theoretically all of cell types. The induced pluripotent stem (iPS) cells overcame the ethical problems of the human embryonic stem (ES) cell, and enable pathologic analysis of intractable diseases and drug discovery. The in vitro disease model using disease-specific iPS cells enables repeated analyses of human cells without influence of environment factors. Even though autoimmune diseases are polygenic diseases, autoimmune disease-specific iPS cells are thought to be a promising tool for analyzing the pathogenesis of the diseases and drug discovery in future.


Subject(s)
Autoimmune Diseases/etiology , Induced Pluripotent Stem Cells , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Cell Differentiation , Drug Discovery/trends , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Lupus Erythematosus, Systemic
16.
Stem Cell Reports ; 8(5): 1155-1163, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28494936

ABSTRACT

Although it is important to clarify the pathogenic functions of T cells in human samples, their examination is often limited due to difficulty in obtaining sufficient numbers of dendritic cells (DCs), used as antigen-presenting cells, especially in autoimmune diseases. We describe the generation of DCs from induced pluripotent stem cells derived from T cells (T-iPSCs). We reprogrammed CD4+ T cell clones from a patient with Sjögren's syndrome (SS) into iPSCs, which were differentiated into DCs (T-iPS-DCs). T-iPS-DCs had dendritic cell-like morphology, and expressed CD11c, HLA-DR, CD80, CD86, and also BDCA-3. Compared with monocyte-derived DCs, the capacity for antigen processing was similar, and T-iPS-DCs induced the proliferative response of autoreactive CD4+ T cells. Moreover, we could evaluate T cell functions of the patient with SS. In conclusion, we obtained adequate numbers of DCs from T-iPSCs, which could be used to characterize pathogenic T cells in autoimmune diseases such as SS.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cellular Reprogramming , Dendritic Cells/cytology , Induced Pluripotent Stem Cells/cytology , Sjogren's Syndrome/pathology , Antigen Presentation , Antigens, Surface/genetics , Antigens, Surface/metabolism , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , CD11 Antigens/genetics , CD11 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cells, Cultured , Dendritic Cells/immunology , HLA-DR Antigens/genetics , HLA-DR Antigens/metabolism , Humans , Thrombomodulin
17.
Stem Cell Reports ; 8(3): 500-508, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28238792

ABSTRACT

Hematopoietic stem cells (HSCs) are considered one of the most promising therapeutic targets for the treatment of various blood disorders. However, due to difficulties in establishing stable maintenance and expansion of HSCs in vitro, their insufficient supply is a major constraint to transplantation studies. To solve these problems we have developed a fully defined, all-recombinant protein-based culture system. Through this system, we have identified hemopexin (HPX) and interleukin-1α as responsible for HSC maintenance in vitro. Subsequent molecular analysis revealed that HPX reduces intracellular reactive oxygen species levels within cultured HSCs. Furthermore, bone marrow immunostaining and 3D immunohistochemistry revealed that HPX is expressed in non-myelinating Schwann cells, known HSC niche constituents. These results highlight the utility of this fully defined all-recombinant protein-based culture system for reproducible in vitro HSC culture and its potential to contribute to the identification of factors responsible for in vitro maintenance, expansion, and differentiation of stem cell populations.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Recombinant Proteins/pharmacology , Animals , Blood Proteins/pharmacology , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Colony-Forming Units Assay , Hematopoietic Stem Cells/metabolism , Hemopexin/pharmacology , Interleukin-1alpha/pharmacology , Mice
18.
Stem Cells ; 35(4): 989-1002, 2017 04.
Article in English | MEDLINE | ID: mdl-27753160

ABSTRACT

Hematopoietic stem cell (HSC) transplantation (HSCT) for malignancy requires toxic pre-conditioning to maximize anti-tumor effects and donor-HSC engraftment. While this induces bone marrow (BM)-localized inflammation, how this BM environmental change affects transplanted HSCs in vivo remains largely unknown. We here report that, depending on interval between irradiation and HSCT, residence within lethally irradiated recipient BM compromises donor-HSC reconstitution ability. Both in vivo and in vitro we demonstrate that, among inflammatory cytokines, TNF-α plays a role in HSC damage: TNF-α stimulation leads to accumulation of reactive oxygen species (ROS) in highly purified hematopoietic stem/progenitor cells (HSCs/HSPCs). Transplantation of flow-cytometry-sorted murine HSCs reveals damaging effects of accumulated ROS on HSCs. Short-term incubation either with an specific inhibitor of tumor necrosis factor receptor 1 signaling or an antioxidant N-acetyl-L-cysteine (NAC) prevents TNF-α-mediated ROS accumulation in HSCs. Importantly, pre-transplantation exposure to NAC successfully demonstrats protective effects in inflammatory BM on graft-HSCs, exhibiting better reconstitution capability than that of nonprotected control grafts. We thus suggest that in vivo protection of graft-HSCs from BM inflammation is a feasible and attractive approach, which may lead to improved hematopoietic reconstitution kinetics in transplantation with myeloablative conditioning that inevitably causes inflammation in recipient BM. Stem Cells 2017;35:989-1002.


Subject(s)
Cytoprotection , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acetylcysteine/pharmacology , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cellular Microenvironment/drug effects , Cytoprotection/drug effects , Hematopoietic Stem Cells/drug effects , Indicators and Reagents , Inflammation/pathology , Mice, Inbred C57BL , Time Factors
19.
Hum Gene Ther Methods ; 27(5): 197-208, 2016 10.
Article in English | MEDLINE | ID: mdl-27763786

ABSTRACT

Stable gene transfer into target cell populations via integrating viral vectors is widely used in stem cell gene therapy (SCGT). Accurate vector copy number (VCN) estimation has become increasingly important. However, existing methods of estimation such as real-time quantitative PCR are more restricted in practicality, especially during clinical trials, given the limited availability of sample materials from patients. This study demonstrates the application of an emerging technology called droplet digital PCR (ddPCR) in estimating VCN states in the context of SCGT. Induced pluripotent stem cells (iPSCs) derived from a patient with X-linked chronic granulomatous disease were used as clonable target cells for transduction with alpharetroviral vectors harboring codon-optimized CYBB cDNA. Precise primer-probe design followed by multiplex analysis conferred assay specificity. Accurate estimation of per-cell VCN values was possible without reliance on a reference standard curve. Sensitivity was high and the dynamic range of detection was wide. Assay reliability was validated by observation of consistent, reproducible, and distinct VCN clustering patterns for clones of transduced iPSCs with varying numbers of transgene copies. Taken together, use of ddPCR appears to offer a practical and robust approach to VCN estimation with a wide range of clinical and research applications.


Subject(s)
DNA Copy Number Variations/genetics , Genetic Therapy , Genetic Vectors/genetics , Granulomatous Disease, Chronic/genetics , Induced Pluripotent Stem Cells/metabolism , DNA Primers/genetics , Genetic Vectors/isolation & purification , Granulomatous Disease, Chronic/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Real-Time Polymerase Chain Reaction , Stem Cell Transplantation , Virus Integration/genetics
20.
Mol Genet Metab ; 119(3): 232-238, 2016 11.
Article in English | MEDLINE | ID: mdl-27590924

ABSTRACT

Mucopolysaccharidosis type II (MPS II) is a lysosomal storage disease caused by the deficient activity of iduronate 2-sulfatase (IDS), which is involved in the lysosomal catabolism of the glycosaminoglycans (GAGs) dermatan and heparan sulfate. Such a deficiency leads to the accumulation of undegraded GAGs in some organs. Although enzyme replacement therapy is available as a treatment of MPS II, there are some limitations, such as the requirement of weekly administration for whole life. To avoid such limitations, hematopoietic cell transplantation (HSCT) is a possible alternative. In fact, some report suggested positive effects of HSCT for MPS II. However, HSCT has also some limitations. Strong conditioning regimens can cause severe side effects. For overcome this obstacle, we studied the efficacy of ACK2, an antibody that blocks KIT, followed by low-dose irradiation as a preconditioning regimen for HSCT using a murine model of MPS II. This protocol achieves 58.7±4.92% donor chimerism at 16weeks after transplantation in the peripheral blood of recipient mice. GAG levels were significantly reduced in liver, spleen, heart and intestine. These results indicated that ACK2-based preconditioning might be one of the choices for MPS II patients who receive HSCT.


Subject(s)
Antibodies, Anti-Idiotypic/administration & dosage , Mucopolysaccharidosis II/therapy , Proto-Oncogene Proteins c-kit/immunology , Animals , Bone Marrow Transplantation , Dermatan Sulfate/metabolism , Disease Models, Animal , Glycoproteins/genetics , Heparitin Sulfate/metabolism , Humans , Lysosomes/enzymology , Lysosomes/pathology , Mice , Mice, Knockout , Mucopolysaccharidosis II/immunology , Mucopolysaccharidosis II/metabolism , Mucopolysaccharidosis II/pathology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...