Network


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

Hotspot


Dive into the research topics where Soo-Kyung Kang is active.

Publication


Featured researches published by Soo-Kyung Kang.


Cytotherapy | 2009

Therapeutic potential of mesenchymal stromal cells in a mouse breast cancer metastasis model

Bo Sun; Kyoung-Hwan Roh; Jeong-Ran Park; Sae-Rom Lee; Sang-Bum Park; Ji-Won Jung; Soo-Kyung Kang; Yong-Soon Lee; Kyung-Sun Kang

BACKGROUND AIMS Mesenchymal stromal cells (MSC) have been studied intensively in regenerative medicine. However, their therapeutic potential against tumor formation and cancer metastasis is still unclear. The effects of transplantation of MSCs in early-stage of carcinogenesis, should be evaluated. METHODS MSC isolated from human umbilical cord blood (UCB) and adipose tissue (AD) were transplanted in a mouse cancer metastasis model. The effects of MSC on tumor growth and metastasis were analyzed. The effects of transplantation of MSC into the mouse model at very early stage carcinogenesis were also evaluated. RESULTS Human MSC reduced lung metastasis and inhibited the growth of human breast cancer cells by inducing apoptosis. In addition, transplantation of both UCB and AD MSC into a cancer model with no detectable clinical symptoms did not appear to promote tumor growth or metastasis. CONCLUSIONS We evaluated the effect of MSC derived from human UCB and AD tissue in a tumor model. Our findings may help to elucidate the interaction between cancer cells and MSC, as well as the application of MSC to clinical trials.


Cellular and Molecular Life Sciences | 2010

Histone deacetylase controls adult stem cell aging by balancing the expression of polycomb genes and jumonji domain containing 3

Ji-Won Jung; Seunghee Lee; Min-Soo Seo; Sang-Bum Park; Andreas Kurtz; Soo-Kyung Kang; Kyung-Sun Kang

Aging is linked to loss of the self-renewal capacity of adult stem cells. Here, we observed that human multipotent stem cells (MSCs) underwent cellular senescence in vitro. Decreased expression of histone deacetylases (HDACs), followed by downregulation of polycomb group genes (PcGs), such as BMI1, EZH2 and SUZ12, and by upregulation of jumonji domain containing 3 (JMJD3), was observed in senescent MSCs. Similarly, HDAC inhibitors induced cellular senescence through downregulation of PcGs and upregulation of JMJD3. Regulation of PcGs was associated with HDAC inhibitor-induced hypophosphorylation of RB, which causes RB to bind to and decrease the transcriptional activity of E2F. JMJD3 expression regulation was dependant on histone acetylation status at its promoter regions. A histone acetyltransferase (HAT) inhibitor prevented replicative senescence of MSCs. These results suggest that HDAC activity might be important for MSC self-renewal by balancing PcGs and JMJD3 expression, which govern cellular senescence by p16INK4A regulation.


Cellular and Molecular Life Sciences | 2011

Histone deacetylase regulates high mobility group A2-targeting microRNAs in human cord blood-derived multipotent stem cell aging

Seunghee Lee; Ji-Won Jung; Sang-Bum Park; Kyoung-Hwan Roh; Su Yeon Lee; Ju Han Kim; Soo-Kyung Kang; Kyung-Sun Kang

Cellular senescence involves a reduction in adult stem cell self-renewal, and epigenetic regulation of gene expression is one of the main underlying mechanisms. Here, we observed that the cellular senescence of human umbilical cord blood-derived multipotent stem cells (hUCB-MSCs) caused by inhibition of histone deacetylase (HDAC) activity leads to down-regulation of high mobility group A2 (HMGA2) and, on the contrary, to up-regulation of p16INK4A, p21CIP1/WAF1 and p27KIP1. We found that let-7a1, let-7d, let-7f1, miR-23a, miR-26a and miR-30a were increased during replicative and HDAC inhibitor-mediated senescence of hUCB-MSCs by microRNA microarray and real-time quantitative PCR. Furthermore, the configurations of chromatins beading on these miRNAs were prone to transcriptional activation during HDAC inhibitor-mediated senescence. We confirmed that miR-23a, miR-26a and miR-30a inhibit HMGA2 to accelerate the progress of senescence. These findings suggest that HDACs may play important roles in cellular senescence by regulating the expression of miRNAs that target HMGA2 through histone modification.


PLOS ONE | 2010

Implication of NOD1 and NOD2 for the Differentiation of Multipotent Mesenchymal Stem Cells Derived from Human Umbilical Cord Blood

Hyung-Sik Kim; Tae-Hoon Shin; Se-Ran Yang; Min-Soo Seo; Dong-Jae Kim; Soo-Kyung Kang; Jong-Hwan Park; Kyung-Sun Kang

Toll-like receptors (TLRs) and Nod-like receptors (NLRs) are known to trigger an innate immune response against microbial infection. Although studies suggest that activation of TLRs modulate the function of mesenchymal stem cells (MSCs), little is known about the role of NLRs on the MSC function. In this study, we investigated whether NOD1 and NOD2 regulate the functions of human umbilical cord blood-derived MSCs (hUCB-MSCs). The genes of TLR2, TLR4, NOD1, and NOD2 were expressed in hUCB-MSCs. Stimulation with each agonist (Pam3CSK4 for TLR2, LPS for TLR4, Tri-DAP for NOD1, and MDP for NOD2) led to IL-8 production in hUCB-MSC, suggesting the expressed receptors are functional in hUCB-MSC. CCK-8 assay revealed that none of agonist influenced proliferation of hUCB-MSCs. We next examined whether TLR and NLR agonists affect osteogenic-, adipogenic-, and chondrogenic differentiation of hUCB-MSCs. Pam3CSK4 and Tri-DAP strongly enhanced osteogenic differentiation and ERK phosphorylation in hUCB-MSCs, and LPS and MDP also slightly did. Treatment of U0126 (MEK1/2 inhibitor) restored osteogenic differentiation enhanced by Pam3CSK4. Tri-DAP and MDP inhibited adipogenic differentiation of hUCB-MSCs, but Pam3CSK4 and LPS did not. On chondrogenic differentiation, all TLR and NLR agonists could promote chondrogenesis of hUCB-MSCs with difference in the ability. Our findings suggest that NOD1 and NOD2 as well as TLRs are involved in regulating the differentiation of MSCs.


PLOS ONE | 2010

REX-1 expression and p38 MAPK activation status can determine proliferation/differentiation fates in human mesenchymal stem cells.

Dilli Ram Bhandari; Kwang-Won Seo; Kyoung-Hwan Roh; Ji-Won Jung; Soo-Kyung Kang; Kyung-Sun Kang

Background REX1/ZFP42 is a well-known embryonic stem cell (ESC) marker. However, the role of REX1, itself, is relatively unknown because the function of REX1 has only been reported in the differentiation of ESCs via STAT signaling pathways. Human mesenchymal stem cells (hMSCs) isolated from young tissues and cancer cells express REX1. Methodology/Principal Finding Human umbilical cord blood-derived MSCs (hUCB-MSCs) and adipose tissue-derived MSCs (hAD-MSCs) strongly express REX1 and have a lower activation status of p38 MAPK, but bone marrow-derived MSCs (hBM-MSCs) have weak REX1 expression and higher activation of p38 MAPK. These results indicated that REX1 expression in hMSCs was positively correlated with proliferation rates but inversely correlated with the phosphorylation of p38 MAPK. In hUCB-MSCs, the roles of REX1 and p38 MAPK were investigated, and a knockdown study was performed using a lentiviral vector-based small hairpin RNA (shRNA). After REX1 knockdown, decreased cell proliferation was observed. In REX1 knocked-down hUCB-MSCs, the osteogenic differentiation ability deteriorated, but the adipogenic potential increased or was similar to that observed in the controls. The phosphorylation of p38 MAPK in hUCB-MSCs significantly increased after REX1 knockdown. After p38 MAPK inhibitor treatment, the cell growth in REX1 knocked-down hUCB-MSCs almost recovered, and the suppressed expression levels of CDK2 and CCND1 were also restored. The expression of MKK3, an upstream regulator of p38 MAPK, significantly increased in REX1 knocked-down hUCB-MSCs. The direct binding of REX1 to the MKK3 gene was confirmed by a chromatin immunoprecipitation (ChIP) assay. Conclusions/Significance These findings showed that REX1 regulates the proliferation/differentiation of hMSCs through the suppression of p38 MAPK signaling via the direct suppression of MKK3. Therefore, p38 MAPK and REX-1 status can determine the cell fate of adult stem cells (ASCs). These results were the first to show the role of REX1 in the proliferation/differentiation of ASCs.


Cell Transplantation | 2011

Human umbilical cord blood-derived mesenchymal stem cells protect against neuronal cell death and ameliorate motor deficits in Niemann Pick type C1 mice.

Yoojin Seo; Se-Ran Yang; Min Ki Jee; Eun Kyung Joo; Kyung-Hwan Roh; Min-Soo Seo; Tae Hee Han; So Yeong Lee; Pan Dong Ryu; Ji-Won Jung; Kwang-Won Seo; Soo-Kyung Kang; Kyung-Sun Kang

Niemann Pick disease type C1 (NPC) is an autosomal recessive disease characterized by progressive neurological deterioration leading to premature death. In this study, we hypothesized that human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) have the multifunctional abilities to ameliorate NPC symptoms in the brain. To test this hypothesis, hUCB-MSCs were transplanted into the hippocampus of NPC mice in the early asymptomatic stage. This transplantation resulted in the recovery of motor function in the Rota Rod test and impaired cholesterol homeostasis leading to increased levels of cholesterol efflux-related genes such as LXRα, ABCA1, and ABCG5 while decreased levels of 3-hydroxy-3-methylglutaryl coenzyme A reductase were observed in NPC mice. In the cerebrum, hUCB-MSCs enhanced neuronal cell survival and proliferation, where they directly differentiated into electrically active MAP2-positive neurons as demonstrated by whole-cell patch clamping. In addition, we observed that hUCB-MSCs reduced Purkinje neuronal loss by suppression of inflammatory and apoptotic signaling in the cerebellum as shown by immunohistochemistry. We further investigated how hUCB-MSCs enhance cellular survival and inhibit apoptosis in NPC mice. Neuronal cell survival was associated with increased PI3K/AKT and JAK2/STAT3 signaling; moreover, hUCB-MSCs modulated the levels of GABA/glutamate transporters such as GAT1, EAAT2, EAAT3, and GAD6 in NPC mice as assessed by Western blot analysis. Taken together, our findings suggest that hUCB-MSCs might play multifunctional roles in neuronal cell survival and ameliorating motor deficits of NPC mice.


Neuroscience Letters | 2009

Transplantation of mesenchymal stem cells enhances axonal outgrowth and cell survival in an organotypic spinal cord slice culture

Jung-Sun Cho; Hwan-Woo Park; Sangkyu Park; Sangho Roh; Soo-Kyung Kang; Ki-Suk Paik; Mi-Sook Chang

Mesenchymal stem cells (MSCs) have demonstrated a measurable therapeutic effect following transplantation into animal models of spinal cord injury. However, the mechanism(s) by which transplanted cells promote nerve regeneration and/or functional recovery remains indeterminate. Several studies have suggested that MSCs promote tissue repair via secretion of trophic factors, but delineating the effect of such factors is difficult due to the complexity of the in vivo systems. Therefore, we developed an organotypic spinal cord slice culture system that can be sustained for sufficient periods of time in vitro to evaluate nerve regeneration as an ex vivo model of spinal cord injury. Using this model, we demonstrate that treatment of lumbar slices of spinal cord with lysolecithin induced a significant degree of cell death and demyelination of nerve fibers, but that these effects were ameliorated to a significant extent following co-culture of slices with human MSCs (hMSCs). The results indicate that transplanted hMSCs alter the tissue microenvironment in a way that promotes survival of endogenous cells, including injured neurons, immature oligodendrocytes and oligodendrocyte progenitor cells. This ex vivo culture system represents a useful tool to further dissect the mechanism(s) by which MSCs promote regeneration of injured nervous tissue.


Biomaterials | 2011

Wnt5a-mediating neurogenesis of human adipose tissue-derived stem cells in a 3D microfluidic cell culture system.

Jeein Choi; Sohyeun Kim; Jinsun Jung; Youngbin Lim; Kyung-Sun Kang; Seungsu Park; Soo-Kyung Kang

In stem cell biology, cell plasticity refers to the ability of stem cells to differentiate into a variety of cell lineages. Recently, cell plasticity has been used to refer to the ability of a given cell type to reversibly de-differentiate, re-differentiate, or transdifferentiate in response to specific stimuli. These processes are regulated by multiple intracellular and extracellular growth and differentiation factors, including low oxygen. Our recent study showed that 3D microfluidic cell culture induces activation of the Wnt5A/β-catenin signaling pathway in hATSCs (human Adipose Tissue-derived Stem Cells). This resulted in self renewal and transdifferentiation of hATSCs into neurons. To improve neurogenic potency of hATSCs in response to low oxygen and other unknown physical factors, we developed a gel-free 3D microfluidic cell culture system (3D-μFCCS). The functional structure was developed for the immobilization of 3D multi-cellular aggregates in a microfluidic channel without the use of a matrix on the chip. Growth of hATSCs neurosphere grown on a chip was higher than the growth of control cells grown in a culture dish. Induction of differentiation in the Chip system resulted in a significant increase in the induction of neuronal-like cell structures and the presentation of TuJ or NF160 positive long neuritis compared to control cells after active migration from the center of the microfluidic channel layer to the outside of the microfluidic channel layer. We also observed that the chip neurogenesis system induced a significantly higher level of GABA secreting neurons and, in addition, almost 60% of cells were GABA + cells. Finally, we observed that 1 month of after the transplantation of each cell type in a mouse SCI lesion, chip cultured and neuronal differentiated hATSCs exhibited the ability to effectively transdifferentiate into NF160 + motor neurons at a high ratio. Interestingly, our CHIP/PCR analysis revealed that HIF1α-induced hATSCs neurogenesis on the chip. This induction was a result of the direct binding of HIF1α to the regulatory regions of the Oct4 and β-catenin genes in nucleus. In the Chip culture of hATSCs that we developed, a low oxygen microenvironment was induced. The low oxygen level induced HIF1α expression, which resulted in increased expression of Wnt5A/β-catenin and Oct4 via the direct binding of HIF1α to the regulatory regions of β-catenin and Oct4.


Growth Factors Journal | 2009

bFGF enhances the IGFs-mediated pluripotent and differentiation potentials in multipotent stem cells

Sang-Bum Park; Kyung-Rok Yu; Ji-Won Jung; Sae-Rom Lee; Kyoung-Hwan Roh; Min-Soo Seo; Jeong-Ran Park; Soo-Kyung Kang; Yong-Soon Lee; Kyung-Sun Kang

It has widely been reported that basic fibroblast growth factor (bFGF) promotes proliferation of human stem cells and contributes to the maintenance of their self-renewal capability through repeated replications. In contrast to embryonic stem cells (ESCs), the effects of growth factors on adult stem cells are poorly understood. In human umbilical cord blood-derived multipotent stem cells (hUCB-MSCs), bFGF is associated with an increased number of proliferating cells. Furthermore, expression levels of ESC markers were increased after treatment with bFGF. bFGF also increased the expression of FGFR, which in turn increased expression of insulin-like growth factor (IGFs). Since IGFs exert autocrine and paracrine effects on stem cells, bFGF-mediated release of IGFs from hUCB-MSCs might enhance FGFR1 and IGF1R expression in neighboring cells. These receptors could subsequently regulate the effects of bFGF and IGFs in adult stem cells. These results suggest that positive feedback regulation of bFGF and IGFs leads to proliferation of hUCB-MSCs.


Archive | 2012

Cancer Stem Cells in Brain Gliomas

Mee-Gyeung Khang; Soo-Kyung Kang

Stem cells are defined as cells that self-renew indefinitely and give rise to differentiated cells (Reya et al., 2001). During the last decade, it has become clear that almost all tissues contain tissue-specific stem cells that continuously generate the residential differentiated cells responsible for tissue functions and homeostasis (Kondo and Raff, 2000). In addition, there is increasing evidence that malignant tumors such as leukemia, myeliod leukemia and breast cancers contain cancer stem cells (CSCs) that maintain the characteristics of tissue-specific stem cells and are malignant (Al-Hajj et al., 2003; Ladipot et al., 2004).

Collaboration


Dive into the Soo-Kyung Kang's collaboration.

Top Co-Authors

Avatar

Kyung-Sun Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ji-Won Jung

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Kyoung-Hwan Roh

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Min-Soo Seo

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Sang-Bum Park

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hwan-Woo Park

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jeong-Ran Park

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jung-Sun Cho

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ki-Suk Paik

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Kwang-Won Seo

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge