Network


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

Hotspot


Dive into the research topics where So Young Chun is active.

Publication


Featured researches published by So Young Chun.


Journal of Korean Medical Science | 2011

Apoptotic Effects of Genistein, Biochanin-A and Apigenin on LNCaP and PC-3 Cells by p21 through Transcriptional Inhibition of Polo-like Kinase-1

Young Jin Seo; Bum Soo Kim; So Young Chun; Yoon Kyu Park; Ku Seong Kang; Tae Gyun Kwon

Natural isoflavones and flavones are important dietary factors for prostate cancer prevention. We investigated the molecular mechanism of these compounds (genistein, biochanin-A and apigenin) in PC-3 (hormone-independent/p53 mutant type) and LNCaP (hormone-dependent/p53 wild type) prostate cancer cells. A cell growth rate and apoptotic activities were analyzed in different concentrations and exposure time to evaluate the antitumor activities of genistein, biochanin-A and apigenin. The real time PCR and Western blot analysis were performed to investigate whether the molecular mechanism of these compounds are involving the p21 and PLK-1 pathway. Apoptosis of prostate cancer cells was associated with p21 up-regulation and PLK-1 suppression. Exposure of genistein, biochanin-A and apigenin on LNCaP and PC-3 prostate cancer cells resulted in same pattern of cell cycle arrest and apoptosis. The inhibition effect for cell proliferation was slightly greater in LNCaP than PC-3 cells. In conclusion, flavonoids treatment induces up-regulation of p21 expression, and p21 inhibits transcription of PLK-1, which promotes apoptosis of cancer cells.


BMC Medicine | 2012

Human amniotic fluid stem cell injection therapy for urethral sphincter regeneration in an animal model

Bum Soo Kim; So Young Chun; Jong Kil Lee; Hyun Ju Lim; Jae-sung Bae; Ho-Yun Chung; Anthony Atala; Shay Soker; James J. Yoo; Tae Gyun Kwon

BackgroundStem cell injection therapies have been proposed to overcome the limited efficacy and adverse reactions of bulking agents. However, most have significant limitations, including painful procurement, requirement for anesthesia, donor site infection and a frequently low cell yield. Recently, human amniotic fluid stem cells (hAFSCs) have been proposed as an ideal cell therapy source. In this study, we investigated whether periurethral injection of hAFSCs can restore urethral sphincter competency in a mouse model.MethodsAmniotic fluids were collected and harvested cells were analyzed for stem cell characteristics and in vitro myogenic differentiation potency. Mice underwent bilateral pudendal nerve transection to generate a stress urinary incontinence (SUI) model and received either periurethral injection of hAFSCs, periurethral injection of Plasma-Lyte (control group), or underwent a sham (normal control group).For in vivo cell tracking, cells were labeled with silica-coated magnetic nanoparticles containing rhodamine B isothiocyanate (MNPs@SiO2 (RITC)) and were injected into the urethral sphincter region (n = 9). Signals were detected by optical imaging. Leak point pressure and closing pressure were recorded serially after injection.Tumorigenicity of hAFSCs was evaluated by implanting hAFSCs into the subcapsular space of the kidney, followed two weeks later by retrieval and histologic analysis.ResultsFlow activated cell sorting showed that hAFSCs expressed mesenchymal stem cell (MSC) markers, but no hematopoietic stem cell markers. Induction of myogenic differentiation in the hAFSCs resulted in expression of PAX7 and MYOD at Day 3, and DYSTROPHIN at Day 7. The nanoparticle-labeled hAFSCs could be tracked in vivo with optical imaging for up to 10 days after injection. Four weeks after injection, the mean LPP and CP were significantly increased in the hAFSC-injected group compared with the control group. Nerve regeneration and neuromuscular junction formation of injected hAFSCs in vivo was confirmed with expression of neuronal markers and acetylcholine receptor. Injection of hAFSCs caused no in vivo host CD8 lymphocyte aggregation or tumor formation.ConclusionshAFSCs displayed MSC characteristics and could differentiate into cells of myogenic lineage. Periurethral injection of hAFSCs into an SUI animal model restored the urethral sphincter to apparently normal histology and function, in absence of immunogenicity and tumorigenicity.


Tissue Engineering Part A | 2009

Enhanced ex vivo expansion of human adipose tissue-derived mesenchymal stromal cells by fibroblast growth factor-2 and dexamethasone.

Sun-Young Lee; Jiwon Lim; Gilson Khang; Youngsook Son; Phil-Hoon Choung; Shin-Sung Kang; So Young Chun; Hong-In Shin; Shin-Yoon Kim; Eui Kyun Park

In the present study, we investigated the ex vivo expansion of human adipose tissue-derived mesenchymal stromal cells (ATSCs) to identify factors that promoted efficient expansion while preserving stem cell potential. We examined several growth factors and steroids, and found that the combination of a low concentration of fibroblast growth factor-2 (FGF-2) (1 ng/mL) and dexamethasone (DEX) or betamethasone (BET) enhanced the proliferation of ATSCs by approximately 30-60% as compared to control. Enhanced proliferation under these conditions was confirmed using ATSCs isolated from three independent donors. ATSCs that were expanded in the presence of FGF-2 and DEX for 5 days were capable of differentiating into either osteoblastic or adipogenic cells, and the cells were positive for the mesenchymal stem cell markers such as CD29, CD44, CD90, CD105, and CD146, suggesting that the stem cell potential of the ATSCs was preserved. Analysis of signaling pathway revealed that tyrosine phosphorylation of Src kinase was dramatically increased in response to FGF-2 and DEX, suggesting the involvement of Src-dependent pathways in the stimulatory mechanism of proliferation of ATSCs by FGF-2 and DEX. Moreover, Src family kinase inhibitors (SU6656 and Src kinase inhibitor I) substantially reduced the FGF-2 and DEX-induced proliferation of ATSCs. SU6656 also inhibited the osteogenic and adipogenic differentiation of ATSCs. The results of the current study demonstrate that FGF-2 in combination with DEX stimulates the proliferation and osteoblastic and adipogenic differentiation of ATSCs through a Src-dependent mechanism, and that FGF-2 and DEX promote the efficient ex vivo expansion of ATSCs.


Journal of Tissue Engineering and Regenerative Medicine | 2015

Pdx1 and controlled culture conditions induced differentiation of human amniotic fluid-derived stem cells to insulin-producing clusters

So Young Chun; David L. Mack; Emily C. Moorefield; Se Heang Oh; Tae Gyun Kwon; Mark J. Pettenati; James J. Yoo; Paolo De Coppi; Anthony Atala; Shay Soker

This study investigated the differentiation of human amniotic fluid‐derived stem cells (hAFSCs) into insulin‐producing clusters in vitro. Adenovirally‐delivered mouse Pdx1 (Ad‐Pdx1) induced human Pdx1 expression in hAFSCs and enhanced the coordinated expression of downstream β‐cell markers. When Ad‐Pdx1‐transduced hAFSCs were sequentially treated with activin A, bFGF and nicotinamide and the culture plate surface coated with poly‐l‐ornithine, the expression of islet‐associated human mRNAs for Pdx1, Pax6, Ngn3 and insulin was increased. C‐peptide ELISA confirmed that Ad‐Pdx1‐transduced hAFSCs processed and secreted insulin in a manner consistent with that pathway in pancreatic β‐cells. To sustain the β‐cell‐like phenotype and investigate the effect of three‐dimensional (3D) conformation on the differentiation of hAFSCs, Pdx1‐transduced cells were encapsulated in alginate and cultured long‐term under serum‐free conditions. Over 2 weeks, partially differentiated hAFSC clusters increased in size and increased insulin secretion. Taken together, these data demonstrate that ectopic Pdx1 expression initiates pancreatic differentiation in hAFSCs and that a β‐cell‐like phenotype can be augmented by culture conditions that mimic the stromal components and 3D geometry associated with pancreatic islets. Copyright


Journal of Korean Medical Science | 2016

Differentiation of Human Dental Pulp Stem Cells into Dopaminergic Neuron-like Cells in Vitro

So Young Chun; Shay Soker; Yu-Jin Jang; Tae Gyun Kwon; Eun Sang Yoo

We investigated the potential of human dental pulp stem cells (hDPSCs) to differentiate into dopaminergic neurons in vitro as an autologous stem cell source for Parkinson’s disease treatment. The hDPSCs were expanded in knockout-embryonic stem cell (KO-ES) medium containing leukemia inhibitory factor (LIF) on gelatin-coated plates for 3–4 days. Then, the medium was replaced with KO-ES medium without LIF to allow the formation of the neurosphere for 4 days. The neurosphere was transferred into ITS medium, containing ITS (human insulin-transferrin-sodium) and fibronectin, to select for Nestin-positive cells for 6–8 days. The cells were then cultured in N-2 medium containing basic fibroblast growth factor (FGF), FGF-8b, sonic hedgehog-N, and ascorbic acid on poly-l-ornithine/fibronectin-coated plates to expand the Nestin-positive cells for up to 2 weeks. Finally, the cells were transferred into N-2/ascorbic acid medium to allow for their differentiation into dopaminergic neurons for 10–15 days. The differentiation stages were confirmed by morphological, immunocytochemical, flow cytometric, real-time PCR, and ELISA analyses. The expressions of mesenchymal stem cell markers were observed at the early stages. The expressions of early neuronal markers were maintained throughout the differentiation stages. The mature neural markers showed increased expression from stage 3 onwards. The percentage of cells positive for tyrosine hydroxylase was 14.49%, and the amount was 0.526 ± 0.033 ng/mL at the last stage. hDPSCs can differentiate into dopaminergic neural cells under experimental cell differentiation conditions, showing potential as an autologous cell source for the treatment of Parkinson’s disease.


ACS Applied Materials & Interfaces | 2016

Biomimetic Porous PLGA Scaffolds Incorporating Decellularized Extracellular Matrix for Kidney Tissue Regeneration

Eugene Lih; Ki Wan Park; So Young Chun; Hyuncheol Kim; Tae Gyun Kwon; Dong Keun Han

Chronic kidney disease is now recognized as a major health problem, but current therapies including dialysis and renal replacement have many limitations. Consequently, biodegradable scaffolds to help repairing injured tissue are emerging as a promising approach in the field of kidney tissue engineering. Poly(lactic-co-glycolic acid) (PLGA) is a useful biomedical material, but its insufficient biocompatibility caused a reduction in cell behavior and function. In this work, we developed the kidney-derived extracellular matrix (ECM) incorporated PLGA scaffolds as a cell supporting material for kidney tissue regeneration. Biomimetic PLGA scaffolds (PLGA/ECM) with different ECM concentrations were prepared by an ice particle leaching method, and their physicochemical and mechanical properties were characterized through various analyses. The proliferation of renal cortical epithelial cells on the PLGA/ECM scaffolds increased with an increase in ECM concentrations (0.2, 1, 5, and 10%) in scaffolds. The PLGA scaffold containing 10% of ECM has been shown to be an effective matrix for the repair and reconstitution of glomerulus and blood vessels in partially nephrectomized mice in vivo, compared with only PLGA control. These results suggest that not only can the tissue-engineering techniques be an effective alternative method for treatment of kidney diseases, but also the ECM incorporated PLGA scaffolds could be promising materials for biomedical applications including tissue engineered scaffolds and biodegradable implants.


Journal of Biomedical Materials Research Part A | 2012

Effect of biological/physical stimulation on guided bone regeneration through asymmetrically porous membrane.

Tae Ho Kim; Se Heang Oh; Seung Yeon Na; So Young Chun; Jin Ho Lee

Asymmetrically porous polycaprolactone (PCL)/Pluronic F127 guided bone regeneration (GBR) membranes were fabricated. The top surface of the membrane had nanosize pores (∼10 nm) which can effectively prevent invasion by fibrous connective tissue but permeate nutrients, whereas the bottom surface had microsize pores (∼200 μm) which can enhance the adhesiveness with bone tissue. Ultrasound was applied to a bone morphogenetic protein (BMP-2)-immobilized PCL/F127 GBR membrane to investigate the feasibility of using dual biological (BMP-2) and physical (ultrasound) stimulation for enhancing bone regeneration through the membrane. In an animal study using SD rats (cranial defect model), the bone regeneration behavior that occurred when using BMP-2-loaded GBR membranes with ultrasound treatment (GBR/BMP-2/US) was much faster than when the same GBR membrane was used without the ultrasound treatment (GBR/BMP-2), as well as when GBR membranes were used without stimulations (GBR). The enhanced bone regeneration of the GBR/BMP-2/US group can be interpreted as resulting from the synergistic or additive effect of the asymmetrically porous PCL/F127 membrane with unique properties (selective permeability, hydrophilicity, and osteoconductivity) and the stimulatory effects of BMP-2 and ultrasound (osteoinductivity). The asymmetrically porous GBR membrane with dual BMP-2 and ultrasound stimulation may be promising for the clinical treatment of delayed and insufficient bone healing.


Journal of Korean Medical Science | 2012

Human Amniotic Fluid Stem Cell-derived Muscle Progenitor Cell Therapy for Stress Urinary Incontinence

So Young Chun; Deok Hyun Cho; Seon Yeong Chae; Kyung Hee Choi; Hyun Ju Lim; Ghil Suk Yoon; Bum Soo Kim; Bup Wan Kim; James J. Yoo; Tae Gyun Kwon

The most promising treatment for stress urinary incontinence can be a cell therapy. We suggest human amniotic fluid stem cells (hAFSCs) as an alternative cell source. We established the optimum in vitro protocol for the differentiation from hAFSCs into muscle progenitors. These progenitors were transplanted into the injured urethral sphincter and their therapeutic effect was analyzed. For the development of an efficient differentiation system in vitro, we examined a commercial medium, co-culture and conditioned medium (CM) systems. After being treated with CM, hAFSCs were effectively developed into a muscle lineage. The progenitors were integrated into the host urethral sphincter and the host cell differentiation was stimulated in vivo. Urodynamic analysis showed significant increase of leak point pressure and closing pressure. Immunohistochemistry revealed the regeneration of circular muscle mass with normal appearance. Molecular analysis observed the expression of a larger number of target markers. In the immunogenicity analysis, the progenitor group had a scant CD8 lymphocyte. In tumorigenicity, the progenitors showed no teratoma formation. These results suggest that hAFSCs can effectively be differentiated into muscle progenitors in CM and that the hAFSC-derived muscle progenitors are an accessible cell source for the regeneration of injured urethral sphincter.


BJUI | 2014

Combined injection of three different lineages of early-differentiating human amniotic fluid-derived cells restores urethral sphincter function in urinary incontinence

So Young Chun; Joon Beom Kwon; Seon Yeong Chae; Jong Kil Lee; Jae-sung Bae; Bum Soo Kim; Hyun Tae Kim; Eun Sang Yoo; Jeong Ok Lim; James J. Yoo; Wun-Jae Kim; Bup Wan Kim; Tae Gyun Kwon

To investigate whether a triple combination of early‐differentiated cells derived from human amniotic fluid stem cells (hAFSCs) would show synergistic effects in urethral sphincter regeneration.


Journal of Biomedical Materials Research Part A | 2015

Development of a porcine renal extracellular matrix scaffold as a platform for kidney regeneration

Seock Hwan Choi; So Young Chun; Seon Yeong Chae; Jin Rae Kim; Se Heang Oh; Sung Kwang Chung; Jin Ho Lee; Phil Hyun Song; Gyu-Seog Choi; Tae-Hwan Kim; Tae Gyun Kwon

Acellular scaffolds, possessing an intact three-dimensional extracellular matrix (ECM) architecture and biochemical components, are promising for regeneration of complex organs, such as the kidney. We have successfully developed a porcine renal acellular scaffold and analyzed its physical/biochemical characteristics, biocompatibility, and kidney reconstructive potential. Segmented porcine kidney cortexes were treated with either 1% (v/v) Triton X-100 (Triton) or sodium dodecyl sulfate (SDS). Scanning electron microscopy showed both treatments preserved native tissue architecture, including porosity and composition. Swelling behavior was higher in the Triton-treated compared with the SDS-treated scaffold. Maximum compressive strength was lower in the Triton-treated compared with the SDS-treated scaffold. Attenuated total reflective-infrared spectroscopy showed the presence of amide II (-NH) in both scaffolds. Furthermore, richer ECM protein and growth factor contents were observed in the Triton-treated compared with SDS-treated scaffold. Primary human kidney cell adherence, viability, and proliferation were enhanced on the Triton-treated scaffold compared with SDS-treated scaffold. Following murine in vivo implantation, tumorigenecity was absent for both scaffolds after 8 weeks and in the Triton-treated scaffold only, glomeruli-like structure formation and neovascularity were observed. We identified 1% Triton X-100 as a more suitable decellularizing agent for porcine renal ECM scaffolds prior to kidney regeneration.

Collaboration


Dive into the So Young Chun's collaboration.

Top Co-Authors

Avatar

Tae Gyun Kwon

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Bum Soo Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Eun Sang Yoo

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Hyun Tae Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yun-Sok Ha

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Bup Wan Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tae-Hwan Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge