Network


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

Hotspot


Dive into the research topics where Gun-Jae Jeong is active.

Publication


Featured researches published by Gun-Jae Jeong.


International Journal of Nanomedicine | 2014

Delivery of bone morphogenetic protein-2 and substance P using graphene oxide for bone regeneration

Wan-Geun La; Min Jin; Saibom Park; Hee-Hun Yoon; Gun-Jae Jeong; Suk Ho Bhang; Hoyoung Park; Kookheon Char; Byung-Soo Kim

In this study, we demonstrate that graphene oxide (GO) can be used for the delivery of bone morphogenetic protein-2 (BMP-2) and substance P (SP), and that this delivery promotes bone formation on titanium (Ti) implants that are coated with GO. GO coating on Ti substrate enabled a sustained release of BMP-2. BMP-2 delivery using GO-coated Ti exhibited a higher alkaline phosphatase activity in bone-forming cells in vitro compared with bare Ti. SP, which is known to recruit mesenchymal stem cells (MSCs), was co-delivered using Ti or GO-coated Ti to further promote bone formation. SP induced the migration of MSCs in vitro. The dual delivery of BMP-2 and SP using GO-coated Ti showed the greatest new bone formation on Ti implanted in the mouse calvaria compared with other groups. This approach may be useful to improve osteointegration of Ti in dental or orthopedic implants.


Biochemical and Biophysical Research Communications | 2014

Graphene enhances the cardiomyogenic differentiation of human embryonic stem cells

Tae-Jin Lee; Subeom Park; Suk Ho Bhang; Jeong-Kee Yoon; Insu Jo; Gun-Jae Jeong; Byung Hee Hong; Byung-Soo Kim

Graphene has drawn attention as a substrate for stem cell culture and has been reported to stimulate the differentiation of multipotent adult stem cells. Here, we report that graphene enhances the cardiomyogenic differentiation of human embryonic stem cells (hESCs) at least in part, due to nanoroughness of graphene. Large-area graphene on glass coverslips was prepared via the chemical vapor deposition method. The coating of the graphene with vitronectin (VN) was required to ensure high viability of the hESCs cultured on the graphene. hESCs were cultured on either VN-coated glass (glass group) or VN-coated graphene (graphene group) for 21 days. The cells were also cultured on glass coated with Matrigel (Matrigel group), which is a substrate used in conventional, directed cardiomyogenic differentiation systems. The culture of hESCs on graphene promoted the expression of genes involved in the stepwise differentiation into mesodermal and endodermal lineage cells and subsequently cardiomyogenic differentiation compared with the culture on glass or Matrigel. In addition, the culture on graphene enhanced the gene expression of cardiac-specific extracellular matrices. Culture on graphene may provide a new platform for the development of stem cell therapies for ischemic heart diseases by enhancing the cardiomyogenic differentiation of hESCs.


Journal of Power Sources | 2001

Particulate-reinforced Al-based composite material for anode in lithium secondary batteries

Gun-Jae Jeong; Younghyun Kim; H.-J. Sohn; Tak Kang

Particulate-reinforced Al/SiC composite materials are prepared by ball-milling technique to be used as an anode material for lithium secondary battery. The microstructure of the composite powders show that the SiC particles are embedded homogeneously in the Al matrix. This feature is distinctively different from any other active/inactive composite anode materials reported recently. The cycle performance of these composite electrodes is superior to that of an unreinforced aluminium electrode. This improved cycelability may be due to an enhanced mechanical stability of the electrode.


Metals and Materials | 1999

Mathematical modeling on the corrosion of unprotected structure due to stray current resulting from cathodic protection system

Y. S. Kim; Gun-Jae Jeong; Hun-Joon Sohn

Mathematical modeling on the corrosion of unprotected structures due to a stray current resulting from a nearby cathodic protection system was carried out using the boundary element method. The model consists Laplace’s equation with non-linear boundary conditions (Tafel equations) and the iterative technique to determine the mixed potential of the unprotected structure. The model is applied to an unprotected bare structure as well as a coated structure with several defects. The amount and the location of corrosion along the unprotected structure correlate strongly with experimental results within the experimental conditions studied.


RSC Advances | 2017

Graphene oxide reinforced hydrogels for osteogenic differentiation of human adipose-derived stem cells

Myungkyung Noh; Su-Hwan Kim; Jiyong Kim; J.H. Lee; Gun-Jae Jeong; Jeong-Kee Yoon; Seokyung Kang; Suk Ho Bhang; Hee Hun Yoon; Jong-Chan Lee; Nathaniel S. Hwang; Byung-Soo Kim

Polyethylene glycol (PEG)-based hydrogels are attractive biomaterials for stem cell culture due to their tunable material properties and mechanical strength. However, the lack of cell adhesion sites has been one of the major obstacles in generating functional tissue constructs using PEG-based hydrogels. To overcome this limitation, we designed graphene oxide (GO)-functionalized polyethylene glycol diacrylate (PEGDA) hydrogels to assign cell adhesion-dependent biofunctionality. The incorporation of GO into three-dimensional PEGDA networks improved cell attachment, engaged focal adhesion, and activated focal adhesion kinase (FAK) signaling of hydrogel-encapsulated human adipose-derived stem cells (hADSCs). Compared to the control PEGDA hydrogel, GO functionalized PEGDA hydrogel (PEGDA-GO) resulted in enhanced cell viability and survival. When subsequently cultured under osteoinductive condition, PEGDA-GO enhanced osteogenic differentiation and stimulated osteogenic phenotypes compared to those in its PEGDA counterpart. Taken together, GO could serve as an effective biofunctionalizing moiety to modulate stem cell adhesion and differentiation.


Biotechnology Progress | 2016

Therapeutic angiogenesis using tumor cell-conditioned medium

Hyeon-Ki Jang; Byung-Soo Kim; Jin Han; Jeong-Kee Yoon; J.H. Lee; Gun-Jae Jeong; Jung-Youn Shin

Stem cell‐conditioned medium (CM), which contains angiogenic factors that are secreted by stem cells, represents a potential therapy for ischemic diseases. Along with stem cells, tumor cells also secrete various angiogenic factors. Here, tumor cells as a cell source of CM for therapeutic angiogenesis was evaluated and the therapeutic efficacy of tumor cell CM in mouse hindlimb ischemia models was demonstrated. CM obtained from a human fibrosarcoma HT1080 cell line culture was compared with CM obtained from a human bone marrow‐derived mesenchymal stem cell (MSC) culture. HT1080 CM contained higher concentrations of angiogenic factors compared with MSC CM, which was attributable to the higher cell density that resulted from a much faster growth rate of HT1080 cells compared with MSCs. For use in in vitro and in vivo angiogenesis studies, HT1080 CM was diluted such that HT1080 CM and MSC CM would have the same cell number basis. The two types of CMs induced the same extent of human umbilical vein endothelial cell (HUVEC) proliferation in vitro. The injection of HT1080 CM into mouse ischemic limbs significantly improved capillary density and blood perfusion compared with the injection of fresh medium. Although the therapeutic outcome of HT1080 CM was similar to that of MSC CM, the preparation of CM by tumor cell line culture would be much more efficient due to the faster growth and unlimited life‐time of the tumor cell line. These data suggest the potential application of tumor cell CM as a therapeutic modality for angiogenesis and ischemic diseases.


ACS Nano | 2018

Dual Roles of Graphene Oxide To Attenuate Inflammation and Elicit Timely Polarization of Macrophage Phenotypes for Cardiac Repair

Jin Han; Yong Sook Kim; Min-Young Lim; Han Young Kim; Saerom Kong; Mikyung Kang; Yeon Woong Choo; Ju Hee Jun; Seungmi Ryu; Hye-yun Jeong; Jooyeon Park; Gun-Jae Jeong; Jong-Chan Lee; Gwang Hyeon Eom; Youngkeun Ahn; Byung-Soo Kim

Development of localized inflammatory environments by M1 macrophages in the cardiac infarction region exacerbates heart failure after myocardial infarction (MI). Therefore, the regulation of inflammation by M1 macrophages and their timely polarization toward regenerative M2 macrophages suggest an immunotherapy. Particularly, controlling cellular generation of reactive oxygen species (ROS), which cause M1 differentiation, and developing M2 macrophage phenotypes in macrophages propose a therapeutic approach. Previously, stem or dendritic cells were used in MI for their anti-inflammatory and cardioprotective potentials and showed inflammation modulation and M2 macrophage progression for cardiac repair. However, cell-based therapeutics are limited due to invasive cell isolation, time-consuming cell expansion, labor-intensive and costly ex vivo cell manipulation, and low grafting efficiency. Here, we report that graphene oxide (GO) can serve as an antioxidant and attenuate inflammation and inflammatory polarization of macrophages via reduction in intracellular ROS. In addition, GO functions as a carrier for interleukin-4 plasmid DNA (IL-4 pDNA) that propagates M2 macrophages. We synthesized a macrophage-targeting/polarizing GO complex (MGC) and demonstrated that MGC decreased ROS in immune-stimulated macrophages. Furthermore, DNA-functionalized MGC (MGC/IL-4 pDNA) polarized M1 to M2 macrophages and enhanced the secretion of cardiac repair-favorable cytokines. Accordingly, injection of MGC/IL-4 pDNA into mouse MI models attenuated inflammation, elicited early polarization toward M2 macrophages, mitigated fibrosis, and improved heart function. Taken together, the present study highlights a biological application of GO in timely modulation of the immune environment in MI for cardiac repair. Current therapy using off-the-shelf material GO may overcome the shortcomings of cell therapies for MI.


International Journal of Molecular Sciences | 2018

A Disposable Photovoltaic Patch Controlling Cellular Microenvironment for Wound Healing

Hyeon-Ki Jang; Jin Oh; Gun-Jae Jeong; Tae-Jin Lee; Gwang-Bum Im; J.H. Lee; Jeong-Kee Yoon; Dong Ik Kim; Byung-Soo Kim; Suk Ho Bhang; Tae Lee

Electrical stimulation (ES) is known to affect the wound healing process by modulating skin cell behaviors. However, the conventional clinical devices that can generate ES for promoting wound healing require patient hospitalization due to large-scale of the extracorporeal devices. Herein, we introduce a disposable photovoltaic patch that can be applied to skin wound sites to control cellular microenvironment for promoting wound healing by generating ES. In vitro experiment results show that exogenous ES could enhance cell migration, proliferation, expression of extracellular matrix proteins, and myoblast differentiation of fibroblasts which are critical for wound healing. Our disposable photovoltaic patches were attached to the back of skin wound induced mice. Our patch successfully provided ES, generated by photovoltaic energy harvested from the organic solar cell under visible light illumination. In vivo experiment results show that the patch promoted cutaneous wound healing via enhanced host-inductive cell proliferation, cytokine secretion, and protein synthesis which is critical for wound healing process. Unlike the current treatments for wound healing that engage passive healing processes and often are unsuccessful, our wearable photovoltaic patch can stimulate regenerative activities of endogenous cells and actively contribute to the wound healing processes.


ACS Nano | 2018

M1 Macrophage-Derived Nanovesicles Potentiate the Anticancer Efficacy of Immune Checkpoint Inhibitors

Yeon Woong Choo; Mikyung Kang; Han Young Kim; Jin Han; Seokyung Kang; J.H. Lee; Gun-Jae Jeong; Sung Pil Kwon; Seuk Young Song; Seokhyeong Go; Mungyo Jung; Jihye Hong; Byung-Soo Kim

Cancer immunotherapy modulates immune cells to induce antitumor immune responses. Tumors employ immune checkpoints to evade immune cell attacks. Immune checkpoint inhibitors such as anti-PD-L1 antibody (aPD-L1), which is being used clinically for cancer treatments, can block immune checkpoints so that the immune system can attack tumors. However, immune checkpoint inhibitor therapy may be hampered by polarization of macrophages within the tumor microenvironment (TME) into M2 tumor-associated macrophages (TAMs), which suppress antitumor immune responses and promote tumor growth by releasing anti-inflammatory cytokines and angiogenic factors. In this study, we used exosome-mimetic nanovesicles derived from M1 macrophages (M1NVs) to repolarize M2 TAMs to M1 macrophages that release pro-inflammatory cytokines and induce antitumor immune responses and investigated whether the macrophage repolarization can potentiate the anticancer efficacy of aPD-L1. M1NV treatment induced successful polarization of M2 macrophages to M1 macrophages in vitro and in vivo. Intravenous injection of M1NVs into tumor-bearing mice suppressed tumor growth. Importantly, injection of a combination of M1NVs and aPD-L1 further reduced the tumor size, compared to the injection of either M1NVs or aPD-L1 alone. Thus, our study indicates that M1NV injection can repolarize M2 TAMs to M1 macrophages and potentiate antitumor efficacy of the checkpoint inhibitor therapy.


ACS Applied Materials & Interfaces | 2017

Therapeutic Angiogenesis via Solar Cell-Facilitated Electrical Stimulation

Gun-Jae Jeong; Jin Young Oh; Yeon-Ju Kim; Suk Ho Bhang; Hyeon-Ki Jang; Jin Han; Jeong-Kee Yoon; Sang-Mo Kwon; Tae Il Lee; Byung-Soo Kim

Cell therapy has been suggested as a treatment modality for ischemic diseases, but the poor survival and engraftment of implanted cells limit its therapeutic efficacy. To overcome such limitation, we used electrical stimulation (ES) derived from a wearable solar cell for inducing angiogenesis in ischemic tissue. ES enhanced the secretion of angiogenic growth factors and the migration of mesenchymal stem cells (MSCs), myoblasts, endothelial progenitor cells, and endothelial cells in vitro. In a mouse ischemic hindlimb model, ES generated by a solar cell and applied to the ischemic region promoted migration of MSCs toward the ischemic site and upregulated expression of angiogenic paracrine factors (vascular endothelial, basic fibroblast, and hepatocyte growth factors; and stromal cell-derived factor-1α). Importantly, solar cell-generated ES promoted the formation of capillaries and arterioles at the ischemic region, attenuated muscle necrosis and fibrosis, and eventually prevented loss of the ischemic limb. Solar cell ES therapy showed higher angiogenic efficacy than conventional MSC therapy. This study shows the feasibility of using solar cell ES as a novel treatment for therapeutic angiogenesis.

Collaboration


Dive into the Gun-Jae Jeong's collaboration.

Top Co-Authors

Avatar

Byung-Soo Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Suk Ho Bhang

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Jeong-Kee Yoon

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Tae-Jin Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

J.H. Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jin Han

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Seokyung Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Han Young Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hyeon-Ki Jang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Mikyung Kang

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge