Network


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

Hotspot


Dive into the research topics where Jong Wook Chang is active.

Publication


Featured researches published by Jong Wook Chang.


International Journal of Molecular Sciences | 2013

Comparative Analysis of Human Mesenchymal Stem Cells from Bone Marrow, Adipose Tissue, and Umbilical Cord Blood as Sources of Cell Therapy

Hye Jin Jin; Yun Kyung Bae; Mi-Yeon Kim; Soon-Jae Kwon; Hong Bae Jeon; Soo Jin Choi; Seong Who Kim; Yoon Sun Yang; Wonil Oh; Jong Wook Chang

Various source-derived mesenchymal stem cells (MSCs) have been considered for cell therapeutics in incurable diseases. To characterize MSCs from different sources, we compared human bone marrow (BM), adipose tissue (AT), and umbilical cord blood-derived MSCs (UCB-MSCs) for surface antigen expression, differentiation ability, proliferation capacity, clonality, tolerance for aging, and paracrine activity. Although MSCs from different tissues have similar levels of surface antigen expression, immunosuppressive activity, and differentiation ability, UCB-MSCs had the highest rate of cell proliferation and clonality, and significantly lower expression of p53, p21, and p16, well known markers of senescence. Since paracrine action is the main action of MSCs, we examined the anti-inflammatory activity of each MSC under lipopolysaccharide (LPS)-induced inflammation. Co-culture of UCB-MSCs with LPS-treated rat alveolar macrophage, reduced expression of inflammatory cytokines including interleukin-1α (IL-1α), IL-6, and IL-8 via angiopoietin-1 (Ang-1). Using recombinant Ang-1 as potential soluble paracrine factor or its small interference RNA (siRNA), we found that Ang-1 secretion was responsible for this beneficial effect in part by preventing inflammation. Our results demonstrate that primitive UCB-MSCs have biological advantages in comparison to adult sources, making UCB-MSCs a useful model for clinical applications of cell therapy.


Neurobiology of Aging | 2012

Human umbilical cord blood-derived mesenchymal stem cells improve neuropathology and cognitive impairment in an Alzheimer's disease mouse model through modulation of neuroinflammation

Hyun Ju Lee; Jong Kil Lee; Hyun Su Lee; Janet E. Carter; Jong Wook Chang; Wonil Oh; Yoon Sun Yang; Jun-Gyo Suh; Byoung-Hee Lee; Hee Kyung Jin; Jae-sung Bae

Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSC) have a potential therapeutic role in the treatment of neurological disorders, but their current clinical usage and mechanism of action has yet to be ascertained in Alzheimers disease (AD). Here we report that hUCB-MSC transplantation into amyloid precursor protein (APP) and presenilin1 (PS1) double-transgenic mice significantly improved spatial learning and memory decline. Furthermore, amyloid-β peptide (Aβ) deposition, β-secretase 1 (BACE-1) levels, and tau hyperphosphorylation were dramatically reduced in hUCB-MSC transplanted APP/PS1 mice. Interestingly, these effects were associated with reversal of disease-associated microglial neuroinflammation, as evidenced by decreased microglia-induced proinflammatory cytokines, elevated alternatively activated microglia, and increased anti-inflammatory cytokines. These findings lead us to suggest that hUCB-MSC produced their sustained neuroprotective effect by inducing a feed-forward loop involving alternative activation of microglial neuroinflammation, thereby ameliorating disease pathophysiology and reversing the cognitive decline associated with Aβ deposition in AD mice.


Stem Cells | 2010

Irradiation Enhances the Tumor Tropism and Therapeutic Potential of Tumor Necrosis Factor‐Related Apoptosis‐Inducing Ligand‐Secreting Human Umbilical Cord Blood‐Derived Mesenchymal Stem Cells in Glioma Therapy

Seong Muk Kim; Ji Hyeon Oh; Soon A Park; Chung Heon Ryu; Jung Yeon Lim; Dal-Soo Kim; Jong Wook Chang; Wonil Oh; Sin-Soo Jeun

Irradiation is a standard therapy for gliomas and many other cancers. Tumor necrosis factor‐related apoptosis‐inducing ligand (TRAIL) is one of the most promising candidates for cancer gene therapy. Here, we show that tumor irradiation enhances the tumor tropism of human umbilical cord blood‐derived mesenchymal stem cells (UCB‐MSCs) and the therapeutic effect of TRAIL delivered by UCB‐MSCs. The sequential treatment with irradiation followed by TRAIL‐secreting UCB‐MSCs (MSC‐TRAIL) synergistically enhanced apoptosis in either TRAIL‐sensitive or TRAIL‐resistant glioma cells by upregulating the death receptor 5 and by inducing caspase activation. Migration assays showed greater MSC migration toward irradiated glioma cells and the tumor site in glioma‐bearing mice compared with unirradiated tumors. Irradiated glioma cells had increased expression of interleukin‐8 (IL‐8), which leads to the upregulation of the IL‐8 receptor on MSCs. This upregulation, which is involved in the migratory capacity of UCB‐MSCs, was confirmed by siRNA inhibition and an antibody‐neutralizing assay. In vivo survival experiments in orthotopic xenografted mice showed that MSC‐based TRAIL gene delivery to irradiated tumors had greater therapeutic efficacy than a single treatment. These results suggest that clinically relevant tumor irradiation increases the therapeutic efficacy of MSC‐TRAIL by increasing tropism of MSCs and TRAIL‐induced apoptosis, which may be a more useful strategy for cancer gene therapy. STEM CELLS 2010;28:2217–2228


Stem Cell Research & Therapy | 2011

Therapeutic effects of human umbilical cord blood-derived mesenchymal stem cells after intrathecal administration by lumbar puncture in a rat model of cerebral ischemia.

Jung Yeon Lim; Chang Hyun Jeong; Jin Ae Jun; Seong Muk Kim; Chung Heon Ryu; Yun Hou; Wonil Oh; Jong Wook Chang; Sin-Soo Jeun

IntroductionStem cell transplantation is a promising therapeutic strategy for the treatment of stroke. Mesenchymal stem cells (MSCs) are a potential cell source for clinical application because they can be easily obtained and cultivated with a high proliferative capacity. The safety and efficacy of cell therapy depends on the mode of cell administration. To determine the therapeutic potential of intrathecal administration of MSCs by lumbar puncture (LP), we administrated human umbilical cord blood-derived MSCs (hUCB-MSCs) intrathecally into the lumbar spinal cord or intravenously into the tail vein in a rat model of stroke, and then investigated whether hUCB-MSCs could enter the brain, survive, and improve post-stroke neurological functional recovery.MethodshUCB-MSCs (1.0 × 106) were administrated three days after stroke induced by occlusion of the middle cerebral artery. The presence of hUCB-MSCs and their survival and differentiation in the brain tissue of the rats was examined by immunohistochemistry. Recovery of coordination of movement after administration of hUCB-MSCs was examined using a Rotarod test and adhesive-removal test on the 7th, 14th, 21st, and 28th days after ischemia. The volume of ischemic lesions seven days after the experimental procedure was evaluated using 2-3-5-triphenyltetrazolium (TTC) staining.ResultsRats receiving hUCB-MSCs intrathecally by LP had a significantly higher number of migrated cells within the ischemic area when compared with animals receiving cells intravenously. In addition, many of the cells administered intrathecally survived and a subset of them expressed mature neural-lineage markers, including the mature neuron marker NeuN and glial fibrillary acidic protein, typical of astrocytes. Animals that received hUCB-MSCs had significantly improved motor function and reduced ischemic damage when compared with untreated control animals. Regardless of the administration route, the group treated with 1 × 106 hUCB-MSCs showed better neurological recovery, without significant differences between the two treatment groups. Importantly, intrathecal administration of 5 × 105 hUCB-MSCs significantly reduced ischemic damage, but not in the intravenously treated group. Furthermore, the cells administered intrathecally survived and migrated into the ischemic area more extensively, and differentiated significantly into neurons and astrocytes.ConclusionsTogether, these results indicate that intrathecal administration of MSCs by LP may be useful and feasible for MSCs treatment of brain injuries, such as stroke, or neurodegenerative disorders.


Stem Cells and Development | 2009

Overexpression of CXC chemokine receptors is required for the superior glioma-tracking property of umbilical cord blood-derived mesenchymal stem cells.

Dal-Soo Kim; Ji Hyun Kim; Jae Kwon Lee; Soo Jin Choi; Jae-Sung Kim; Sin-Soo Jeun; Wonil Oh; Yoon Sun Yang; Jong Wook Chang

Our observations indicate that umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) have a strong migration capacity toward the human glioma cell line, U-87 MG, LN18, U138, and U251, when compared to several other cancer cell lines. In order to identify soluble factors that function to attract UCB-MSCs, we used cytokine antibody arrays to screen changed cytokines in conditioned media from U-87 MG cells. Among these, interleukin-8 (IL-8) and growth-related oncogene (GRO-alpha) enhanced UCB-MSC migration. Furthermore, antibodies treatment against the IL-8 receptors reduced these migration events and overexpression of IL-8 in cells with lower level of IL-8 such as A549 could induce UCB-MSC migration. Since we found that the capacity of UCB-MSC migration is much higher than that of bone marrow-derived MSCs (BM-MSCs) toward either U-87 MG cells or recombinant IL-8, we compared the levels of the IL-8 receptor, CXC chemokine receptor 1 (CXCR1) and CXCR2 between two kinds of MSCs by RT-PCR and immunostaining. Expression levels of two receptors were much higher in UCB-MSCs than in BM-MSCs. These data suggest that higher levels of two IL-8 receptors could influence downstream signaling events affecting superior UCB-MSC migration toward the glioma cells.


World Journal of Stem Cells | 2010

Application of human umbilical cord blood-derived mesenchymal stem cells in disease models

J.I. Kim; Hong Bae Jeon; Yoon Sun Yang; Wonil Oh; Jong Wook Chang

Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) are regarded as an alternative source of bone marrow-derived mesenchymal stem cells because collection of cord blood is less invasive than that of bone marrow. hUCB-MSCs have recently been studied for evaluation of their potential as a source of cell therapy. In this review, the general characteristics of hUCB-MSCs and their therapeutic effects on various diseases in vitro and in vivo will be discussed.


Biochemical and Biophysical Research Communications | 2011

CXC chemokine receptor 1 enhances the ability of human umbilical cord blood-derived mesenchymal stem cells to migrate toward gliomas

Seong Muk Kim; Dal-Soo Kim; Chang Hyun Jeong; Dong Hyun Kim; Ji Hyun Kim; Hong Bae Jeon; Soon-Jae Kwon; Sin-Soo Jeun; Yoon Sun Yang; Wonil Oh; Jong Wook Chang

In this study, we showed that knocking-down interleukin-8 (IL-8) in glioma cells, or its receptor, CXC chemokine receptor 1 (CXCR1) in hUCB-MSCs reduced hUCB-MSC migration toward glioma cells in a Transwell chamber. In contrast, CXCR1-transfected hUCB-MSCs (CXCR1-MSCs) showed a superior capacity to migrate toward glioma cells in a Transwell chamber compared to primary hUCB-MSCs. Furthermore, these transfected cells also demonstrated the same ability to migrate toward tumors in mice bearing intracranial human gliomas as shown by histological and in vivo imaging analysis. Our findings indicate that overexpression of CXCR1 could be a useful tool for MSC-based gene therapy to achieve a sufficient quantity of therapeutic MSCs that are localized within tumors.


Stem Cells | 2013

Thrombospondin-2 secreted by human umbilical cord blood-derived mesenchymal stem cells promotes chondrogenic differentiation

Sang Young Jeong; Dong Hyun Kim; Jueun Ha; Hye Jin Jin; Soon-Jae Kwon; Jong Wook Chang; Soo Jin Choi; Wonil Oh; Yoon Sun Yang; Gonhyung Kim; Jae-Sung Kim; Jung-Ro Yoon; Dong Hyung Cho; Hong Bae Jeon

Increasing evidence indicates that the secretome of mesenchymal stem cells (MSCs) has therapeutic potential for the treatment of various diseases, including cartilage disorders. However, the paracrine mechanisms underlying cartilage repair by MSCs are poorly understood. Here, we show that human umbilical cord blood‐derived MSCs (hUCB‐MSCs) promoted differentiation of chondroprogenitor cells by paracrine action. This paracrine effect of hUCB‐MSCs on chondroprogenitor cells was increased by treatment with synovial fluid (SF) obtained from osteoarthritis (OA) patients but was decreased by SF of fracture patients, compared to that of an untreated group. To identify paracrine factors underlying the chondrogenic effect of hUCB‐MSCs, the secretomes of hUCB‐MSCs stimulated by OA SF or fracture SF were analyzed using a biotin label‐based antibody array. Among the proteins increased in response to these two kinds of SF, thrombospondin‐2 (TSP‐2) was specifically increased in only OA SF‐treated hUCB‐MSCs. In order to determine the role of TSP‐2, exogenous TSP‐2 was added to a micromass culture of chondroprogenitor cells. We found that TSP‐2 had chondrogenic effects on chondroprogenitor cells via PKCα, ERK, p38/MAPK, and Notch signaling pathways. Knockdown of TSP‐2 expression on hUCB‐MSCs using small interfering RNA abolished the chondrogenic effects of hUCB‐MSCs on chondroprogenitor cells. In parallel with in vitro analysis, the cartilage regenerating effect of hUCB‐MSCs and TSP‐2 was also demonstrated using a rabbit full‐thickness osteochondral‐defect model. Our findings suggested that hUCB‐MSCs can stimulate the differentiation of locally presented endogenous chondroprogenitor cells by TSP‐2, which finally leads to cartilage regeneration. Stem Cells 2013;31:2136–2148


BMC Biotechnology | 2010

Microporation is a valuable transfection method for efficient gene delivery into human umbilical cord blood-derived mesenchymal stem cells

Jung Yeon Lim; Sun Hwa Park; Chang Hyun Jeong; Ji Hyeon Oh; Seong Muk Kim; Chung Hun Ryu; Soon A Park; Jae Geun Ahn; Wonil Oh; Sin-Soo Jeun; Jong Wook Chang

BackgroundMesenchymal stem cells (MSCs) are an attractive source of adult stem cells for therapeutic application in clinical study. Genetic modification of MSCs with beneficial genes makes them more effective for therapeutic use. However, it is difficult to transduce genes into MSCs by common transfection methods, especially nonviral methods. In this study, we applied microporation technology as a novel electroporation technique to introduce enhanced green fluorescent protein (EGFP) and brain-derived neurotropfic factor (BDNF) plasmid DNA into human umbilical cord blood-derived MSCs (hUCB-MSCs) with significant efficiency, and investigated the stem cell potentiality of engineered MSCs through their phenotypes, proliferative capacity, ability to differentiate into multiple lineages, and migration ability towards malignant glioma cells.ResultsUsing microporation with EGFP as a reporter gene, hUCB-MSCs were transfected with higher efficiency (83%) and only minimal cell damage than when conventional liposome-based reagent (<20%) or established electroporation methods were used (30-40%). More importantly, microporation did not affect the immunophenotype of hUCB-MSCs, their proliferation activity, ability to differentiate into mesodermal and ectodermal lineages, or migration ability towards cancer cells. In addition, the BDNF gene could be successfully transfected into hUCB-MSCs, and BDNF expression remained fairly constant for the first 2 weeks in vitro and in vivo. Moreover, microporation of BDNF gene into hUCB-MSCs promoted their in vitro differentiation into neural cells.ConclusionTaken together, the present data demonstrates the value of microporation as an efficient means of transfection of MSCs without changing their multiple properties. Gene delivery by microporation may enhance the feasibility of transgenic stem cell therapy.


FEBS Letters | 2010

Galectin-3 secreted by human umbilical cord blood-derived mesenchymal stem cells reduces amyloid-β42 neurotoxicity in vitro

J.I. Kim; Dong Hyun Kim; Dal-Soo Kim; Ji Hyun Kim; Sang Young Jeong; Hong Bae Jeon; Eun Hui Lee; Yoon Sun Yang; Wonil Oh; Jong Wook Chang

In this study, we found that expression and secretion of galectin‐3 (GAL‐3) were upregulated by amyloid‐β42 (Aβ42) exposure in human umbilical cord blood‐derived mesenchymal stem cell (hUCB‐MSC) without cell death. Aβ42‐exposed rat primary cortical neuronal cells co‐treated with recombinant GAL‐3 were protected from neuronal death in a dose‐dependent manner. hUCB‐MSCs were cocultured with Aβ42‐exposed rat primary neuronal cells or the neuroblastoma cell line, SH‐SY5Y in a Transwell chamber. Coculture of hUCB‐MSCs reduced cell death of Aβ42‐exposed neurons and SH‐SY5Y cells. This neuroprotective effect of hUCB‐MSCs was reduced significantly by GAL‐3 siRNA. These data suggested that hUCB‐MSC‐derived GAL‐3 is a survival factor against Aβ42 neurotoxicity.

Collaboration


Dive into the Jong Wook Chang's collaboration.

Top Co-Authors

Avatar

Yoon Sun Yang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Duk L. Na

Samsung Medical Center

View shared research outputs
Top Co-Authors

Avatar

Na Kyung Lee

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

J.I. Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ji Hyun Kim

Ulsan National Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Won Il Oh

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge