• Title/Summary/Keyword: Human pluripotent stem cells

Search Result 87, Processing Time 0.025 seconds

A STUDY ON THE OSTEOGENIC DIFFERENTIATION OF ADIPOSE-DERIVED ADULT STEM CELL (지방조직 유래 줄기세포의 조골세포로의 분화에 대한 실험적 연구)

  • Lee, Eui-Seok;Jang, Hyon-Seok;Kwon, Jong-Jin;Rim, Jae-Suk
    • Maxillofacial Plastic and Reconstructive Surgery
    • /
    • v.30 no.2
    • /
    • pp.133-141
    • /
    • 2008
  • Stem cells have self-renewal capacity, long-term viability, and multiline age potential. Adult bone marrow contains mesenchymal stem cells. Bone marrow-derived mesenchymal stem cells (BMSCs) are progenitors of skeletal tissue components and can differentiate into adipocytes, chondrocytes, osteoblasts, and myoblasts in vitro and undergo differentiation in vivo. However, the clinical use of BMSCs has presented problems, including pain, morbidity, and low cell number upon harvest. Recent studies have identified a putative stem cell population within the adipose tissue. Human adipose tissue contains pluripotent stem cells simillar to bone marrow-derived stem cells that can differentiate toward the osteogenic, adipogenic, myogenic, and chondrogenic lineages. Human adipose tissue-derived stem cells (ATSCs) could be proposed as an alternative source of adult bone marrow stem cells, and could be obtained in large quantities, under local anesthesia, with minimal discomfort. Human adipose tissue obtained by liposuction was processed to obtain ATSCs. In this study, we compared the osteogenic differentiation of ATSCs in a specific osteogenic induction medium with that in a non-osteogenic medium. ATSCs were incubated in an osteogenic medium for 28 days to induce osteogenesis respectively. Osteogenic differentiation was assessed by von Kossa and alkaline phosphatase staining. Expression of osteocyte specific bone sialoprotein, osteocalcin, collagen type I and alkaline phosphatase, bone morphogenic protein 2, bone morphogenic protein 6 was confirmed by RT-PCR. ATSCs incubated in the osteogenic medium were stained positively for von Kossa and alkaline phosphatase staining. Expression of osteocyte specific genes was also detected. Since this cell population can be easily identified through fluorescence microscopy, it may be an ideal source of ATSCs for further experiments on stem cell biology and tissue engineering. The present results show that ADSCs have an ability to differentiate into osteoblasts. In the present study, we extend this approach to characterize adipose tissue-derived stem cells.

Artificial gametes from stem cells

  • Moreno, Inmaculada;Miguez-Forjan, Jose Manuel;Simon, Carlos
    • Clinical and Experimental Reproductive Medicine
    • /
    • v.42 no.2
    • /
    • pp.33-44
    • /
    • 2015
  • The generation of artificial gametes is a real challenge for the scientific community today. In vitro development of human eggs and sperm will pave the way for the understanding of the complex process of human gametogenesis and will provide with human gametes for the study of infertility and the onset of some inherited disorders. However, the great promise of artificial gametes resides in their future application on reproductive treatments for all these people wishing to have genetically related children and for which gamete donation is now their unique option of parenthood. This is the case of infertile patients devoid of suitable gametes, same sex couples, singles and those fertile couples in a high risk of transmitting serious diseases to their progeny. In the search of the best method to obtain artificial gametes, many researchers have successfully obtained human germ cell-like cells from stem cells at different stages of differentiation. In the near future, this field will evolve to new methods providing not only viable but also functional and safe artificial germ cells. These artificial sperm and eggs should be able to recapitulate all the genetic and epigenetic processes needed for the correct gametogenesis, fertilization and embryogenesis leading to the birth of a healthy and fertile newborn.

Transcriptional Profiles of Imprinted Genes in Human Embryonic Stem Cells During In vitro Differentiation

  • Park, Sang-Wook;Do, Hyo-Sang;Kim, Dongkyu;Ko, Ji-Yun;Lee, Sang-Hun;Han, Yong-Mahn
    • International Journal of Stem Cells
    • /
    • v.7 no.2
    • /
    • pp.108-117
    • /
    • 2014
  • Background and Objectives: Genomic imprinting is an inheritance phenomenon by which a subset of genes are expressed from one allele of two homologous chromosomes in a parent of origin-specific manner. Even though fine-tuned regulation of genomic imprinting process is essential for normal development, no other means are available to study genomic imprinting in human during embryonic development. In relation with this bottleneck, differentiation of human embryonic stem cells (hESCs) into specialized lineages may be considered as an alternative to mimic human development. Methods and Results: In this study, hESCs were differentiated into three lineage cell types to analyze temporal and spatial expression of imprinted genes. Of 19 imprinted genes examined, 15 imprinted genes showed similar transcriptional level among two hESC lines and two human induced pluripotent stem cell (hiPSC) lines. Expressional patterns of most imprinted genes were varied in progenitors and fully differentiated cells which were derived from hESCs. Also, no consistence was observed in the expression pattern of imprinted genes within an imprinting domain during in vitro differentiation of hESCs into three lineage cell types. Conclusions: Transcriptional expression of imprinted genes is regulated in a cell type- specific manner in hESCs during in vitro differentiation.

Efficient Derivation of New Human Embryonic Stem Cell Lines

  • Kim, Sun Jong;Lee, Jeoung Eun;Park, Jong Hyuk;Lee, Jung Bok;Kim, Jin Mee;Yoon, Byung Sun;Song, Ji Min;Roh, Sung Il;Kim, Chul Geun;Yoon, Hyun Soo
    • Molecules and Cells
    • /
    • v.19 no.1
    • /
    • pp.46-53
    • /
    • 2005
  • Human embryonic stem (hES) cells, unlike most cells derived from adult or fetal human tissues, represent a potentially unlimited source of various cell types for basic clinical research. To meet the increased demand for characterized hES cell lines, we established and characterized nine new lines obtained from frozen-thawed pronucleus-stage embryos. In addition, we improved the derivation efficiency from inner cell masses (to 47.4%) and optimized culture conditions for undifferentiated hES cells. After these cell lines had been maintained for over a year in vitro, they were characterized comprehensively for expression of markers of undifferentiated hES cells, karyotype, and in vitro/in vivo differentiation capacity. All of the cell lines were pluripotent, and one cell line was trisomic for chromosome 3. Improved culture techniques for hES cells should make them a good source for diverse applications in regenerative medicine, but further investigation is needed of their basic biology.

iPSC technology-Powerful hand for disease modeling and therapeutic screen

  • Kim, Changsung
    • BMB Reports
    • /
    • v.48 no.5
    • /
    • pp.256-265
    • /
    • 2015
  • Cardiovascular and neurodegenerative diseases are major health threats in many developed countries. Recently, target tissues derived from human embryonic stem (hES) cells and induced pluripotent stem cells (iPSCs), such as cardiomyocytes (CMs) or neurons, have been actively mobilized for drug screening. Knowledge of drug toxicity and efficacy obtained using stem cell-derived tissues could parallel that obtained from human trials. Furthermore, iPSC disease models could be advantageous in the development of personalized medicine in various parts of disease sectors. To obtain the maximum benefit from iPSCs in disease modeling, researchers are now focusing on aging, maturation, and metabolism to recapitulate the pathological features seen in patients. Compared to pediatric disease modeling, adult-onset disease modeling with iPSCs requires proper maturation for full manifestation of pathological features. Herein, the success of iPSC technology, focusing on patient-specific drug treatment, maturation-based disease modeling, and alternative approaches to compensate for the current limitations of patient iPSC modeling, will be further discussed. [BMB Reports 2015; 48(5): 256-265]

THE EFFECT OF GROWTH FACTORS ON OSTEOGENIC DIFFERENTIATION OF ADIPOSE TISSUE-DERIVED STROMAL CELLS (지방기질유래 줄기세포의 골 분화 시 성장인자의 효과)

  • Kim, Uk-Kyu;Choi, Yeon-Sik;Jung, Jin-Sup
    • Journal of the Korean Association of Oral and Maxillofacial Surgeons
    • /
    • v.32 no.4
    • /
    • pp.327-333
    • /
    • 2006
  • Future cell-based therapies such as tissue engineering will benefit from a source of autogenous pluripotent stem cells. There are embryonic stem cells (ESC) and autologous adult stem cells, two general types of stem cells potentilally useful for these applications. But practical use of ESC is limited due to potential problems of cell regulation and ethical considerations. To get bone marrow stem cells is relatively burden to patients because of pain, anesthesia requirement. The ideal stem cells are required of such as the following advantages: easy to obtain, minimal patient discomfort and a capability of yielding enough cell numbers. Adipose autologus tissue taken from intraoral fatty pad or abdomen may represent such a source. Our study designed to demonstrate the ability of human adipose tissue-derived stromal cells (hATSC) from human abdominal adipose tissue diffentiating into osteocyte and adipocyte under culture in vitro conditions. As a result of experiment, we identified stromal cell derived adipose tissue has the multilineage potentiality under appropriate culture conditions. And the adipose stromal cells expressed several mesenchymal stem cell related antigen (CD29, CD44) reactions. Secondary, we compared the culture results of a group of hATSC stimulated with TGF-${\beta}$1, bFGF with a hATSC group without growth factors to confirm whether cytokines have a important role of the proliferation in osteogenic differentiation. The role of cytokines such as TGF-${\beta}$1, bFGF increased hATSC's osteogenic differentiation especially when TGF-${\beta}$1 and bFGF were used together. These results suggest that adipose stromal cells with growth factors could be efficiently available for cell-based bone regeneration.

ENDOTHELIAL PROGENITOR CELLS AND MESENCHYMAL STEM CELLS FROM HUMAN CORD BLOOD (제대혈 내피기원세포 및 간엽줄기세포의 분화에 대한 연구)

  • Kim, Eun-Seok;Kim, Hyun-Ok
    • Journal of the Korean Association of Oral and Maxillofacial Surgeons
    • /
    • v.31 no.1
    • /
    • pp.39-45
    • /
    • 2005
  • Stem cell therapy using mesenchymal stem cells(MSCs) transplantation have been paid attention because of their powerful proliferation and pluripotent differentiating ability. Although umbilical cord blood (UCB) is well known to be a rich source of hematopoietic stem cells with practical and ethical advantages, the presence of mesenchymal stem cells (MSCs) in UCB has been controversial and it remains to be validated. In this study, we examine the presence of MSCs in UCB harvests and the prevalence of them is compared to that of endothelial progenitor cells. For this, CD34+ and CD34- cells were isolated and cultured under the endothelial cell growth medium and mesenchymal stem cell growth medium respectively. The present study showed that ESC-like cells could be isolated and expanded from preterm UCBs but were not acquired efficiently from full-terms. They expressed CD14-, CD34-, CD45-, CD29+, CD44+, CD105+ cell surface marker and could differentiate into adipogenic and osteogenic lineages. Our results suggest that MSCs are fewer in full-term UCB compared to endothelial progenitor cells.

Derivation of endothelial cells from porcine induced pluripotent stem cells by optimized single layer culture system

  • Wei, Renyue;Lv, Jiawei;Li, Xuechun;Li, Yan;Xu, Qianqian;Jin, Junxue;Zhang, Yu;Liu, Zhonghua
    • Journal of Veterinary Science
    • /
    • v.21 no.1
    • /
    • pp.9.1-9.15
    • /
    • 2020
  • Regenerative therapy holds great promise in the development of cures of some untreatable diseases such as cardiovascular diseases, and pluripotent stem cells (PSCs) including induced PSCs (iPSCs) are the most important regenerative seed cells. Recently, differentiation of human PSCs into functional tissues and cells in vitro has been widely reported. However, although porcine reports are rare they are quite essential, as the pig is an important animal model for the in vitro generation of human organs. In this study, we reprogramed porcine embryonic fibroblasts into porcine iPSCs (piPSCs), and differentiated them into cluster of differentiation 31 (CD31)-positive endothelial cells (ECs) (piPSC-derived ECs, piPS-ECs) using an optimized single-layer culture method. During differentiation, we observed that a combination of GSK3β inhibitor (CHIR99021) and bone morphogenetic protein 4 (BMP4) promoted mesodermal differentiation, resulting in higher proportions of CD31-positive cells than those from separate CHIR99021 or BMP4 treatment. Importantly, the piPS-ECs showed comparable morphological and functional properties to immortalized porcine aortic ECs, which are capable of taking up low-density lipoprotein and forming network structures on Matrigel. Our study, which is the first trial on a species other than human and mouse, has provided an optimized single-layer culture method for obtaining ECs from porcine PSCs. Our approach can be beneficial when evaluating autologous EC transplantation in pig models.

Propagation of Human Embryonic Stem Cells on Human Amniotic Fluid Cells as Feeder Cells in Xeno-Free Culture Conditions

  • Jung, Juwon;Baek, Jin Ah;Seol, Hye Won;Choi, Young Min
    • Development and Reproduction
    • /
    • v.20 no.1
    • /
    • pp.63-71
    • /
    • 2016
  • Human embryonic stem cells (hESCs) have been routinely cultured on mouse embryonic fibroblast feeder layers with a medium containing animal materials. For clinical application of hESCs, animal-derived products from the animal feeder cells, animal substrates such as gelatin or Matrigel and animal serum are strictly to be eliminated in the culture system. In this study, we performed that SNUhES32 and H1 were cultured on human amniotic fluid cells (hAFCs) with KO-SR XenoFree and a humanized substrate. All of hESCs were relatively well propagated on hAFCs feeders with xeno-free conditions and they expressed pluripotent stem cell markers, alkaline phosphatase, SSEA-4, TRA1-60, TRA1-81, Oct-4, and Nanog like hESCs cultured on STO or human foreskin fibroblast feeders. In addition, we observed the expression of nonhuman N-glycolylneuraminic acid (Neu5GC) molecules by flow cytometry, which was xenotransplantation components of contamination in hESCs cultured on animal feeder conditions, was not detected in this xeno-free condition. In conclusion, SNUhES32 and H1 could be maintained on hAFCs for humanized culture conditions, therefore, we suggested that new xeno-free conditions for clinical grade hESCs culture will be useful data in future clinical studies.

Stem Cell Biotechnology for Cell Therapy

  • LEE Dong-Ree;KIM Ha Won
    • Biomolecules & Therapeutics
    • /
    • v.13 no.4
    • /
    • pp.199-206
    • /
    • 2005
  • Cell therapy (CT) is a group of techniques to treat human disorders by transplantation of cells which have been processed and propagated independent of the living body. Blood transfusion and bone marrow transplant have been the primary examples of cell therapy. With introduction of stem cell (SC) technologies, however, CT is perceived as the next generation of biologies to treat human diseases such as cancer, neurological diseases, and heart disease. Despite potential of cell therapy, insufficient guidelines have been implemented concerning safety test and regulation of cell therapy. This review addresses the safety issues to be resolved for the cell therapy, especially SC therapy, to be successfully utilized for clinical practice. Adequate donor cell screening must preceed to ensure safety in cell therapy. In terms of SC culture, controlled, standardized practices and procedures should be established. Further molecular studies should be done on SC development and differentiation to enhance safety level in cell therapy. Finally, animal model must be further installed to evaluate toxicity, new concepts, and proliferative potential of SC including alternative feeder layer of animal cells.