Network


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

Hotspot


Dive into the research topics where So Jin Bing is active.

Publication


Featured researches published by So Jin Bing.


Journal of Biological Chemistry | 2014

Lipocalin-2 Protein Deficiency Ameliorates Experimental Autoimmune Encephalomyelitis THE PATHOGENIC ROLE OF LIPOCALIN-2 IN THE CENTRAL NERVOUS SYSTEM AND PERIPHERAL LYMPHOID TISSUES

Youngpyo Nam; Jong-Heon Kim; Minchul Seo; Jae-Hong Kim; Myungwon Jin; Sangmin Jeon; Jung-Wan Seo; Won-Ha Lee; So Jin Bing; Youngheun Jee; Won Kee Lee; Dong Ho Park; Hyun Kook; Kyoungho Suk

Background: The role of LCN2 in EAE is not clear. Results: LCN2 expression increased in EAE. Lcn2 deficiency attenuated EAE symptoms and pathology. LCN2 enhanced glial expression of inflammatory mediators and peripheral encephalitogenic T cell activation in vitro and in vivo. Conclusion: Both central and peripheral LCN2 contributed to EAE development. Significance: LCN2 can be targeted for treatment of multiple sclerosis. Lipocalin-2 (LCN2) plays an important role in cellular processes as diverse as cell growth, migration/invasion, differentiation, and death/survival. Furthermore, recent studies indicate that LCN2 expression and secretion by glial cells are induced by inflammatory stimuli in the central nervous system. The present study was undertaken to examine the regulation of LCN2 expression in experimental autoimmune encephalomyelitis (EAE) and to determine the role of LCN2 in the disease process. LCN2 expression was found to be strongly increased in spinal cord and secondary lymphoid tissues after EAE induction. In spinal cords astrocytes and microglia were the major cell types expressing LCN2 and its receptor 24p3R, respectively, whereas in spleens, LCN2 and 24p3R were highly expressed in neutrophils and dendritic cells, respectively. Furthermore, disease severity, inflammatory infiltration, demyelination, glial activation, the expression of inflammatory mediators, and the proliferation of MOG-specific T cells were significantly attenuated in Lcn2-deficient mice as compared with wild-type animals. Myelin oligodendrocyte glycoprotein-specific T cells in culture exhibited an increased expression of Il17a, Ifng, Rorc, and Tbet after treatment with recombinant LCN2 protein. Moreover, LCN2-treated glial cells expressed higher levels of proinflammatory cytokines, chemokines, and MMP-9. Adoptive transfer and recombinant LCN2 protein injection experiments suggested that LCN2 expression in spinal cord and peripheral immune organs contributes to EAE development. Taken together, these results imply LCN2 is a critical mediator of autoimmune inflammation and disease development in EAE and suggest that LCN2 be regarded a potential therapeutic target in multiple sclerosis.


International Journal of Radiation Biology | 2010

Dieckol rescues mice from lethal irradiation by accelerating hemopoiesis and curtailing immunosuppression

Eun-Jin Park; Ginnae Ahn; Jin-Suk Yun; Min Ju Kim; So Jin Bing; Dae Seung Kim; Jehee Lee; Nam Ho Lee; Jae Woo Park; Youngheun Jee

Purpose: To evaluate the radioprotective effect of dieckol, a component of the brown algae Ecklonia cava. Materials and methods: Mice were lethally irradiated, treated with dieckol and surveyed for survival and the mechanism of radioprotection. Results: Dieckol treatment lengthened the survival of irradiated mice and significantly accelerated the hemopoietic recovery of bone marrow cells and peripheral immune cells compared with irradiated, but untreated controls. Dieckol also enhanced the proliferation and differentiation of immune cells, which had been suppressed by ionising radiation. It turned out that the increased expression of oxygen radical-scavenging enzyme manganese superoxide dismutase (MnSOD) and subsequent reduction in DNA damage and lipid peroxidation was crucial for dieckols radioprotective effect. In addition, the dieckol-induced modulation of protein 53 (p53)-dependent pathways further strengthened the radioresistance of immune cells. Conclusion: Dieckol can be a promising agent for reducing the time needed for reconstitution of hemopoietic cells after exposure to irradiation.


Food and Chemical Toxicology | 2013

Geraniin down regulates gamma radiation-induced apoptosis by suppressing DNA damage.

So Jin Bing; Danbee Ha; Min Ju Kim; Eun-Jin Park; Ginnae Ahn; Dae Seung Kim; Ryeo Kyeong Ko; Jae Woo Park; Nam Ho Lee; Youngheun Jee

Gamma ray irradiation triggers DNA damage and apoptosis of proliferating stem cells and peripheral immune cells, resulting in the destruction of intestinal crypts and lymphoid system. Geraniin is a natural compound extracts from an aquatic plant Nymphaea tetragona and possesses good antioxidant property. In this study, we demonstrate that geraniin rescues radiosensitive splenocytes and jejunal crypt cells from radiation-induced DNA damage and apoptosis. Isolated splenocytes from C57BL/6 mice treated with geraniin were protected against radiation injury of 2 Gy irradiation through the enhancement of the proliferation and attenuation of DNA damage. Also, geraniin inhibited apoptosis in radiosensitive splenocytes by reducing the expression level and immunoreactivity of proapoptotic p53 and Bax and increasing those of anti-apoptotic Bcl-2. In mice exposed to radiation, geraniin treatment protected splenocytes and intestinal crypt cells from radiation-induced cell death. Our results suggest that geraniin presents radioprotective effects by regulating DNA damage on splenocytes, exerting immunostimulatory capacities and inhibiting apoptosis of radiosensitive immune cells and jejunal crypt cells. Therefore, geraniin can be a radioprotective agent against γ-irradiation exposure.


Acta Histochemica | 2014

Acidic polysaccharide of Panax ginseng regulates the mitochondria/caspase-dependent apoptotic pathway in radiation-induced damage to the jejunum in mice

So Jin Bing; Min Ju Kim; Ginnae Ahn; Jaehak Im; Dae Seung Kim; Danbee Ha; Jinhee Cho; Areum Kim; Youngheun Jee

Owing to its susceptibility to radiation, the small intestine of mice is valuable for studying radioprotective effects. When exposed to radiation, intestinal crypt cells immediately go through apoptosis, which impairs swift differentiation necessary for the regeneration of intestinal villi. Our previous studies have elucidated that acidic polysaccharide of Panax ginseng (APG) protects the mouse small intestine from radiation-induced damage by lengthening villi with proliferation and repopulation of crypt cells. In the present study, we identified the molecular mechanism involved. C57BL/6 mice were irradiated with gamma-rays with or without APG and the expression levels of apoptosis-related molecules in the jejunum were investigated using immunohistochemistry. APG pretreatment strongly decreased the radiation-induced apoptosis in the jejunum. It increased the expression levels of anti-apoptotic proteins (Bcl-2 and Bcl-XS/L) and dramatically reduced the expression levels of pro-apoptotic proteins (p53, BAX, cytochrome c and caspase-3). Therefore, APG attenuated the apoptosis through the intrinsic pathway, which is controlled by p53 and Bcl-2 family members. Results presented in this study suggest that APG protects the mouse small intestine from irradiation-induced apoptosis through inhibition of the p53-dependent pathway and the mitochondria/caspase pathway. Thus, APG may be a potential agent for preventing radiation induced injuries in intestinal cells during radio-therapy such as in cancer treatment.


Journal of Histochemistry and Cytochemistry | 2013

Phloroglucinol protects small intestines of mice from ionizing radiation by regulating apoptosis-related molecules: a comparative immunohistochemical study.

Danbee Ha; So Jin Bing; Jinhee Cho; Ginnae Ahn; Dae Seung Kim; Mohammad Al-Amin; Suk Jae Park; Youngheun Jee

Phloroglucinol (PG) is a phenolic compound isolated from Ecklonia cava, a brown algae abundant on Jeju island, Korea. Previous reports have suggested that PG exerts antioxidative and cytoprotective effects against oxidative stress. In this study, we confirmed that PG protected against small intestinal damage caused by ionizing radiation, and we investigated its protective mechanism in detail. Regeneration of intestinal crypts in the PG-treated irradiated group was significantly promoted compared with that in irradiated controls. The expression level of proapoptotic molecules such as p53, Bax, and Bak in the small intestine was downregulated and that of antiapoptotic molecules such as Bcl-2 and Bcl-XS/L was augmented in the PG-treated group. On histological observation of the small intestine, PG inhibited the immunoreactivity of p53, Bax, and Bak and increased that of Bcl-2 and Bcl-XS/L. These results demonstrate the protective mechanisms of PG in mice against intestinal damage from ionizing radiation, providing the benefit of raising the apoptosis threshold of jejunal crypt cells.


Molecular Neurodegeneration | 2016

IKKβ-mediated inflammatory myeloid cell activation exacerbates experimental autoimmune encephalomyelitis by potentiating Th1/Th17 cell activation and compromising blood brain barrier

Min Jung Lee; So Jin Bing; Jonghee Choi; Minhee Jang; Gihyun Lee; Hyunkyoung Lee; Byung Soo Chang; Youngheun Jee; Sung Joong Lee; Ik-Hyun Cho

BackgroundThe inflammatory myeloid cell activation is one of the hallmarks of experimental autoimmune encephalomyelitis (EAE), yet the in vivo role of the inflammatory myeloid cell activation in EAE has not been clearly resolved. It is well-known that IKK/NF-κB is a key signaling pathway that regulates inflammatory myeloid activation.MethodsWe investigated the in vivo role of inflammatory myeloid cell activation in myelin oligodendrocyte glycoprotein (MOG) peptides-induced EAE using myeloid cell type-specific ikkβ gene conditional knockout-mice (LysM-Cre/IkkβF/F).ResultsIn our study, LysM-Cre/IkkβF/F mice had alleviated clinical signs of EAE corresponding to the decreased spinal demyelination, microglial activation, and immune cell infiltration in the spinal cord, compared to the wild-type mice (WT, IkkβF/F). Myeloid ikkβ gene deletion significantly reduced the percentage of CD4+/IFN-γ+ (Th1) and CD4+/IL-17+ (Th17) cells but increased the percentages of CD4+/CD25+/Foxp3+ (Treg) cells in the spinal cord and lymph nodes, corresponding to the altered mRNA expression of IFN-γ, IL-17, IL-23, and Foxp3 in the spinal cords of LysM-Cre/IkkβF/F EAE mice. Also, the beneficial effect of myeloid IKKβ deletion in EAE corresponded to the decreased permeability of the blood brain barrier (BBB).ConclusionsOur findings strongly suggest that IKK/NF-kB-induced myeloid cell activation exacerbates EAE by activating Th1 and Th17 responses and compromising the BBB. The development of NF-κB inhibitory agents with high efficacy through specific targeting of IKKβ in myeloid cells might be of therapeutic potential in MS and other autoimmune disorders.


Food and Chemical Toxicology | 2012

Jeju ground water containing vanadium induced immune activation on splenocytes of low dose γ-rays-irradiated mice

Danbee Ha; Haejin Joo; Ginnae Ahn; Min Ju Kim; So Jin Bing; Subin An; Hyunki Kim; Kyung-goo Kang; Yoon-Kyu Lim; Youngheun Jee

Vanadium, an essential micronutrient, has been implicated in controlling diabetes and carcinogenesis and in impeding reactive oxygen species (ROS) generation. γ-ray irradiation triggers DNA damage by inducing ROS production and causes diminution in radiosensitive immunocytes. In this study, we elucidate the immune activation capacities of Jeju water containing vanadium on immunosuppression caused by γ-ray irradiation, and identify its mechanism using various low doses of NaVO(3). We examined the intracellular ROS generation, DNA damage, cell proliferation, population of splenocytes, and cytokine/antibody profiles in irradiated mice drinking Jeju water for 180 days and in non-irradiated and in irradiated splenocytes both of which were treated with NaVO(3). Both Jeju water and 0.245 μM NaVO(3) attenuated the intracellular ROS generation and DNA damage in splenocytes against γ-ray irradiation. Splenocytes were significantly proliferated by the long-term intake of Jeju water and by 0.245 μM NaVO(3) treatment, and the expansion of B cells accounted for the increased number of splenocytes. Also, 0.245 μM NaVO(3) treatment showed the potency to amplify the production of IFN-γ and total IgG in irradiated splenocytes, which correlated with the expansion of B cells. Collectively, Jeju water containing vanadium possesses the immune activation property against damages caused by γ-irradiation.


FEBS Journal | 2016

Blocking Glutamate Carboxypeptidase II Inhibits Glutamate Excitotoxicity and Regulates Immune Responses in Experimental Autoimmune Encephalomyelitis

Danbee Ha; So Jin Bing; Ginnae Ahn; Jinhee Kim; Jinhee Cho; Areum Kim; Kalahe Hewage Iresha Nadeeka Madushani Herath; Hak Sun Yu; Sangmee Ahn Jo; Ik-Hyun Cho; Youngheun Jee

Experimental autoimmune encephalomyelitis (EAE) is an inflammatory disease in the murine central nervous system (CNS) and recapitulates the clinical and pathological features of human multiple sclerosis (MS). Glutamate carboxipeptidase II (GCPII), an enzyme expressed exclusively on astrocytes, is known to affect the disease progression of various neurological disorders by producing glutamate. Despite several findings indicating possible link between glutamate and MS/EAE, however, the involvement of astrocyte or GCPII on glutamate excitotoxicity has not received much attention in MS/EAE. When we examined GCPII expression during EAE progression in this study, we observed significantly elevated GCPII expression in peak stage of disease localized mainly in astrocytes. Intrigued by these results, we tried a potent GCPII inhibitor, 2‐phosphonomethyl pentanedioic acid (2‐PMPA), on EAE mice and noticed markedly attenuated EAE clinical signs along with significantly inhibited infiltration of inflammatory cells into CNS. Furthermore, 2‐PMPA dampened the function of Th1 cell lineage and down‐regulated mGluR1 expression in both periphery and CNS contributing to glutamate‐mediated immune regulation. Our observations identify a sequence of events triggering EAE through GCPII overexpression, which may offer a novel therapeutic approach to the treatment of MS.


Pharmaceutical Biology | 2017

Beetroot (Beta vulgaris) rescues mice from γ-ray irradiation by accelerating hematopoiesis and curtailing immunosuppression

Jinhee Cho; So Jin Bing; Areum Kim; Nam Ho Lee; Sang-Hee Byeon; Gi-Ok Kim; Youngheun Jee

Abstract Context: Beetroot [Beta vulgaris Linné (Chenopodiaceae)], a vegetable usually consumed as a food or a medicinal plant in Europe, has been reported to have antioxidant and anti-inflammatory properties. Since the lymphohematopoietic system is the most sensitive tissue to ionizing radiation, protecting it from radiation damage is one of the best ways to decrease detrimental effects from radiation exposure. Objective: In this study, we evaluated the radio-protective effects of beetroot in hematopoietic stem cells (HSCs) and progenitor cells. Materials and methods: Beetroot extract was administered at a dose of 400 mg/mouse per os (p.o.) three times into C57BL/6 mice and, at day 10 after γ-ray irradiation, diverse molecular presentations were measured and compared against non-irradiated and irradiated mice with PBS treatments. Survival of beetroot-fed and unfed irradiated animal was also compared. Results: Beetroot not only stimulated cell proliferation, but also minimized DNA damage of splenocytes. Beetroot also repopulated S-phase cells and increased Ki-67 or c-Kit positive cells in bone marrow. Moreover, beetroot-treated mice showed notable boosting of differentiation of HSCs into burst-forming units-erythroid along with increased production of IL-3. Also, beetroot-treated mice displayed enhancement in the level of hematocrit and hemoglobin as well as the number of red blood cell in peripheral blood. Beetroot diet improved survival rate of lethally exposed mice with a dose reduction factor (DRF) of 1.1. Discussion and conclusion: These results suggest that beetroot has the potency to preserve bone marrow integrity and stimulate the differentiation of HSCs against ionizing radiation.


Biomedicine & Pharmacotherapy | 2016

Anti-inflammatory activities of Dangyuja (Citrus grandis Osbeck) in concanavalin A stimulated murine splenocytes and 12-O-tetradecanoylphorbol-13-acetate-induced murine skin edema

Kalahe Hewage Iresha Nadeeka Madushani Herath; So Jin Bing; Jinhee Cho; Areum Kim; Sumin Shin; Gi-Ok Kim; Jong-chul Lee; Youngheun Jee

Dangyuja (Citrus grandis Osbeck), a citrus cultivated in southern Korea, has been used in traditional medicine for its anti-inflammatory effect. In this study, we investigated the anti-inflammatory potential of extract of Citrus grandis Osbeck (ECGO). In in vitro assays, ECGO treatment of concanavalin A (10μg/ml, for 24h) stimulated splenocytes showed significant reduction in CD44/CD62L+ T cell population and a marked decrease in the production of inflammatory cytokines IL-2, IFN-γ and IL-4. Interestingly, in vivo assays of ECGO topical treatment (100μg/20μl/ear) significantly mitigated the TPA (4μg/20μl/ear) induced edema induction and Myeloperoxidase activity. Anti-inflammatory potential of ECGO were further evidenced through its potent decrease in expression of inducible nitric oxide, cyclooxygenase-2, IL-1β and TNF-α and suppressed homing of CD3+ T cells and F4/80+ macrophages to site of inflammation. This study emphasizes the possibility of developing ECGO as an alternative natural topical agent to combat inflammatory diseases.

Collaboration


Dive into the So Jin Bing's collaboration.

Top Co-Authors

Avatar

Jinhee Cho

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Areum Kim

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Ginnae Ahn

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Youngheun Jee

St. Joseph's Hospital and Medical Center

View shared research outputs
Top Co-Authors

Avatar

Youngheun Jee

St. Joseph's Hospital and Medical Center

View shared research outputs
Top Co-Authors

Avatar

Danbee Ha

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Dae Seung Kim

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Min Ju Kim

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Eun-Jin Park

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge