Network


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

Hotspot


Dive into the research topics where Si Jun Park is active.

Publication


Featured researches published by Si Jun Park.


Cancer Prevention Research | 2013

Direct Targeting of MEK1/2 and RSK2 by Silybin Induces Cell-Cycle Arrest and Inhibits Melanoma Cell Growth

Mee Hyun Lee; Zunnan Huang; Dong Joon Kim; Sung Hyun Kim; Myoung Ok Kim; Sung Young Lee; Hua Xie; Si Jun Park; Jae Young Kim; Joydeb Kumar Kundu; Ann M. Bode; Young-Joon Surh; Zigang Dong

Abnormal functioning of multiple gene products underlies the neoplastic transformation of cells. Thus, chemopreventive and/or chemotherapeutic agents with multigene targets hold promise in the development of effective anticancer drugs. Silybin, a component of milk thistle, is a natural anticancer agent. In the present study, we investigated the effect of silybin on melanoma cell growth and elucidated its molecular targets. Our study revealed that silybin attenuated the growth of melanoma xenograft tumors in nude mice. Silybin inhibited the kinase activity of mitogen-activated protein kinase (MEK)-1/2 and ribosomal S6 kinase (RSK)-2 in melanoma cells. The direct binding of silybin with MEK1/2 and RSK2 was explored using a computational docking model. Treatment of melanoma cells with silybin attenuated the phosphorylation of extracellular signal-regulated kinase (ERK)-1/2 and RSK2, which are regulated by the upstream kinases MEK1/2. The blockade of MEK1/2-ERK1/2-RSK2 signaling by silybin resulted in a reduced activation of NF-κB, activator protein-1, and STAT3, which are transcriptional regulators of a variety of proliferative genes in melanomas. Silybin, by blocking the activation of these transcription factors, induced cell-cycle arrest at the G1 phase and inhibited melanoma cell growth in vitro and in vivo. Taken together, silybin suppresses melanoma growth by directly targeting MEK- and RSK-mediated signaling pathways. Cancer Prev Res; 6(5); 455–65. ©2013 AACR.


Carcinogenesis | 2014

[6]-Shogaol inhibits growth and induces apoptosis of non-small cell lung cancer cells by directly regulating Akt1/2

Myoung Ok Kim; Mee Hyun Lee; Naomi Oi; Sung Hyun Kim; Ki Beom Bae; Zunnan Huang; Dong Joon Kim; Kanamata Reddy; Sung Young Lee; Si Jun Park; Jae Young Kim; Hua Xie; Joydeb Kumar Kundu; Zae Young Ryoo; Ann M. Bode; Young-Joon Surh; Zigang Dong

Non-small cell lung cancer (NSCLC) is the leading cause of cancer mortality worldwide. Despite progress in developing chemotherapeutics for the treatment of NSCLC, primary and secondary resistance limits therapeutic success. NSCLC cells exhibit multiple mutations in the epidermal growth factor receptor (EGFR), which cause aberrant activation of diverse cell signaling pathways. Therefore, suppression of the inappropriate amplification of EGFR downstream signaling cascades is considered to be a rational therapeutic and preventive strategy for the management of NSCLC. Our initial molecular target-oriented virtual screening revealed that the ginger components, including [6]-shogaol, [6]-paradol and [6]-gingerol, seem to be potential candidates for the prevention and treatment of NSCLC. Among the compounds, [6]-shogaol showed the greatest inhibitory effects on the NSCLC cell proliferation and anchorage-independent growth. [6]-Shogaol induced cell cycle arrest (G1 or G2/M) and apoptosis. Furthermore, [6]-shogaol inhibited Akt kinase activity, a downstream mediator of EGFR signaling, by binding with an allosteric site of Akt. In NCI-H1650 lung cancer cells, [6]-shogaol reduced the constitutive phosphorylation of signal transducer and activator of transcription-3 (STAT3) and decreased the expression of cyclin D1/3, which are target proteins in the Akt signaling pathway. The induction of apoptosis in NCI-H1650 cells by [6]-shogaol corresponded with the cleavage of caspase-3 and caspase-7. Moreover, intraperitoneal administration of [6]-shogaol inhibited the growth of NCI-H1650 cells as tumor xenografts in nude mice. [6]-Shogaol suppressed the expression of Ki-67, cyclin D1 and phosphorylated Akt and STAT3 and increased terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling-positivity in xenograft tumors. The current study clearly indicates that [6]-shogaol can be exploited for the prevention and/or treatment of NSCLC.


Experimental and Molecular Medicine | 2012

Over-expression of extracellular superoxide dismutase in mouse synovial tissue attenuates the inflammatory arthritis.

Dong Hoon Yu; Jun Koo Yi; Hyung Soo Yuh; Seo Jin Park; Hei Jung Kim; Ki Beom Bae; Young Rae Ji; Na Ri Kim; Si Jun Park; Do-Hyung Kim; Sung Hyun Kim; Myoung Ok Kim; Jeong Woong Lee; Zae Young Ryoo

Oxidative stress such as reactive oxygen species (ROS) within the inflamed joint have been indicated as being involved as inflammatory mediators in the induction of arthritis. Correlations between extracellular-superoxide dismutase (EC-SOD) and inflammatory arthritis have been shown in several animal models of RA. However, there is a question whether the over-expression of EC-SOD on arthritic joint also could suppress the progression of disease or not. In the present study, the effect on the synovial tissue of experimental arthritis was investigated using EC-SOD over-expressing transgenic mice. The over-expression of EC-SOD in joint tissue was confirmed by RT-PCR and immunohistochemistry. The degree of the inflammation in EC-SOD transgenic mice was suppressed in the collagen-induced arthritis model. In a cytokine assay, the production of pro-inflammatory cytokines such as, IL-1β, TNFα, and matrix metalloproteinases (MMPs) was decreased in fibroblast-like synoviocyte (FLS) but not in peripheral blood. Histological examination also showed repressed cartilage destruction and bone in EC-SOD transgenic mice. In conclusion, these data suggest that the over-expression of EC-SOD in FLS contributes to the activation of FLS and protection from joint destruction by depressing the production of the pro-inflammatory cytokines and MMPs. These results provide EC-SOD transgenic mice with a useful animal model for inflammatory arthritis research.


Journal of Biological Chemistry | 2012

Enforced Expression of Roquin Protein in T Cells Exacerbates the Incidence and Severity of Experimental Arthritis

Young Rae Ji; Hei Jung Kim; Dong Hoon Yu; Ki Beom Bae; Seo Jin Park; Jun Koo Yi; Nari Kim; Si Jun Park; Keon Bong Oh; Sung Soo Hwang; Sanggyu Lee; Sung Hyun Kim; Myoung Ok Kim; Jeong Woong Lee; Zae Young Ryoo

Background: Roquin regulates inflammation in autoimmune disease. Results: Roquin overexpression promoted the induction of collagen-induced arthritis (CIA). Conclusion: Enforced Roquin expression was produced in mice and used to demonstrate the importance of inflammation and CIA development in experimental arthritis. Significance: Roquin overexpression may induce inflammation in some autoimmune diseases. To investigate the role of Roquin, a RING-type ubiquitin ligase family member, we used transgenic mice with enforced Roquin expression in T cells, with collagen-induced arthritis (CIA). Wild-type (WT) and Roquin transgenic (Tg) mice were immunized with bovine type II collagen (CII). Arthritis severity was evaluated by clinical score; histopathologic CIA severity; proinflammatory and anti-inflammatory cytokine levels; anti-CII antibody levels; and populations of Th1, Th2, germinal center B cells, and follicular helper T cells in CIA. T cell proliferation in vitro and cytokine levels were determined to assess the response to CII. Roquin Tg mice developed more severe CIA and joint destruction compared with WT mice. Production of TNF-α, IFN-γ, IL-6, and pathogenic anti-collagen CII-specific IgG and IgG2a antibodies was increased in Roquin Tg mice. In addition, in vitro T cell assays showed increased proliferation and proinflammatory cytokine production in response to CII as a result of enforced Roquin expression in T cells. Furthermore, the Th1/Th2 balance was altered by an increased Th1 and decreased Th2 population. These findings suggest that overexpression of Roquin exacerbates the development of CIA and that enforced expression of Roquin in T cells may promote autoimmune diseases such as CIA.


Biochemical and Biophysical Research Communications | 2014

Overexpression of Jazf1 reduces body weight gain and regulates lipid metabolism in high fat diet.

Woo Young Jang; Ki Beom Bae; Sung Hyun Kim; Dong Hun Yu; Hei Jung Kim; Young Rae Ji; Seo Jin Park; Si Jun Park; Min Cheol Kang; Ja In Jeong; Sang Joon Park; Sang Gyu Lee; In-Kyu Lee; Myoung Ok Kim; Duhak Yoon; Zae Young Ryoo

Jazf1 is a 27 kDa nuclear protein containing three putative zinc finger motifs that is associated with diabetes mellitus and prostate cancer; however, little is known about the role that this gene plays in regulation of metabolism. Recent evidence indicates that Jazf1 transcription factors bind to the nuclear orphan receptor TR4. This receptor regulates PEPCK, the key enzyme involved in gluconeogenesis. To elucidate Jazf1s role in metabolism, we fed a 60% fat diet for up to 15 weeks. In Jazf1 overexpression mice, weight gain was found to be significantly decreased. The expression of Jazf1 in the liver also suppressed lipid accumulation and decreased droplet size. These results suggest that Jazf1 plays a critical role in the regulation of lipid homeostasis. Finally, Jazf1 may provide a new therapeutic target in the management of obesity and diabetes.


Arthritis & Rheumatism | 2012

Regulation of inflammatory responses and fibroblast‐like synoviocyte apoptosis by calcineurin‐binding protein 1 in mice with collagen‐induced arthritis

Jun Koo Yi; Hei Jung Kim; Dong Hoon Yu; Seo Jin Park; Mi Jung Shin; Hyung Soo Yuh; Ki Beom Bae; Young Rae Ji; Na Ri Kim; Si Jun Park; Jae Young Kim; Hyun Shik Lee; Sang Gyu Lee; Du Hak Yoon; Byung Hwa Hyun; Wan Uk Kim; Zae Young Ryoo

OBJECTIVE Calcineurin-binding protein 1 (CABIN-1) regulates calcineurin phosphatase activity as well as the activation, apoptosis, and inflammatory responses of fibroblast-like synoviocytes (FLS), which actively participate in the chronic inflammatory responses in rheumatoid arthritis (RA). However, the mechanism of action of CABIN-1 in FLS apoptosis is not clear. This study was undertaken to define the regulatory role of CABIN-1 in FLS from mice with collagen-induced arthritis (CIA). METHODS Transgenic mice overexpressing human CABIN-1 in joint tissue under the control of a type II collagen promoter were generated. Expression of human CABIN-1 (hCABIN-1) in joints and FLS was determined by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot analysis. The expression of cytokines, matrix metalloproteinases (MMPs), and apoptosis-related genes in FLS was determined by enzyme-linked immunosorbent assay, gelatin zymography, and RT-PCR, respectively. Joints were stained with hematoxylin and eosin and with tartrate-resistant acid phosphatase for histologic analysis. RESULTS Human CABIN-1-transgenic mice with CIA had less severe arthritis than wild-type mice with CIA, as assessed according to hind paw thickness and histologic features. The milder arthritis was accompanied by significantly enhanced apoptosis in transgenic mice, evidenced by a significantly greater number of TUNEL-positive cells in synovial tissue. Expression of inflammatory cytokines and MMPs in the transgenic mice with CIA was reduced, and they exhibited decreased Akt activation and increased expression of p53, caspase 3, caspase 9, and Bax. CONCLUSION Our findings demonstrate that hCABIN-1 plays a critical role in promoting apoptosis of FLS and in attenuating inflammation and cartilage and bone destruction in RA. These results help elucidate the pathogenic mechanisms of RA and suggest that CABIN-1 is a potential target for treatment of this disease.


Biochemical and Biophysical Research Communications | 2012

The role of Roquin overexpression in the modulation of signaling during in vitro and ex vivo T-cell activation

Hei Jung Kim; Young Rae Ji; Myoung Ok Kim; Dong Hoon Yu; Mi Jung Shin; Hyung Soo Yuh; Ki Beom Bae; Seo Jin Park; Jun Koo Yi; Na Ri Kim; Si Jun Park; Du Hak Yoon; Won-Ha Lee; Sanggyu Lee; Zae Young Ryoo

The T-cell receptor (TCR) engages with an antigen and initiates a signaling cascade that leads to the activation of transcription factors. Roquin, a protein encoded by the RC3H1 gene and characterized as an immune regulator, was recently identified as a novel RING-type ubiquitin ligase family member, but the mechanisms by which Roquin regulates T-cell responses are unclear. We used the EL-4 murine lymphoma cell line to elucidate the role of Roquin in vitro. Roquin-overexpressing EL-4 cells became hyper-responsive after anti-CD3/CD28 stimulation in vitro and were a major source of the cytokines IL-2 and TNF-α. Upon activation, these cells showed particularly enhanced production of IL-2 and TNF-α. To clarify the important role played by Roquin in T-cell responses ex vivo, we generated T-cell-specific Roquin transgenic (Tg) mice. Roquin-Tg CD4(+) T-cells showed enhanced production of IL-2 and TNF-α in response to TCR stimulation with anti-CD28 co-stimulation. Further studies are necessary to investigate the role of Roquin in the regulation of primary T-cell activation, survival, and differentiation.


Cellular Signalling | 2015

Non-receptor tyrosine kinase inhibitors enhances β-cell survival by suppressing the PKCδ signal transduction pathway in streptozotocin-induced β-cell apoptosis.

Udayakumar Karunakaran; Si Jun Park; Do Youn Jun; Taebo Sim; Keun-Gyu Park; Myoung Ok Kim; In-Kyu Lee

GNF-2 and GNF-5 are members of a new class of non-receptor tyrosine kinases inhibitors that possess excellent selectivity towards imatinib-resistant mutations found in chronic myeloid leukemia patients. On the other hand recent reports implicate abnormal tyrosine kinase signaling in β-cell death in Type I and Type II diabetes. In this work we determined the effects of GNF-2, GNF-5 on pancreatic β-cell death caused by streptozotocin (STZ). STZ treatment causes apoptosis of INS-1 cells by activation of intracellular ROS, c-jun N-terminal kinase (JNK), caspase 3, and caspase 3-dependent activation of protein kinase C delta (PKCδ). GNF-2 and GNF-5 increased cell viability and attenuated STZ-induced intracellular ROS and significantly reduced the activation of JNK, caspase 3, and caspase 3-dependent activation of PKCδ. In studies with intact mice, GFN-2 and GNF-5 prevented the loss of beta cells and the increase in blood glucose produced by STZ-treated control mice. Furthermore, immunohistochemical analysis revealed that GNF-2 and GNF-5 increased insulin protein levels in STZ-treated mice when compared with control mice. These findings suggest that non-receptor tyrosine kinase inhibitors provide a new approach for the treatment of new-onset Type I and Type II diabetes.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Gestational Loss and Growth Restriction by Angiogenic Defects in Placental Growth Factor Transgenic Mice

Min Cheol Kang; Seo Jin Park; Hei Jung Kim; Jinhee Lee; Dong Hoon Yu; Ki Beom Bae; Young Rae Ji; Si Jun Park; Jain Jeong; Woo Young Jang; Jung Hak Kim; Myung Sook Choi; Dong Seok Lee; Hyun Shik Lee; Sanggyu Lee; Sung Hyun Kim; Myoung Ok Kim; Gyeongsin Park; Yeon Sik Choo; Je Yoel Cho; Zae Young Ryoo

Objective— Angiogenesis is an important biological process during development, reproduction, and in immune responses. Placental growth factor (PlGF) is a member of vascular endothelial growth factor that is critical for angiogenesis and vasculogenesis. We generated transgenic mice overexpressing PlGF in specifically T cells using the human CD2-promoter to investigate the effects of PlGF overexpression. Approach and Results— Transgenic mice were difficult to obtain owing to high lethality; for this reason, we investigated why gestational loss occurred in these transgenic mice. Here, we report that placenta detachment and inhibition of angiogenesis occurred in PlGF transgenic mice during the gestational period. Moreover, even when transgenic mice were born, their growth was restricted. Conclusions— Conclusively, PlGF overexpression prevents angiogenesis by inhibiting Braf, extracellular signal–regulated kinase activation, and downregulation of HIF-1&agr; in the mouse placenta. Furthermore, it affected regulatory T cells, which are important for maintenance of pregnancy.


Biochemical and Biophysical Research Communications | 2014

Over-expression of Roquin aggravates T cell mediated hepatitis in transgenic mice using T cell specific promoter.

Young Rae Ji; Hei Jung Kim; Dong Hun Yu; Ki Beom Bae; Seo Jin Park; Si Jun Park; Woo Young Jang; Min Cheol Kang; Jain Jeong; Yong Hun Sung; Minjee Choi; Taejun Park; Taesun Park; Jong Won Yun; Hyun Shik Lee; Sanggyu Lee; Myoung Ok Kim; Zae Young Ryoo

Chronic hepatitis is a major cause of liver cancer, so earlier treatment of hepatitis might be reducing liver cancer incidence. Hepatitis can be induced in mice by treatment with Concanavalin A (Con A); the resulting liver injury causes significant CD4(+) T cell activation and infiltration. In these T cells, Roquin, a ring-type E3 ubiquitin ligase, is activated. To investigate the role of Roquin, we examined Con A-induced liver injury and T cell infiltration in transgenic (Tg) mice overexpressing Roquin specifically in T cells. In Roquin Tg mice, Con A treatment caused greater increases in both the levels of liver injury enzymes and liver tissue apoptosis, as revealed by TUNEL and H&E staining, than wild type (WT) mice. Further, Roquin Tg mice respond to Con A treatment with greater increases in the T cell population, particularly Th17 cells, though Treg cell counts are lower. Roquin overexpression also enhances increases in pro-inflammatory cytokines, including IFN-γ, TNF-α and IL-6, upon liver injury. Furthermore, Roquin regulates the immune response and apoptosis in Con A induced hepatitis via STATs, Bax and Bcl2. These findings suggest that over-expression of Roquin exacerbates T-cell mediated hepatitis.

Collaboration


Dive into the Si Jun Park's collaboration.

Top Co-Authors

Avatar

Myoung Ok Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Zae Young Ryoo

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Sung Hyun Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Hei Jung Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Seo Jin Park

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Young Rae Ji

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Ki Beom Bae

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Dong Hoon Yu

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Jain Jeong

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Sanggyu Lee

Kyungpook National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge