Network


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

Hotspot


Dive into the research topics where Sun-O Ka is active.

Publication


Featured researches published by Sun-O Ka.


Molecular and Cellular Endocrinology | 2007

Genistein protects pancreatic β cells against cytokine-mediated toxicity

Eun-Kyung Kim; Kang-Beom Kwon; Mi-Young Song; Sang-Wan Seo; Sung-Joo Park; Sun-O Ka; Lv Na; Kyung-Ah Kim; Do-Gon Ryu; Hong-Seob So; Raekil Park; Jin-Woo Park; Byung-Hyun Park

In the past few decades, the use of genistein as an anti-inflammatory agent has gained much attention. Our current study focuses on the preventive effects of genistein on cytokine-induced pancreatic beta-cell damage. Treatment of RINm5F (RIN) rat insulinoma cells with interleukin (IL)-1beta and interferon (IFN)-gamma induced cell damage, which was correlated with nitric oxide (NO) production. Genistein completely prevented cytokine-mediated cytotoxicity and NO production, a finding that correlated well with reduced levels of the inducible form of NO synthase (iNOS) mRNA and protein. The molecular mechanism of genistein inhibition of iNOS gene expression appeared to involve the inhibition of NFkappaB activation. The cytokine induced increases in NFkappaB binding activity, nuclear p50 and p65 subunit levels, and IkappaBalpha degradation in cytosol compared to unstimulated cells; genistein abolished all of these parameters. The cytoprotective effects of genistein are also mediated through the suppression of ERK-1/2 and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathways. In a second set of experiments, rat islets were used. The findings on beta-cell protective effects of genistein were essentially the same as for the RIN cell data, namely genistein prevented cytokine-induced NO production, iNOS expression, ERK-1/2 activation, JAK/STAT activation, and impairment of glucose-stimulated insulin secretion. Collectively, these results suggest that genistein might be used to preserve functional beta-cell mass.


Arthritis & Rheumatism | 2013

Overexpression of Sirtuin 6 Suppresses Inflammatory Responses and Bone Destruction in Mice With Collagen-Induced Arthritis

Hwa-Suk Lee; Sun-O Ka; Sang-Myeong Lee; Sang-Il Lee; Jin-Woo Park; Byung-Hyun Park

OBJECTIVE Sirtuin 6 (SIRT-6) is an NAD(+) -dependent deacetylase and mono-ADP-ribosyltransferase. It is known to interfere with the NF-κB signaling pathway and thereby has an antiinflammatory function. Due to the central role of NF-κB in rheumatoid arthritis (RA) development, we undertook this study to test our hypothesis that SIRT-6 could have antiarthritic effects. METHODS An adenovirus containing SIRT-6 complementary DNA (Ad-SIRT6) was used to deliver SIRT-6 to human RA fibroblast-like synoviocytes in vitro as well as to mice with collagen-induced arthritis (CIA) in vivo via bilateral intraarticular injections into the ankle joints. RESULTS In vitro experiments demonstrated that SIRT-6 overexpression suppressed NF-κB target gene expression induced by tumor necrosis factor α. SIRT-6 overexpression inhibited osteoclast differentiation induced by macrophage colony-stimulating factor and RANKL in bone marrow-derived macrophages. Mice with CIA had an increased incidence of disease and developed arthritis in the hind paws. In contrast, mice injected with Ad-SIRT6 showed attenuated severity of arthritis based on clinical scores, hind paw thickness, and radiographic and pathologic findings. Moreover, the injection of Ad-SIRT6 down-regulated local and systemic levels of proinflammatory cytokines. After induction of CIA, mice injected with Ad-SIRT6 showed significantly decreased arthritis severity, from the onset of clinical signs to the end of the study. CONCLUSION These results suggest that blocking the NF-κB pathway by SIRT-6 in rheumatoid joints reduces both the inflammatory response and tissue destruction. Therefore, the development of an immunoregulatory strategy based on SIRT-6 may have therapeutic potential for the treatment of RA.


Experimental and Molecular Medicine | 2010

Sulfuretin protects against cytokine-induced

Mi-Young Song; Gil-Saeng Jeong; Kang-Beom Kwon; Sun-O Ka; Hyun-Young Jang; Jin-Woo Park; Youn-Chul Kim; Byung-Hyun Park

NF-κB activation has been implicated as a key signaling mechanism for pancreatic β-cell damage. Sulfuretin is one of the main flavonoids produced by Rhus verniciflua, which is reported to inhibit the inflammatory response by suppressing the NF-κB pathway. Therefore, we isolated sulfuretin from Rhus verniciflua and evaluated if sulfuretin could inhibit cytokine- or streptozotocin-induced β-cell damage. Rat insulinoma RINm5F cells and isolated rat islets were treated with IL-1β and IFN-γ to induce cytotoxicity. Incubation of cells and islets with sulfuretin resulted in a significant reduction of cytokine-induced NF-κB activation and its downstream events, iNOS expression, and nitric oxide production. The cytotoxic effects of cytokines were completely abolished when cells or islets were pretreated with sulfuretin. The protective effect of sulfuretin was further demonstrated by normal insulin secretion of cytokine-treated islets in response to glucose. Treatment of mice with streptozotocin resulted in hyperglycemia and hypoinsulinemia, which was further evidenced by immunohistochemical staining of islets. However, the diabetogenic effects of streptozotocin were completely prevented when mice were pretreated with sulfuretin. The anti-diabetogenic effects of sulfuretin were also mediated by suppression of NF-κB activation. Collectively, these results indicate that sulfuretin may have therapeutic value in preventing β-cell damage.


Experimental and Molecular Medicine | 2007

{\beta}

Eun-Kyung Kim; Kang-Beom Kwon; Mi-Jeong Han; Mi-Young Song; Ji-Hyun Lee; Na Lv; Sun-O Ka; Seung-Ryong Yeom; Young-Dal Kwon; Do-Gon Ryu; Kang-San Kim; Jin-Woo Park; Raekil Park; Byung-Hyun Park

We demonstrated previously that Coptidis rhizoma extract (CRE) prevented S-nitroso-N-acetylpenicillamine-induced apoptotic cell death via the inhibition of mitochondrial membrane potential disruption and cytochrome c release in RINm5F (RIN) rat insulinoma cells. In this study, the preventive effects of CRE against cytokine-induced β-cell death was assessed. Cytokines generated by immune cells infiltrating pancreatic islets are crucial mediators of β-cell destruction in insulin-dependent diabetes mellitus. The treatment of RIN cells with IL-1β and IFN-γ resulted in a reduction of cell viability. CRE completely protected IL-1β and IFN-γ-mediated cell death in a concentration-dependent manner. Incubation with CRE induced a significant suppression of IL-1β and IFN-γ-induced nitric oxide (NO) production, a finding which correlated well with reduced levels of the iNOS mRNA and protein. The molecular mechanism by which CRE inhibited iNOS gene expression appeared to involve the inhibition of NF-κ B activation. The IL-1β and IFN-γ-stimulated RIN cells showed increases in NF-κ B binding activity and p65 subunit levels in nucleus, and IκBα degradation in cytosol compared to unstimulated cells. Furthermore, the protective effects of CRE were verified via the observation of reduced NO generation and iNOS expression, and normal insulin-secretion responses to glucose in IL-1β and IFN-γ-treated islets.


Journal of Endocrinology | 2015

-cell damage and prevents streptozotocin-induced diabetes

Sun-O Ka; Mi-Young Song; Eun Ju Bae; Byung-Hyun Park

Inflammation is an important factor in the development of insulin resistance. SIRT1, a class 3 histone/protein deacetylase, has anti-inflammatory functions. Myeloid-specific deletion of Sirt1 promotes macrophage infiltration into insulin-sensitive organs and aggravates tissue inflammation. In this study, we investigated how SIRT1 in macrophages alters tissue inflammation in the pancreas as well as liver and adipose tissue, and further explored the role of SIRT1 in locomotion of macrophages. Myeloid-specific Sirt1-deleted mice (mS1KO) and WT littermates were fed a 60% calorie high-fat diet (HFD) for 16 weeks. Tissue inflammation and metabolic phenotypes were compared. Bone marrow macrophages (BMMs) from WT or mS1KO mice were used in in vitro chemotaxis assays and macrophage polarization studies. mS1KO mice fed a HFD exhibited glucose intolerance, reduced insulin secretion, and insulin sensitivity with a slight decrease in body weight. Consistent with these results, pancreatic islets of mS1KO mice fed a HFD displayed decreased mass with profound apoptotic cell damage and increased macrophage infiltration and inflammation. Liver and adipose tissues from mS1KO HFD mice also showed greater accumulation of macrophages and tissue inflammation. Results from in vitro experiments indicated that deletion of myeloid Sirt1 stimulated proinflammatory M1-like polarization of BMMs and augmented the adipocyte-mediated macrophage chemotaxis. The latter effect was accompanied by increased expression and acetylation of focal adhesion kinase, as well as nuclear factor kappa B. Our results indicate that myeloid SIRT1 plays a crucial role in macrophage polarization and chemotaxis, and thus regulates the development of HFD-induced pancreatic inflammation and insulin secretion, and metabolic derangements in liver and adipose tissue.


Free Radical Biology and Medicine | 2015

Coptidis rhizoma extract protects against cytokine-induced death of pancreatic β-cells through suppression of NF-κB activation

Ji Hye Yang; Kyu Min Kim; Mi Gwang Kim; Kyu Hwa Seo; Jae Yoon Han; Sun-O Ka; Byung-Hyun Park; Sang Mi Shin; Sae Kwang Ku; Il Je Cho; Sung Hwan Ki

Sestrins (Sesns) are conserved antioxidant proteins that accumulate in cells in response to various stresses. However, the regulatory roles of Sesn2 in the immune system and in inflammatory responses remain obscure. In the present study, we investigated whether Sesn2 regulates Toll like receptor (TLR)-mediated inflammatory signaling and sought to identify the molecular mechanism responsible. In cells expressing Sesn2, it was found that Sesn2 almost completely inhibited lipopolysaccharide (LPS)-induced NO release and iNOS expression. A gene knockdown experiment confirmed the role of Sesn2 in LPS-activated RAW264.7 cells. Consistently, proinflammatory cytokine (e.g., TNF-α, IL-6, and IL-1β) release and expression were inhibited in Sesn2-expressing cells. Furthermore, Sesn2 prevented LPS-elicited cell death and ROS production via inhibition of NADPH oxidase. NF-κB and AP-1 are redox-sensitive transcription factors that regulate the expressions of diverse inflammatory genes. Surprisingly, Sesn2 specifically inhibited AP-1 luciferase activity and its DNA binding, but not those of NF-κB. AP-1 inhibition by Sesn2 was found to be due to a lack of JNK, p38, and c-Jun phosphorylation. Next, we investigated whether Sesn2 protects galactosamine (Gal)/LPS-induced liver injury in mice infected with a recombinant adenovirus Sesn2 (Ad-Sesn2). Ad-Sesn2 present less severe hepatic injury as supported by decreases in the ALT, AST, and hepatocyte degeneration. Moreover, Ad-Sesn2 attenuated Gal/LPS-induced proinflammatory gene expression in mice. The study shows that Sesn2 inhibits TLR-induced proinflammatory signaling and protects cells by inhibiting JNK- or p38-mediated c-Jun phosphorylation.


Arthritis & Rheumatism | 2014

Myeloid Sirt1 regulates macrophage infiltration and insulin sensitivity in mice fed a high-fat diet

Hwa-Suk Lee; Seong Ji Woo; Hyoung-Won Koh; Sun-O Ka; Lu Zhou; Kyu Yun Jang; Hye Song Lim; Hyun-Ok Kim; Sang-Il Lee; Byung-Hyun Park

Insulin‐like growth factor binding protein 3 (IGFBP‐3) is known to interfere with the NF‐κB signaling pathway, and it effectively promotes apoptosis in tumor cells by a variety of mechanisms. NF‐κB activation and apoptosis resistance of fibroblast‐like synoviocytes (FLS) play pivotal roles in rheumatoid arthritis (RA). This study was undertaken to evaluate whether IGFBP‐3 has antiarthritic effects.


PLOS ONE | 2014

Role of sestrin2 in the regulation of proinflammatory signaling in macrophages.

Young-Sool Hah; Yun-Hong Cheon; Hye Song Lim; Hee Young Cho; Byung-Hyun Park; Sun-O Ka; Young-Rae Lee; Dong-Won Jeong; Hyun-Ok Kim; Myung-Kwan Han; Sang-Il Lee

Objective SIRT1 modulates the acetylation of the p65 subunit of nuclear factor-κB (NF-κB) and plays a pivotal role in the inflammatory response. This study sought to assess the role of SIRT1 in rheumatoid arthritis (RA) using a myeloid cell-specific SIRT1 knockout (mSIRT1 KO) mouse. Methods mSIRT1 KO mice were generated using the loxP/Cre recombinase system. K/BxN serum transfer arthritis was induced in mSIRT1 KO mice and age-matched littermate loxP control mice. Arthritis severity was assessed by clinical and pathological scoring. The levels of inflammatory cytokines in the serum and joints were measured by ELISA. Migration, M1 polarization, cytokine production, osteoclastogenesis, and p65 acetylation were assessed in bone marrow-derived monocytes/macrophages (BMMs). Results mSIRT1 KO mice showed more severe inflammatory arthritis and aggravated pathological findings than control mice. These effects were paralleled by increases in IL-1, TNF-α, TRAP-positive osteoclasts, and F4/80+ macrophages in the ankles of mSIRT1 KO mice. In addition, BMMs from mSIRT1 KO mice displayed hyperacetylated p65 and increased NF-κB binding activity when compared to control mice, which resulted in increased M1 polarization, migration, pro-inflammatory cytokine production, and osteoclastogenesis. Conclusion Our study provides in vivo evidence that myeloid cell-specific deletion of SIRT1 exacerbates inflammatory arthritis via the hyperactivation of NF-κB signaling, which suggests that SIRT1 activation may be beneficial in the treatment of inflammatory arthritis.


Scientific Reports | 2016

Regulation of Apoptosis and Inflammatory Responses by Insulin‐like Growth Factor Binding Protein 3 in Fibroblast‐like Synoviocytes and Experimental Animal Models of Rheumatoid Arthritis

Mi-Young Song; Jie Wang; Sun-O Ka; Eun Ju Bae; Byung-Hyun Park

Sirtuin 6 (Sirt6), a chromatin associated class III deacetylase, controls whole-body energy homeostasis and has a critical role in glucose-stimulated insulin secretion (GSIS) in pancreatic β cells. However, its underlying molecular mechanism remains poorly understood. To gain further insights, we studied the pathway by which Sirt6 regulates GSIS utilizing mice lacking Sirt6 in their β cells (βS6KO). Further, we overexpressed wild type or deacetylase-inactive mutant Sirt6 in isolated islets as well as in MIN6 cells. We confirmed that βS6KO mice developed glucose intolerance with severely impaired GSIS. Gene expression analysis of knockout islets and overexpression studies demonstrated that Sirt6 deacetylates forkhead box protein O1 (FoxO1) to trigger its nuclear export and releases its transcriptional repression of key glucose sensing genes such as Pdx1 and Glut2. Ectopic overexpression of Sirt6 in knockout islets resulted in rescue of the defective insulin secretion and restoration of the expression of Pdx1 and Glut2. These results show that Sirt6 in pancreatic β cells deacetylates FoxO1 and subsequently increases the expression of Pdx1 and Glut2 to maintain the glucose-sensing ability of pancreatic β cells and systemic glucose tolerance.


Diabetes | 2017

Myeloid Deletion of SIRT1 Aggravates Serum Transfer Arthritis in Mice via Nuclear Factor-κB Activation

Youngyi Lee; Sun-O Ka; Hye-Na Cha; Yu-Na Chae; Mi-Kyung Kim; So-Young Park; Eun Ju Bae; Byung-Hyun Park

Obesity-related insulin resistance is closely associated with macrophage accumulation and subsequent cytokine release in local tissues. Sirtuin 6 (Sirt6) is known to exert an anti-inflammatory function, but its role in macrophages in the context of obesity has not been investigated. We generated myeloid-specific Sirt6 knockout (mS6KO) mice and investigated the metabolic characteristics after high-fat diet (HFD) feeding for 16 weeks. Compared with their wild-type littermates, HFD-fed mS6KO mice exhibited greater increases in body weight, fasting blood glucose and insulin levels, hepatic steatosis, glucose intolerance, and insulin resistance. Gene expression, histology, and flow cytometric analyses demonstrated that liver and adipose tissue inflammation were elevated in HFD-fed mS6KO mice relative to wild type, with a greater accumulation of F4/80+CD11b+CD11c+ adipose tissue macrophages. Myeloid Sirt6 deletion facilitated proinflammatory M1 polarization of bone marrow macrophages and augmented the migration potential of macrophages toward adipose-derived chemoattractants. Mechanistically, Sirt6 deletion in macrophages promoted the activation of nuclear factor-κB (NF-κB) and endogenous production of interleukin-6, which led to STAT3 activation and the positive feedback circuits for NF-κB stimulation; this cross talk expedited an M1 polarization. We conclude that Sirt6 in macrophages is required for the prevention of obesity-associated tissue inflammation and insulin resistance.

Collaboration


Dive into the Sun-O Ka's collaboration.

Top Co-Authors

Avatar

Byung-Hyun Park

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jin-Woo Park

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mi-Young Song

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Kyung-Ah Kim

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Youngyi Lee

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Eun-Kyung Kim

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Hwa-Suk Lee

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

In Hyuk Bang

Chonbuk National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge