Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Soon Won Choi is active.

Publication


Featured researches published by Soon Won Choi.


Stem Cells | 2012

CD49f Enhances Multipotency and Maintains Stemness Through the Direct Regulation of OCT4 and SOX2

Kyung-Rok Yu; Se-Ran Yang; Ji-Won Jung; Hyongbum Kim; Kinarm Ko; Dong Wook Han; Sang-Bum Park; Soon Won Choi; Soo-Kyung Kang; Hans R. Schöler; Kyung-Sun Kang

CD49f (integrin subunit α6) regulates signaling pathways in a variety of cellular activities. However, the role of CD49f in regulating the differentiation and pluripotency of stem cells has not been fully investigated. Therefore, in this study, human mesenchymal stem cells (hMSCs) were induced to form spheres under nonadherent culture conditions, and we found that the CD49f‐positive population was enriched in MSC spheres compared with MSCs in a monolayer. The expression of CD49f regulated the ability of hMSCs to form spheres and was associated with an activation of the phosphatidylinositol 3‐kinase (PI3K)/AKT signaling pathway. Furthermore, the forced expression of CD49f modulated the proliferation and differentiation potentials of hMSCs through prolonged activation of PI3K/AKT and suppressed the level of p53. We showed that the pluripotency factors OCT4 and SOX2 were recruited to the putative promoter region of CD49f, indicating that OCT4 and SOX2 play positive roles in the expression of CD49f. Indeed, CD49f expression was upregulated in human embryonic stem cells (hESCs) compared with hMSCs. The elevated level of CD49f expression was significantly decreased upon embryoid body formation in hESCs. In hESCs, the knockdown of CD49f downregulated PI3K/AKT signaling and upregulated the level of p53, inducing differentiation into three germ layers. Taken together, our data suggest that the cell‐surface protein CD49f has novel and dynamic roles in regulating the differentiation potential of hMSCs and maintaining pluripotency. STEM CELLS 2012;30:876–887


Gastroenterology | 2013

Human umbilical cord blood mesenchymal stem cells reduce colitis in mice by activating NOD2 signaling to COX2.

Hyung Sik Kim; Tae–Hoon Shin; Byung-Chul Lee; Kyung Rok Yu; Yoojin Seo; Seunghee Lee; Min Soo Seo; In–Sun Hong; Soon Won Choi; Kwang Won Seo; Gabriel Núñez; Jong Hwan Park; Kyung Sun Kang

BACKGROUND & AIMS Decreased levels or function of nucleotide-binding oligomerization domain 2 (NOD2) are associated with Crohns disease. NOD2 regulates intestinal inflammation, and also is expressed by human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs), to regulate their differentiation. We investigated whether NOD2 is required for the anti-inflammatory activities of MSCs in mice with colitis. METHODS Colitis was induced in mice by administration of dextran sulfate sodium or trinitrobenzene sulfonic acid. Mice then were given intraperitoneal injections of NOD2-activated hUCB-MSCs; colon tissues and mesenteric lymph nodes were collected for histologic analyses. A bromodeoxyuridine assay was used to determine the ability of hUCB-MSCs to inhibit proliferation of human mononuclear cells in culture. RESULTS Administration of hUCB-MSCs reduced the severity of colitis in mice. The anti-inflammatory effects of hUCB-MSCs were greatly increased by activation of NOD2 by its ligand, muramyl dipeptide (MDP). Administration of NOD2-activated hUCB-MSCs increased anti-inflammatory responses in colons of mice, such as production of interleukin (IL)-10 and infiltration by T regulatory cells, and reduced production of inflammatory cytokines. Proliferation of mononuclear cells was inhibited significantly by co-culture with hUCB-MSCs that had been stimulated with MDP. MDP induced prolonged production of prostaglandin (PG)E2 in hUCB-MSCs via the NOD2-RIP2 pathway, which suppressed proliferation of mononuclear cells derived from hUCB. PGE2 produced by hUCB-MSCs in response to MDP increased production of IL-10 and T regulatory cells. In mice, production of PGE2 by MSCs and subsequent production of IL-10 were required to reduce the severity of colitis. CONCLUSIONS Activation of NOD2 is required for the ability of hUCB-MSCs to reduce the severity of colitis in mice. NOD2 signaling increases the ability of these cells to suppress mononuclear cell proliferation by inducing production of PGE2.


Stem Cells | 2015

Human Umbilical Cord Blood Mesenchymal Stem Cell‐Derived PGE2 and TGF‐β1 Alleviate Atopic Dermatitis by Reducing Mast Cell Degranulation

Hyung-Sik Kim; Jun-Won Yun; Tae-Hoon Shin; Sung-Hoon Lee; Byung-Chul Lee; Kyung-Rok Yu; Yoojin Seo; Seunghee Lee; Taewook Kang; Soon Won Choi; Kwang-Won Seo; Kyung-Sun Kang

Mesenchymal stem cell (MSC) is a promising tool for the therapy of immune disorders. However, their efficacy and mechanisms in treating allergic skin disorders are less verified. We sought to investigate the therapeutic efficacy of human umbilical cord blood‐derived MSCs (hUCB‐MSCs) against murine atopic dermatitis (AD) and to explore distinct mechanisms that regulate their efficacy. AD was induced in mice by the topical application of Dermatophagoides farinae. Naïve or activated‐hUCB‐MSCs were administered to mice, and clinical severity was determined. The subcutaneous administration of nucleotide‐binding oligomerization domain 2 (NOD2)‐activated hUCB‐MSCs exhibited prominent protective effects against AD, and suppressed the infiltration and degranulation of mast cells (MCs). A β‐hexosaminidase assay was performed to evaluate the effect of hUCB‐MSCs on MC degranulation. NOD2‐activated MSCs reduced the MC degranulation via NOD2‐cyclooxygenase‐2 signaling. In contrast to bone marrow‐derived MSCs, hUCB‐MSCs exerted a cell‐to‐cell contact‐independent suppressive effect on MC degranulation through the higher production of prostaglandin E2 (PGE2). Additionally, transforming growth factor (TGF)‐β1 production from hUCB‐MSCs in response to interleukin‐4 contributed to the attenuation of MC degranulation by downregulating FcεRI expression in MCs. In conclusion, the subcutaneous application of NOD2‐activated hUCB‐MSCs can efficiently ameliorate AD, and MSC‐derived PGE2 and TGF‐β1 are required for the inhibition of MC degranulation. Stem Cells 2015;33:1254–1266


PLOS ONE | 2014

A p38 MAPK-Mediated Alteration of COX-2/PGE2 Regulates Immunomodulatory Properties in Human Mesenchymal Stem Cell Aging

Kyung-Rok Yu; Jin Young Lee; Hyung-Sik Kim; In-Sun Hong; Soon Won Choi; Yoojin Seo; Insung Kang; Jae-Jun Kim; Byung-Chul Lee; Seunghee Lee; Andreas Kurtz; Kwang-Won Seo; Kyung-Sun Kang

Because human mesenchymal stem cells (hMSC) have profound immunomodulatory effects, many attempts have been made to use hMSCs in preclinical and clinical trials. For hMSCs to be used in therapy, a large population of hMSCs must be generated by in vitro expansion. However, the immunomodulatory changes following the in vitro expansion of hMSCs have not been elucidated. In this study, we evaluated the effect of replicative senescence on the immunomodulatory ability of hMSCs in vitro and in vivo. Late-passage hMSCs showed impaired suppressive effect on mitogen-induced mononuclear cell proliferation. Strikingly, late-passage hMSCs had a significantly compromised protective effect against mouse experimental colitis, which was confirmed by gross and histologic examination. Among the anti-inflammatory cytokines, the production of prostaglandin E2 (PGE2) and the expression of its primary enzyme, cyclooxygenase-2 (COX-2), were profoundly increased by pre-stimulation with interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α), and this response was significantly decreased with consecutive passages. We demonstrated that the impaired phosphorylation activity of p38 MAP kinase (p38 MAPK) in late-passage hMSCs led to a compromised immunomodulatory ability through the regulation of COX-2. In conclusion, our data indicate that the immunomodulatory ability of hMSCs gradually declines with consecutive passages via a p38-mediated alteration of COX-2 and PGE2 levels.


Cell Death and Disease | 2016

Human umbilical cord blood-stem cells direct macrophage polarization and block inflammasome activation to alleviate rheumatoid arthritis

Tae-Hoon Shin; Hyung-Sik Kim; Taewook Kang; Byung-Chul Lee; Hwa-Yong Lee; Yoon-Jin Kim; Ji-Hee Shin; Yoojin Seo; Soon Won Choi; Seunghee Lee; Ki-Chul Shin; Kwang-Won Seo; Kyung-Sun Kang

Rheumatoid arthritis (RA) is a long-lasting intractable autoimmune disorder, which has become a substantial public health problem. Despite widespread use of biologic drugs, there have been uncertainties in efficacy and long-term safety. Mesenchymal stem cells (MSCs) have been suggested as a promising alternative for the treatment of RA because of their immunomodulatory properties. However, the precise mechanisms of MSCs on RA-related immune cells are not fully elucidated. The aim of this study was to investigate the therapeutic potential of human umbilical cord blood-derived MSCs (hUCB-MSCs) as a new therapeutic strategy for patients with RA and to explore the mechanisms underlying hUCB-MSC-mediated immunomodulation. Mice with collagen-induced arthritis (CIA) were administered with hUCB-MSCs after the onset of disease, and therapeutic efficacy was assessed. Systemic delivery of hUCB-MSCs significantly ameliorated the severity of CIA to a similar extent observed in the etanercept-treated group. hUCB-MSCs exerted this therapeutic effect by regulating macrophage function. To verify the regulatory effects of hUCB-MSCs on macrophages, macrophages were co-cultured with hUCB-MSCs. The tumor necrosis factor (TNF)-α-mediated activation of cyclooxygenase-2 and TNF-stimulated gene/protein 6 in hUCB-MSCs polarized naive macrophages toward an M2 phenotype. In addition, hUCB-MSCs down-regulated the activation of nucleotide-binding domain and leucine-rich repeat pyrin 3 inflammasome via a paracrine loop of interleukin-1β signaling. These immune-balancing effects of hUCB-MSCs were reproducible in co-culture experiments using peripheral blood mononuclear cells from patients with active RA. hUCB-MSCs can simultaneously regulate multiple cytokine pathways in response to pro-inflammatory cytokines elevated in RA microenvironment, suggesting that treatment with hUCB-MSCs could be an attractive candidate for patients with treatment-refractory RA.


Scientific Reports | 2015

Growth arrest and forced differentiation of human primary glioblastoma multiforme by a novel small molecule

Taewook Kang; Soon Won Choi; Se-Ran Yang; Tae-Hoon Shin; Hyung-Sik Kim; Kyung-Rok Yu; In-Sun Hong; Seonggu Ro; Joong Myung Cho; Kyung-Sun Kang

Glioblastoma multiforme is the most common malignant brain tumor in adults, with an average survival of less than one year due to its resistance to therapy. Recent studies reported that GBM initiates from CD133-expressing cancer stem cells (CSC). However, the efficacy of CSC targeting is limited. A newly developed approach in cancer treatment is the forced differentiation of cancer cells. Here, we show that the treatment of the novel small molecule, CG500354, into CD133-expressing human primary GBM cells induces growth arrest by cell cycle regulators, p53, p21, p27 and phase-specific cyclins, and neural differentiation, as confirmed by neural progenitor/precursor markers, nestin, GFAP and Tuj1. When GBM-derived cells caused the tumors in NOD/SCID mice, CG500354 induced GBM-derived cells differentiation into Tuj1 and GFAP expressing cells. We next demonstrated that CG500354 plays a tumor-suppressive role via cAMP/CREB signaling pathway. CG500354 increases not only the extracellular cAMP level but also the protein level of PKA and CREB. Additionally, both mimetic substances, Forskolin and Rolipram, revealed comparable results with CG500354. Our findings indicate that induction of growth arrest and neural differentiation via cAMP/CREB signaling pathway by CG500354 treatment suggests the novel targeting of PDE4D in the development of new drugs for brain tumor therapy.


Scientific Reports | 2016

PGE2 maintains self-renewal of human adult stem cells via EP2-mediated autocrine signaling and its production is regulated by cell-to-cell contact.

Byung-Chul Lee; Hyung-Sik Kim; Tae-Hoon Shin; Insung Kang; Jin Young Lee; Jae-Jun Kim; Hyun Kyoung Kang; Yoojin Seo; Seunghee Lee; Kyung-Rok Yu; Soon Won Choi; Kyung-Sun Kang

Mesenchymal stem cells (MSCs) possess unique immunomodulatory abilities. Many studies have elucidated the clinical efficacy and underlying mechanisms of MSCs in immune disorders. Although immunoregulatory factors, such as Prostaglandin E2 (PGE2), and their mechanisms of action on immune cells have been revealed, their effects on MSCs and regulation of their production by the culture environment are less clear. Therefore, we investigated the autocrine effect of PGE2 on human adult stem cells from cord blood or adipose tissue, and the regulation of its production by cell-to-cell contact, followed by the determination of its immunomodulatory properties. MSCs were treated with specific inhibitors to suppress PGE2 secretion, and proliferation was assessed. PGE2 exerted an autocrine regulatory function in MSCs by triggering E-Prostanoid (EP) 2 receptor. Inhibiting PGE2 production led to growth arrest, whereas addition of MSC-derived PGE2 restored proliferation. The level of PGE2 production from an equivalent number of MSCs was down-regulated via gap junctional intercellular communication. This cell contact-mediated decrease in PGE2 secretion down-regulated the suppressive effect of MSCs on immune cells. In conclusion, PGE2 produced by MSCs contributes to maintenance of self-renewal capacity through EP2 in an autocrine manner, and PGE2 secretion is down-regulated by cell-to-cell contact, attenuating its immunomodulatory potency.


Stem Cells | 2008

Androgenetic Embryonic Stem Cells Form Neural Progenitor Cells In Vivo and In Vitro

Timo C. Dinger; Sigrid Eckardt; Soon Won Choi; Guadelupe Camarero; Satoshi Kurosaka; Vroni Hornich; K. John McLaughlin; Albrecht M. Müller

Uniparental zygotes with two paternal (androgenetic [AG]) or two maternal (gynogenetic [GG]; parthenogenetic [PG]) genomes are not able to develop into viable offspring but can form blastocysts from which embryonic stem cells (ESCs) can be derived. Although some aspects of the in vitro and in vivo differentiation potential of PG and GG ESCs of several species have been studied, the developmental capacity of AG ESCs is much less clear. Here, we investigate the potential of murine AG ESCs to undergo neural differentiation. We observed that AG ESCs differentiate in vitro into pan‐neural progenitor cells (pnPCs) that further give rise to cells that express neuronal‐ and astroglial‐specific markers. Neural progeny of in vitro‐differentiated AG ESCs exhibited fidelity of expression of six imprinted genes analyzed, with the exception of Ube3a. Bisulfite sequencing for two imprinting control regions suggested that pnPCs predominantly maintained their methylation pattern. Following blastocyst injection of AG and biparental (normal fertilized [N]) ESCs, we found widespread and evenly distributed contribution of ESC‐derived cells in both AG and N chimeric early fetal brains. AG and N ESC‐derived cells isolated from chimeric fetal brains by fluorescence‐activated cell sorting exhibited similar neurosphere‐initiating cell frequencies and neural multilineage differentiation potential. Our results indicate that AG ESC‐derived neural progenitor/stem cells do not differ from N neural progenitor/stem cells in their self‐renewal and neural multilineage differentiation potential.


Stem Cell Reviews and Reports | 2015

miR-410 Inhibition Induces RPE Differentiation of Amniotic Epithelial Stem Cells via Overexpression of OTX2 and RPE65

Soon Won Choi; Jae-Jun Kim; Min-Soo Seo; Sang-Bum Park; Taewook Kang; Jin Young Lee; Byung-Chul Lee; Insung Kang; Tae-Hoon Shin; Hyung-Sik Kim; Kyung-Rok Yu; Kyung-Sun Kang

The retinal pigment epithelium (RPE) is a highly specialized cell type located between the choroid and neural retina of the eye. RPE degeneration causes irreversible visual impairment, extending to blindness. Cell therapy has recently emerged as a potential therapeutic approach for retinal degeneration. MicroRNA-based differentiation of stem cells is a new strategy for producing tissue-specific cell types. In this study, we developed a novel microRNA-based strategy for RPE induction from human amniotic epithelial stem cells (AESCs). We identified microRNAs involved in RPE development in AESCs. Of 29 putative human RPE-relevant microRNAs, microRNA-410 (miR-410) was predicted to target multiple RPE development-relevant genes. Inhibition of miR-410 induces overexpression of immature and mature RPE-specific factors, including OTX2, RPE65, Bestrophin and EMMPRIN. These RPE-like cells were morphologically altered toward a cobblestone-like shape and were able to phagocytize microbeads. We showed that miR-410 directly regulates predicted target genes OTX2 and RPE65. Our microRNA-based strategy demonstrated RPE differentiation in AESCs by treatment of an antisense microRNA-410 (anti-miR-410), without the use of additional factors or exogenous transduction. These findings suggest that miR-410 inhibition can be a useful tool for directed cell differentiation and an attractive method for cell therapy in human retinal degenerative diseases.


Biochimica et Biophysica Acta | 2014

Excessive microglial activation aggravates olfactory dysfunction by impeding the survival of newborn neurons in the olfactory bulb of Niemann–Pick disease type C1 mice

Yoojin Seo; Hyung-Sik Kim; Yooyoung Shin; Insung Kang; Soon Won Choi; Kyung-Rok Yu; Kwang-Won Seo; Kyung-Sun Kang

Progressive olfactory impairment is one of the earliest markers of neurodegeneration. However, the underlying mechanism for this dysfunction remains unclear. The present study investigated the possible role of microgliosis in olfactory deficits using a mouse model of Niemann-Pick disease type C1 (NPC1), which is an incurable neurodegenerative disorder with disrupted lipid trafficking. At 7weeks of age, NPC1 mutants showed a distinct olfactory impairment in an olfactory test compared with age-matched wild-type controls (WT). The marked loss of olfactory sensory neurons within the NPC1 affected olfactory bulb (NPC1-OB) suggests that NPC1 dysfunction impairs olfactory structure. Furthermore, the pool of neuroblasts in the OB was diminished in NPC1 mice despite the intact proliferative capacity of neural stem/progenitor cells in the subventricular zone. Instead, pro-inflammatory proliferating microglia accumulated extensively in the NPC1-OB as the disease progressed. To evaluate the impact of abnormal microglial activation on olfaction in NPC1 mice, a microglial inhibition study was performed using the anti-inflammatory agent Cyclosporin A (CsA). Importantly, long-term CsA treatment in NPC1 mice reduced reactive microgliosis, restored the survival of newly generated neurons in the OB and improved overall performance on the olfactory test. Therefore, our study highlights the possible role of microglia in the regulation of neuronal turnover in the OB and provides insight into the possible therapeutic applications of microglial inhibition in the attenuation or reversal of olfactory impairment.

Collaboration


Dive into the Soon Won Choi's collaboration.

Top Co-Authors

Avatar

Kyung-Sun Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hyung-Sik Kim

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Yoojin Seo

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Insung Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jae-Jun Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jin Young Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Tae-Hoon Shin

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Byung-Chul Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ji-Hee Shin

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Kyung-Rok Yu

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge