Network


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

Hotspot


Dive into the research topics where Jin Soo Oh is active.

Publication


Featured researches published by Jin Soo Oh.


Neuroscience Letters | 2010

Hypoxia-preconditioned adipose tissue-derived mesenchymal stem cell increase the survival and gene expression of engineered neural stem cells in a spinal cord injury model.

Jin Soo Oh; Yoon Ha; Sung Su An; Momin Khan; William A. Pennant; Hyo Jin Kim; Do Heum Yoon; Minhyung Lee; Keung Nyun Kim

Hypoxic preconditioning (HP) is a novel strategy to make stem cells resistant to the ischemic environment they encounter after transplantation into injured tissue; this strategy improves survival of both the transplanted cells and the host cells at the injury site. Using both in vitro and in vivo injury models, we confirmed that HP-treated adipose tissue-derived mesenchymal stem cells (HP-AT-MSCs) increased cell survival and enhanced the expression of marker genes in DsRed-engineered neural stem cells (NSCs-DsRed). Similar to untreated AT-MSCs, HP-AT-MSCs had normal morphology and were positive for the cell surface markers CD90, CD105, and CD29, but not CD31. In three in vitro ischemic-mimicking injury models, HP-AT-MSCs significantly increased both the viability of NSCs-DsRed and the expression of DsRed and clearly reduced the number of annexin-V-positive apoptotic NSCs-DsRed and the expression of the apoptotic factor Bax. Consistent with the in vitro assay, co-transplantation of NSCs-DsRed with HP-AT-MSCs significantly improved the survival of the NSCs-DsRed, resulting in an increased expression of the DsRed reporter gene at the transplantation site in a rat spinal cord injury (SCI) model. These findings suggest that the co-transplantation of HP-AT-MSCs with engineered NSCs can improve both the cell survival and the gene expression of the engineered NSCs, indicating that this novel strategy can be used to augment the therapeutic efficacy of combined stem cell and gene therapies for SCI.


Cell Transplantation | 2011

Cotransplantation of mouse neural stem cells (mNSCs) with adipose tissue-derived mesenchymal stem cells improves mNSC survival in a rat spinal cord injury model.

Jin Soo Oh; Keung Nyun Kim; Sung Su An; William A. Pennant; Hyo Jin Kim; So-Jung Gwak; Do Heum Yoon; Mi Hyun Lim; Byung Hyune Choi; Yoon Ha

The low survival rate of graft stem cells after transplantation into recipient tissue is a major obstacle for successful stem cell therapy. After transplantation into the site of spinal cord injury, the stem cells face not only hypoxia due to low oxygen conditions, but also a lack of nutrients caused by damaged tissues and poor vascular supply. To improve the survival of therapeutic stem cells after grafting into the injured spinal cord, we examined the effects of cotransplanting mouse neural stem cells (mNSCs) and adipose tissue-derived mesenchymal stem cells (AT-MSCs) on mNSC viability. The viability of mNSCs in coculture with AT-MSCs was significantly increased compared to mNSCs alone in an in vitro injury model using serum deprivation (SD), hydrogen peroxide (H2O2), and combined (SD + H2O2) injury mimicking the ischemic environment of the injured spinal cord. We demonstrated that AT-MSCs inhibited the apoptosis of mNSCs in SD, H2O2, and combined injury models. Consistent with these in vitro results, mNSCs transplanted into rat spinal cords with AT-MSCs showed better survival rates than mNSCs transplanted alone. These findings suggest that cotransplantation of mNSCs with AT-MSCs may be a more effective transplantation protocol to improve the survival of cells transplanted into the injured spinal cord.


Neuroreport | 2012

Transplantation of an adipose stem cell cluster in a spinal cord injury.

Jin Soo Oh; In Su Park; Keung Nyun Kim; Do Heum Yoon; Sang-Heon Kim; Yoon Ha

We investigated whether transplantation of a three-dimensional cell mass of adipose-derived stem cells (3DCM-ASCs) improved hind limb functional recovery by the stimulation of angiogenesis and neurogenesis in a spinal cord injury. In in-vitro experiments, we confirmed that 3DCM-ASCs differentiated into CD31-positive endothelial cells. To evaluate the therapeutic effect of 3DCM-ASCs in vivo, PBS, human adipose tissue-derived stem cells, and 3DCM-ASCs were transplanted into a spinal cord injury model. The 3DCM-ASCs transplanted into the injured spinal cord differentiated into CD31-positive endothelial cells and remained differentiated. Transplantation of 3DCM-ASCs into the injured spinal cord significantly elevated the density of vascular formations through angiogenic factors released by the 3DCM-ASCs at the lesion site, and enhanced axonal outgrowth at the lesion site. Consistent with these results, the transplantation of 3DCM-ASCs significantly improved functional recovery compared with both ASC transplantation and PBS treatment. These findings suggest that transplantation of 3DCM-ASCs may be an effective stem cell therapy for the treatment of spinal cord injuries and neural ischemia.


Journal of Gene Medicine | 2010

Controlled nonviral gene delivery and expression using stable neural stem cell line transfected with a hypoxia-inducible gene expression system.

Meng-Lu Liu; Jin Soo Oh; Sung Su An; William A. Pennant; Hyo Jin Kim; So-Jung Gwak; Do Heum Yoon; Keung Nyun Kim; Minhyung Lee; Yoon Ha

Nonviral ex vivo local gene therapy systems consisting of regulated gene expression vectors and cellular delivery platforms represent a novel strategy for tissue repair and regeneration. We introduced a hypoxia‐regulated plasmid‐based system into mouse neural stem cells (NSCs) as an efficient gene expression and delivery platform for rapid, robust and persistent hypoxic/ischemic‐regulated gene expression in the spinal cord.


Spine | 2011

Neural stem cells modified by a hypoxia-inducible VEGF gene expression system improve cell viability under hypoxic conditions and spinal cord injury.

Hong Lian Jin; William A. Pennant; Min Hyung Lee; Sung Su; Hyun Ah Kim; Meng Lu Liu; Jin Soo Oh; Joon Cho; Keung Nyun Kim; Do Heum Yoon; Yoon Ha

Study Design. An in vitro neural hypoxia model and rat spinal cord injury (SCI) model were used to assess the regulation of therapeutic vascular endothelial growth factor (VEGF) gene expression in mouse neural stem cells (mNSCs) by the EPO (erythropoietin) enhancer or RTP801 promoter. Objective. To increase VEGF gene expression in mNSCs under hypoxic conditions in SCI lesions but avoid unwanted overexpression of VEGF in normal sites, we developed a hypoxia-inducible gene expression system consisting of the EPO enhancer and RTP801 promoter fused to VEGF or the luciferase gene, then transfected into mNSCs. Summary of Background Data. On the basis of the ischemic response in the injured area, poor cell survival at the transplantation site is a consistent problem with NSC transplantation after SCI. Although VEGF directly protects neurons and enhances neurite outgrowth, uncontrolled overexpression of VEGF in uninjured tissue may cause serious adverse effects. To effectively improve NSC survival in ischemic sites after transplantation, we evaluated mNSCs modified by a hypoxia-inducible VEGF gene expression system in an SCI model. Methods. Hypoxia-inducible luciferase or VEGF plasmids were constructed using the EPO enhancer or RTP801 promoter. The effect of these systems on targeted gene expression and cell viability was evaluated in mNSCs in both hypoxic in vitro injury and a rat SCI model in vivo. Results. The gene expression system containing the EPO enhancer or RTP801 promoter significantly increased the expression of the luciferase reporter gene and therapeutic VEGF gene under hypoxic conditions. The Epo-SV-VEGF plasmid transfection group had significantly fewer apoptotic cells in vitro. This system also augmented cell viability in the in vivo SCI model. Conclusion. These results strongly suggest the potential utility of mNSCs modified by a hypoxia-inducible VEGF gene expression system in the development of effective stem cell transplantation protocols in SCI.


Journal of Biomaterials Science-polymer Edition | 2012

Chitosan/TPP-Hyaluronic Acid Nanoparticles: A New Vehicle for Gene Delivery to the Spinal Cord

So-Jung Gwak; Jong Kwon Jung; Sung Su An; Hyo Jin Kim; Jin Soo Oh; William A. Pennant; Hye Yeong Lee; Min Ho Kong; Keung Nyun Kim; Do Heum Yoon; Yoon Ha

Abstract Gene delivery offers therapeutic promise for the treatment of neurological diseases and spinal cord injury. Several studies have offered viral vectors as vehicles to deliver therapeutic agents, yet their toxicity and immunogenicity, along with the cost of their large-scale formulation, limits their clinical use. As such, non-viral vectors are attractive in that they offer improved safety profiles compared to viruses. Poly(ethylene imine) (PEI) is one of the most extensively studied non-viral vectors, but its clinical value is limited y its cytotoxicity. Recently, chitosan/DNA complex nanoparticles have een considered as a vector for gene delivery. Here, we demonstrate that DNA nanoparticles made of hyaluronic acid (HA) and chitosan have low cytotoxicity and induce high transgene expression in neural stem cells and organotypic spinal cord slice tissue. Chitosan-TPP/HA nanoparticles were significantly less cytotoxic than PEI at various concentrations. Additionally, chitosan-TPP/HA nanoparticles with pDNA induced higher transgene expression in vitro for a longer duration than PEI in neural stem cells. These results suggest chitosan-TPP/HA nanoparticles may have the potential to serve as an option for gene delivery to the spinal cord.


Biomedical Materials | 2015

Membrane-reinforced three-dimensional electrospun silk fibroin scaffolds for bone tissue engineering.

Sung Yeun Yang; Tae Heon Hwang; Lihua Che; Jin Soo Oh; Yoon Ha; WonHyoung Ryu

Electrospun silk fibroin (SF) scaffolds have drawn much attention because of their resemblance to natural tissue architecture such as extracellular matrix, and the biocompatibility of SF as a candidate material to replace collagen. However, electrospun scaffolds lack the physical integrity of bone tissue scaffolds, which require resistance to mechanical loadings. In this work, we propose membrane-reinforced electrospun SF scaffolds by a serial process of electrospinning and freeze-drying of SF solutions in two different solvents: formic acid and water, respectively. After wet electrospinning followed by replacement of methanol with water, SF nanofibers dispersed in water were mixed with aqueous SF solution. Freeze-drying of the mixed solution resulted in 3D membrane-connected SF nanofibrous scaffolds (SF scaffolds) with a thickness of a few centimeters. We demonstrated that the SF concentration of aqueous SF solution controlled the degree of membrane reinforcement between nanofibers. It was also shown that both increase in degree of membrane reinforcement and inclusion of hydroxyapatite (HAP) nanoparticles resulted in higher resistance to compressive loadings of the SF scaffolds. Culture of human osteoblasts on collagen, SF, and SF-HAP scaffolds showed that both SF and SF-HAP scaffolds had biocompatibility and cell proliferation superior to that of the collagen scaffolds. SF-HAP scaffolds with and without BMP-2 were used for in vivo studies for 4 and 8 weeks, and they showed enhanced bone tissue formation in rat calvarial defect models.


Neuroreport | 2012

Hypoxia-specific VEGF-expressing Neural Stem Cells in Spinal Cord Injury Model

Jin Soo Oh; Sung Su An; So-Jung Gwak; William A. Pennant; Keung Nyun Kim; Do Heum Yoon; Yoon Ha

We established three stable neural stem cell (NSC) lines to explore the possibility of using hypoxia-specific vascular endothelial growth factor (VEGF) expressing NSC lines (EpoSV-VEGF NSCs) to treat spinal cord injury. The application of EpoSV-VEGF NSCs into the injured spinal cord after clip compression injury not only showed therapeutic effects such as extended survival and angiogenesis, but also displayed its safety profile as it did not cause unwanted cell proliferation or angiogenesis in normal spinal cord tissue, as EpoSV-VEGF NSCs consistently showed hypoxia-specific VEGF expression patterns. This suggests that our EpoSV-VEGF NSCs are both safe and therapeutically efficacious for the treatment of spinal cord injury. Furthermore, this hypoxia-inducible gene expression system may represent a safe tool suitable for gene therapy.


Neuroreport | 2011

Hypoxia-induced expression of VEGF in the organotypic spinal cord slice culture.

Sung Su An; William A. Pennant; Yoon Ha; Jin Soo Oh; Hyo Jin Kim; So-Jung Gwak; Do Heum Yoon; Keung Nyun Kim

We used the erythropoietin enhancer and Simian virus-40 promoter to create a hypoxia-inducible gene expression system to investigate the effect of vascular endothelial growth factor (VEGF) gene therapy on neuroprotection and neurogenesis in organotypic spinal cord slice culture. The organotypic spinal cord slice culture transfected with pEpo-SV-VEGF expressed the highest amount of VEGF under hypoxic conditions and showed decreased apoptosis and increased proliferation, and evidence of neurogenesis. Our results show that the hypoxia-induced VEGF expression in an organotypic spinal cord slice culture may lead to an optimal treatment for spinal cord injury.


Neuroreport | 2010

Effect of primate bone marrow stromal cells on survival and neurite outgrowth

Keung Nyun Kim; James D. Guest; Jin Soo Oh; William A. Pennant; Do Heum Yoon; Yoon Ha

We tested whether bone marrow stromal cells (BMSCs) could enhance the survival and neurite growth of dorsal root ganglia (DRG) through substrate effects or secreted factors. Our results showed that in DRG with BMSCs and BMSC-conditioned media cultures compared with DRG-fibroblast cultures, there was a significant increase in the number and length of, area covered by, and number of cells with definite neurites. In cytokine assays with conditioned media, vascular endothelial growth factor, granulocyte macrophage colony-stimulating factor, and IL-6 secreted by BMSCs may contribute to observed neurotrophic effects. These findings indicate that BMSCs of adult Macaca fascicularis increased neuronal survival and promoted neurite outgrowth of DRG by means of secretory factors.

Collaboration


Dive into the Jin Soo Oh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge