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










Publication year range
2.
Regen Ther ; 21: 389-397, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36196449

ABSTRACT

Introduction: Human induced pluripotent stem cells (hiPSCs) are generated through the reprogramming of somatic cells expressing a defined set of transcription factors. The advent of autologous iPSCs has enabled the generation of patient-specific iPSC lines and is expected to contribute to the exploration of cures and causes of diseases, drug screening, and tailor-made regenerative medicines. Efficient control of hiPSC derivation is beneficial for industrial applications. However, the mechanisms underlying somatic cell reprogramming remain unknown, while reprogramming efficiency remains extremely low, especially in human cells. Methods and results: We previously reported that chemical inhibition of the NOTCH signaling pathway and DOT1L promoted the generation of hiPSCs from keratinocytes, but the mechanisms and effect of this double inhibition on other types of cells remain to be investigated. Here, we found that the NOTCH/DOT1L inhibition markedly increased iPSC colony generation from human fibroblast cells via mRNA reprogramming, and mesenchymal to epithelial transition (MET)-related genes are significantly expressed in the early phase of the reprogramming. We successfully derived hiPSC lines using a single-cell sorting system under efficient reprogramming conditions. Conclusions: This user-friendly reprogramming approach paves the way for the development of hiPSC derivations in industrial applications of disease modeling and drug screening.

5.
Stem Cells Int ; 2017: 7541734, 2017.
Article in English | MEDLINE | ID: mdl-28706537

ABSTRACT

A large number of myocytes are necessary to treat intractable muscular disorders such as Duchenne muscular dystrophy with cell-based therapies. However, starting materials for cellular therapy products such as myoblasts, marrow stromal cells, menstrual blood-derived cells, and placenta-derived cells have a limited lifespan and cease to proliferate in vitro. From the viewpoints of manufacturing and quality control, cells with a long lifespan are more suitable as a starting material. In this study, we generated stromal cells for future myoblast therapy from a working cell bank of human embryonic stem cells (ESCs). The ESC-derived CD105+ cells with extensive in vitro proliferation capability exhibited myogenesis and genetic stability in vitro. These results imply that ESC-derived CD105+ cells are another cell source for myoblasts in cell-based therapy for patients with genetic muscular disorders. Since ESCs are immortal, mesenchymal stromal cells generated from ESCs can be manufactured at a large scale in one lot for pharmaceutical purposes.

6.
JCI Insight ; 2(1): e86492, 2017 01 12.
Article in English | MEDLINE | ID: mdl-28097227

ABSTRACT

Functional intestines are composed of cell types from all 3 primary germ layers and are generated through a highly orchestrated and serial developmental process. Directed differentiation of human pluripotent stem cells (hPSCs) has been shown to yield gut-specific cell types; however, these structures do not reproduce critical functional interactions between cell types of different germ layers. Here, we developed a simple protocol for the generation of mature functional intestinal organoids from hPSCs under xenogeneic-free conditions. The stem cell-derived gut organoids produced here were found to contain distinct types of intestinal cells, including enterocytes, goblet cells, Paneth cells, and enteroendocrine cells, that were derived from all 3 germ layers; moreover, they demonstrated intestinal functions, including peptide absorption, and showed innervated bowel movements in response to stimulation with histamine and anticholinergic drugs. Importantly, the gut organoids obtained using this xenogeneic-free system could be stably maintained in culture for prolonged periods and were successfully engrafted in vivo. Our xenogeneic-free approach for generating gut organoids from hPSCs provides a platform for studying human intestinal diseases and for pharmacological testing.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Intestines/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Cell Differentiation/genetics , Enterocytes/cytology , Enterocytes/metabolism , Goblet Cells/cytology , Goblet Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Octamer Transcription Factor-3 , Organoids/metabolism , Paneth Cells/cytology , Paneth Cells/metabolism , SOXB1 Transcription Factors
7.
Regen Ther ; 5: 31-39, 2016 Dec.
Article in English | MEDLINE | ID: mdl-31245498

ABSTRACT

Transformation of human embryonic stem cells (hESC) is of interest to scientists who use them as a raw material for cell-processed therapeutic products. However, the WHO and ICH guidelines provide only study design advice and general principles for tumorigenicity tests. In this study, we performed in vivo tumorigenicity tests (teratoma formation) and genome-wide sequencing analysis of undifferentiated hESCs i.e. SEES-1, -2 and -3 cells. We followed up with teratoma formation histopathologically after subcutaneous injection of SEES cells into immunodeficient mice in a qualitative manner and investigated the transforming potential of the teratomas. Maturity of SEES-teratomas perceptibly increased after long-term implantation, while areas of each tissue component remained unchanged. We found neither atypical cells/structures nor cancer in the teratomas even after long-term implantation. The teratomas generated by SEES cells matured histologically over time and did not increase in size. We also analyzed genomic structures and sequences of SEES cells during cultivation by SNP bead arrays and next-generation sequencing, respectively. The nucleotide substitution rate was 3.1 × 10-9, 4.0 × 10-9, and 4.6 × 10-9 per each division in SEES-1, SEES-2, and SEES-3 cells, respectively. Heterozygous single-nucleotide variations were detected, but no significant homologous mutations were found. Taken together, these results imply that SEES-1, -2, and -3 cells do not exhibit in vivo transformation and in vitro genomic instability.

8.
Regen Ther ; 1: 18-29, 2015 Jun.
Article in English | MEDLINE | ID: mdl-31245438

ABSTRACT

The potential applications of human embryonic stem cells (hESCs) in regenerative medicine and developmental research have made stem cell biology one of the most fascinating and rapidly expanding fields of biomedicine. The first clinical trial of hESCs in humans has begun, and the field of stem cell therapy has just entered a new era. Here, we report seven hESC lines (SEES-1, -2, -3, -4, -5, -6, and -7). Four of them were derived and maintained on irradiated human mesenchymal stem cells (hMSCs) grown in xenogeneic-free defined media and substrate. Xenogeneic-free hMSCs isolated from the subcutaneous tissue of extra fingers from individuals with polydactyly showed appropriate potentials as feeder layers in the pluripotency and growth of hESCs. In this report, we describe a comprehensive characterization of these newly derived SEES cell lines. In addition, we developed a scalable culture system for hESCs having high biological safety by using gamma-irradiated serum replacement and pharmaceutical-grade recombinant basic fibroblast growth factor (bFGF, also known as trafermin). This is first report describing the maintenance of hESC pluripotency using pharmaceutical-grade human recombinant bFGF (trafermin) and gamma-irradiated serum replacement. Our defined medium system provides a path to scalability in Good Manufacturing Practice (GMP) settings for the generation of clinically relevant cell types from pluripotent cells for therapeutic applications.

9.
Biotechnol Bioeng ; 106(6): 860-70, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20589667

ABSTRACT

We generated transgenic silkworms that synthesized human type I collagen alpha1 chain [alpha1(I) chain] in the middle silk glands and secreted it into cocoons. The initial content of the recombinant alpha1(I) chain in the cocoons of the transgenic silkworms was 0.8%. The IE1 gene, a trans-activator from the baculovirus, was introduced into the transgenic silkworm to increase the content of the chain. We also generated silkworms homozygous for the transgenes. These manipulations increased the alpha1(I) chain content to 8.0% (4.24 mg per cocoon). The alpha1(I) chain was extracted and purified from the cocoons using a very simple method. The alpha1(I) chain contained no hydroxyprolines due to the absence of prolyl-hydroxylase activity in the silk glands. Circular dichroism analysis showed that the secondary structure of the alpha1(I) chain is similar to that of denatured type I collagen, demonstrating the absence of the triple helical structure. Human skin fibroblasts were seeded on the alpha1(I) chain-coated dishes. The cells attached and spread, although at decreased chain concentrations the spreading rate was lower than that of the collagen and gelatin. Cynomolgus monkey embryonic stem cells cultured on the alpha1(I) chain-coated dishes maintained an undifferentiated state after 30 passages, and their pluripotency was confirmed by teratoma formation in severe combined immunodeficient mice. These results show that the recombinant human alpha1(I) chain is a promising candidate biomaterial as a high-quality and safe gelatin substitute for cell culture.


Subject(s)
Collagen Type I/genetics , Collagen Type I/metabolism , Culture Media/chemistry , Animals , Animals, Genetically Modified , Baculoviridae/genetics , Bombyx/chemistry , Bombyx/genetics , Bombyx/metabolism , Cell Culture Techniques/methods , Circular Dichroism , Collagen Type I/chemistry , Collagen Type I/isolation & purification , Fibroblasts , Humans , Macaca fascicularis , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Stem Cells , Trans-Activators/genetics , Viral Proteins/genetics
10.
Differentiation ; 78(2-3): 137-42, 2009.
Article in English | MEDLINE | ID: mdl-19716222

ABSTRACT

We have previously demonstrated that the tetraspanin CD9 is necessary for membrane fusion between sperm and oocyte during fertilization. While knockout mice for CD9 are viable, CD9(-/-) females are sterile due to the inability of their oocytes to fuse with sperm. While CD9 is not essential for subsequent development, a role in embryonic stem (ES) cell self-renewal was hypothesised on the basis of two observations: CD9 is highly expressed in murine and human ES cells and the CD9-blocking antibody inhibits mouse ES cell colony formation and survival. To investigate whether CD9 has a direct effect on ES cells, we generated and characterised several CD9 knockout murine ES cell lines. These CD9(-/-) ES cell lines exhibited equivalent morphology and growth properties to wild-type ES cells. Furthermore, the CD9(-/-) ES cell lines also displayed similar expression of pluripotency factors Oct3/4, Sox2 and Nanog. CD9(-/-) ES cells were found to be pluripotent in vivo, as their cells injected into immunocompromised mice gave rise to teratomas consisting of tissues representative of all three germ layers. Additionally several high contribution mouse chimeras were generated by blastocyst injection with several CD9(-/-) ES cell lines. Taken together, our results reveal that CD9 is dispensable for mouse ES cell self-renewal and pluripotency. The generation of CD9(-/-) ES cells should prove to be a useful tool with which to study the function of this protein and a range of other associated cellular processes.


Subject(s)
Antigens, CD/physiology , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Germ Layers/cytology , Membrane Glycoproteins/physiology , Regeneration , Alkaline Phosphatase/metabolism , Animals , Cell Survival , Female , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Membrane Glycoproteins/antagonists & inhibitors , Mice , Mice, Inbred ICR , Mice, Knockout , Mice, Nude , Oligonucleotide Array Sequence Analysis , Teratoma/metabolism , Teratoma/pathology , Tetraspanin 29
11.
Exp Cell Res ; 315(16): 2727-40, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19559696

ABSTRACT

POU5F1 (more commonly known as OCT4/3) is one of the stem cell markers, and affects direction of differentiation in embryonic stem cells. To investigate whether cells of mesenchymal origin acquire embryonic phenotypes, we generated human cells of mesodermal origin with overexpression of the chimeric OCT4/3 gene with physiological co-activator EWS (product of the EWSR1 gene), which is driven by the potent EWS promoter by translocation. The cells expressed embryonic stem cell genes such as NANOG, lost mesenchymal phenotypes, and exhibited embryonal stem cell-like alveolar structures when implanted into the subcutaneous tissue of immunodeficient mice. Hierarchical analysis by microchip analysis and cell surface analysis revealed that the cells are subcategorized into the group of human embryonic stem cells and embryonal carcinoma cells. These results imply that cells of mesenchymal origin can be traced back to cells of embryonic phenotype by the OCT4/3 gene in collaboration with the potent cis-regulatory element and the fused co-activator. The cells generated in this study with overexpression of chimeric OCT4/3 provide us with insight into cell plasticity involving OCT4/3 that is essential for embryonic cell maintenance, and the complexity required for changing cellular identity.


Subject(s)
Embryo, Mammalian , Mesoderm/physiology , Morphogenesis/physiology , Octamer Transcription Factor-3 , RNA-Binding Protein EWS/metabolism , Recombinant Fusion Proteins , Animals , Biomarkers/metabolism , Cell Lineage , Cells, Cultured , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Karyotyping , Mice , Mice, Inbred NOD , Mice, SCID , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Principal Component Analysis , RNA-Binding Protein EWS/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Teratoma/metabolism , Teratoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...