Network


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

Hotspot


Dive into the research topics where Byung Yong Rhim is active.

Publication


Featured researches published by Byung Yong Rhim.


Journal of Neuroscience Research | 2014

Cilostazol suppresses β-amyloid production by activating a disintegrin and metalloproteinase 10 via the upregulation of SIRT1-coupled retinoic acid receptor-β

Hye Rin Lee; Hwa Kyoung Shin; So Youn Park; Hye Young Kim; Won Suk Lee; Byung Yong Rhim; Ki Whan Hong; Chi Dae Kim

The accumulation of plaques of β‐amyloid (Aβ) peptides, a hallmark of Alzheimers disease, results from the sequential cleavage of amyloid precursor protein (APP) by activation of β‐ and γ‐secretases. However, the production of Aβ can be avoided by alternate cleavage of APP by α‐and γ‐secretases. We hypothesized that cilostazol attenuates Aβ production by increasing a disintegrin and metalloproteinase 10 (ADAM10)/α‐secretase activity via SIRT1‐coupled retinoic acid receptor‐β (RARβ) activation in N2a cells expressing human APP Swedish mutation (N2aSwe). To evoke endogenous Aβ overproduction, the culture medium was switched from medium containing 10% fetal bovine serum (FBS) to medium containing 1% FBS, and cells were cultured for 3∼24 hr. After depletion of FBS in media, N2aSwe cells showed increased accumulations of full‐length APP (FL‐APP) and Aβ in a time‐dependent manner (3–24 hr) in association with decreased ADAM10 protein expression. When pretreated with cilostazol (10–30 μM), FL‐APP and Aβ levels were significantly reduced, and ADAM10 and α‐secretase activities were restored. Furthermore, the effect of cilostazol on ADAM10 expression was antagonized by pretreating Rp‐cAMPS and sirtinol and by SIRT1‐gene silencing. In the N2aSwe cells overexpressing the SIRT1 gene, ADAM10, and sAPPα levels were significantly elevated. In addition, like all‐trans retinoic acid, cilostazol enhanced the protein expressions of RARβ and ADAM10, and the cilostazol‐stimulated ADAM10 elevation was significantly attenuated by LE135 (a RARβ inhibitor), sirtinol, and RARβ‐gene silencing. In conclusion, cilostazol suppresses the accumulations of FL‐APP and Aβ by activating ADAM10 via the upregulation of SIRT1‐coupled RARβ.


Arthritis & Rheumatism | 2010

Cilostazol enhances apoptosis of synovial cells from rheumatoid arthritis patients with inhibition of cytokine formation via Nrf2-linked heme oxygenase 1 induction

So Youn Park; Sung Won Lee; Hwa Kyoung Shin; Won Tae Chung; Won Suk Lee; Byung Yong Rhim; Ki Whan Hong; Chi Dae Kim

OBJECTIVE To assess the effects of cilostazol in inhibiting proliferation and enhancing apoptosis in synovial cells from patients with rheumatoid arthritis (RA). METHODS Synovial cell proliferation was measured by MTT assay. The expression of NF-kappaB, IkappaBalpha, Bcl-2, Bax, heme oxygenase 1 (HO-1), and Nrf2 was determined by Western blotting. RESULTS Cilostazol suppressed synovial cell proliferation by arresting the G(2)/M phases of the cell cycle, and this was reversed by KT5720, an inhibitor of protein kinase A. Cilostazol increased the number of TUNEL-positive cells, with increased cytochrome c release and apoptosis-inducing factor translocation as well as increased caspase 3 activation. Cilostazol (10 microM) and cobalt protoporphyrin IX (CoPP) increased HO-1 messenger RNA and protein expression. These effects were suppressed by zinc protoporphyrin IX (ZnPP), an HO-1 inhibitor. Cilostazol and CoPP significantly increased IkappaBalpha in the cytosol and decreased NF-kappaB p65 expression in the nucleus. Increased expression of tumor necrosis factor alpha (TNFalpha), interleukin-1beta (IL-1beta), and IL-6 induced by lipopolysaccharide was attenuated by cilostazol and CoPP, and this was reversed by ZnPP. In mice with collagen-induced arthritis treated with cilostazol (10 and 30 mg/kg/day), paw thickness was decreased with increased apoptotic cells in the joints. In synovial cells transfected with small interfering RNA (siRNA) targeting HO-1, cilostazol did not suppress expression of TNFalpha, IL-1beta, and IL-6, in contrast to findings with negative control cells. Cilostazol- and CoPP-induced HO-1 expression was diminished in cells transfected with Nrf2 siRNA. CONCLUSION Cilostazol suppressed proliferation of synovial cells from RA patients by enhancing apoptosis, and also inhibited cytokine production via mediation of cAMP-dependent protein kinase activation-coupled Nrf2-linked HO-1 expression.


Journal of Pharmacology and Experimental Therapeutics | 2009

Cilostazol Ameliorates Metabolic Abnormalities with Suppression of Proinflammatory Markers in a db/db Mouse Model of Type 2 Diabetes via Activation of Peroxisome Proliferator-Activated Receptor γ Transcription

So Youn Park; Hwa Kyoung Shin; Jeong Hyun Lee; Chi Dae Kim; Won Suk Lee; Byung Yong Rhim; Ki Whan Hong

In a previous study, cilostazol promoted differentiation of 3T3-L1 fibroblasts into adipocytes and improved insulin sensitivity by stimulating peroxisome proliferator-activated receptor (PPAR) γ transcription. This study evaluated the in vivo efficacy of cilostazol to protect a db/db mouse model of type 2 diabetes against altered metabolic abnormalities and proinflammatory markers via activation of PPARγ transcription. Eight-week-old db/db mice were treated with cilostazol or rosiglitazone for 12 days. Cilostazol significantly decreased plasma glucose and triglyceride levels, as did rosiglitazone, a PPARγ agonist. Elevated plasma insulin and resistin levels were significantly decreased by cilostazol, and decreased adiponectin mRNA expression was elevated along with increased plasma adiponectin. Cilostazol significantly increased both adipocyte fatty acid binding protein and fatty acid transport protein-1 mRNA expressions with increased glucose transport 4 in the adipose tissue. Cilostazol and rosiglitazone significantly suppressed proinflammatory markers (superoxide, tumor necrosis factor-α, and vascular cell adhesion molecule-1) in the carotid artery of db/db mice. In an in vitro study with 3T3-L1 fibroblasts, cilostazol significantly increased PPARγ transcription activity, as did rosiglitazone. The transcription activity stimulated by cilostazol was attenuated by KT5720 [(9R,10S,12S)-2,3,9,10,11,12-hexahydro-10-hydroxy-9-methyl-1-oxo-9, 12-epoxy-1H-diindolo[1,2,3-fg:3′,2′,1′-kl]pyrrolo [3,4-I][1,6]-benzodiazocine-10-carboxylic acid hexyl ester], a cAMP-dependent protein kinase inhibitor, and GW9662 (2-chloro-5-nitrobenzanilide), an antagonist of PPARγ activity, indicative of implication of the phosphatidylinositol 3-kinase/Akt signal pathway. These results suggest that cilostazol may improve insulin sensitivity along with anti-inflammatory effects in type 2 diabetic patients via activation of both cAMP-dependent protein kinase and PPARγ transcription.


Journal of Cerebral Blood Flow and Metabolism | 2002

Impairment of autoregulatory vasodilation by NAD(P)H oxidase-dependent superoxide generation during acute stage of subarachnoid hemorrhage in rat pial artery.

Hwa Kyoung Shin; Jeong Hyun Lee; Ki Young Kim; Chi Dae Kim; Won Suk Lee; Byung Yong Rhim; Ki Whan Hong

This study assessed the mechanism(s) by which the autoregulatory vasodilation of rat pial artery in response to acute hypotension during the acute phase of subarachnoid hemorrhage (SAH) was markedly blunted. Increased superoxide production from the cerebral vessels in response to NAD(P)H at 24 hours after SAH +NG-nitro-L-arginine methyl ester (L-NAME) (10 mg/kg) was inhibited by intracisternal administration of a tyrosine kinase inhibitor genistein (10 μmol/L) and Rac inhibitor Clostridium difficile toxin B (1 ng/mL) and a flavoenzyme inhibitor diphenyleneiodonium (10 μmol/L). The expression of gp91phox was enhanced by SAH + L-NAME from 12 to 24 hours, which was inhibited by genistein and toxin B, but not the p22phox. Increased membrane translocation of Rac after SAH + L-NAME was attenuated by both genistein and toxin B, whereas increased tyrosine kinase activity was blocked by genistein, but not by toxin B. The blunted autoregulatory vasodilation to acute hypotension was effectively recovered by genistein and C. difficile toxin B as well as by diphenyleneiodonium. In conclusion, SAH during acute stage causes an increase in NAD(P)H oxidase—dependent superoxide formation in cerebral vessels, which is due to activation of tyrosine phosphorylation-dependent increased expression of gp91phox mRNA and translocation of Rac protein, thereby resulting in a significant reduction of autoregulatory vasodilation.


Journal of Cerebral Blood Flow and Metabolism | 1999

Effect of ω-Conotoxin GVIA and ω-Agatoxin IVA on the Capsaicin-Sensitive Calcitonin Gene-Related Peptide Release and Autoregulatory Vasodilation in Rat Pial Arteries

Ki Whan Hong; Chi Dae Kim; Byung Yong Rhim; Won Suk Lee

This study assesses the effect of neuronal voltage-sensitive Ca2+ channel blockers, ω-conotoxin GVIA (CTX), and ω-agatoxin IVA (AgTX) on the vasodilation and release of calcitonin gene-related peptide (CGRP), both of which were induced by either application of capsaicin or acute stepwise hypotension. Changes in pial arterial diameter were determined directly through a closed cranial window. The vasodilation of pial artery induced by either CGRP (0.1 μmol/L) or capsaicin (0.3 μmol/L) was significantly inhibited by CGRP8–37 (0.1 μmol/L) (P < 0.05 and P < 0.05, respectively). The autoregulatory vasodilation to acute stepwise hypotension was severely attenuated by pretreatment with either CTX or AgTX. When the hypotension was kept for 2, 4, and 10 minutes, the releasable CGRP-like immunoreactivity (CGRP-LI) level (vehicle, 13.4 ± 1.5 fmol/mm2/30 min) by 10 μmol/L capsaicin from the isolated pial arteries was significantly reduced in the 4- and 10-minute hypotension groups (11.3 ± 1.2 fmol/mm2/30 min, P < 0.05, and 11.1 ± 1.5 fmol/mm2/30 min, P < 0.05, respectively), but not in 2-min group. Moreover, the CGRP-LI level released by 10 μmol/L capsaicin (13.7 ± 0.9 fmol/mm2/30 min) also was significantly depressed by pretreatment with 1 μmol/L CTX to 10.4 ± 1.0 fmol/mm2/30 min (P < 0.01) and with 0.1 μmol/L AgTX to 8.7 (1.7 fmol/mm2/30 min (P < 0.001), as well as by pretreatment with 10 μmol/L capsaicin (6.0 ± 1.6 fmol/mm2/30 min, P < 0.001). These results suggest that the neuronal N- and P-type voltage-sensitive Ca2+ channels are implicated in the release of CGRP from capsaicin-sensitive perivascular sensory nerves in response to acute hypotension, and that the released CGRP may contribute to the autoregulatory vasodilation in the cerebral microcirculation.


Brain Research | 2007

Beneficial synergistic effects of concurrent treatment with cilostazol and probucol against focal cerebral ischemic injury in rats.

So Youn Park; Jeong Hyun Lee; Chi Dae Kim; Byung Yong Rhim; Ki Whan Hong; Won Suk Lee

In the present study, we assessed the beneficial synergistic effects of concurrent treatment with low doses of cilostazol and probucol against focal cerebral ischemic infarct in rats. The ischemic infarct induced by 2-h occlusion of middle cerebral artery (MCA) and 22-h reperfusion was significantly reduced in rat brain that received cilostazol (20 mg/kg) and probucol (30 mg/kg) twice together with prominent improvement of neurological function compared to the effect of cilostazol or probucol monotherapy. Increased myeloperoxidase activity, a marker of neutrophil infiltration, observed in the penumbral zone of vehicle-treated brain was more significantly reduced by cilostazol plus probucol in combination. Increased superoxide-, nitrotyrosine (a marker of peroxynitrite)-, poly(ADP-ribose) [a marker for poly(ADP-ribose) polymerase activity]-, and cleaved caspase-3-positive cells (a proapoptotic marker) in the vehicle sample were significantly attenuated by the combination therapy, while individual treatment with low dose of cilostazol or probucol showed a marginal effect. Taken together, it is suggested that the neuroprotective potentials of combination therapy with low doses of cilostazol plus probucol may provide beneficial therapeutic intervention in reducing the focal cerebral ischemic infarct in rats.


Life Sciences | 1993

Disturbances in autoregulatory responses of rat pial arteries by sulfonylureas

Won Suk Lee; Yung Jae Kwon; Sung Suk Yu; Byung Yong Rhim; Ki Whan Hong

It was aimed to test the role of ATP-sensitive K+ channels in the autoregulatory response of cerebral arterioles in vivo. Changes in pial arterial caliber (mean, 43.2 +/- 2.3 microns in diameter) in response to changes in systemic arterial blood pressure (mean, 104.3 +/- 1.4 mmHg) were observed directly through closed cranial windows in anesthetized normotensive rats. During superfusion with vehicle, pial arterial caliber automatically increased in response to hypotension induced by arterial bleeding into a reservoir and decreased on reverse of arterial blood pressure by infusion of blood. After pretreatment with sulfonylureas, glibenclamide (1 and 3 microM) and glipizide (30 and 100 microM), arteriolar dilatation and constriction observed during hypotension and its reverse were disturbed. A similarity was evidenced when hypotension was induced by sodium nitroprusside (750 nmol kg-1min-1, i.v.). Cromakalim, a K+ channel opener, exerted a concentration-dependent vasodilatation of the pial artery and its effect was antagonized by glibenclamide. These data suggest that the endogenous glibenclamide-sensitive K+ channel opener is involved in the modulation of cerebral microvascular autoregulation.


European Journal of Pharmacology | 1986

Enhancement of central and peripheral α1-adrenoceptor sensitivity and reduction of α2-adrenoceptor sensitivity following chronic imipramine treatment in rats

Ki Whan Hong; Byung Yong Rhim; Won Suk Lee

Abstract After chronic imipramine treatment (20 mg/kg i.p., once daily for 14 days) the dose-response curve of the isolated rat anococcygeal muscle to phenylephrine shifted to the left, and furthermore, the -log K A value (affinity) for phenylephrine was significantly increased without affecting the affinity for guanfacine. On the other hand, such treatment caused a shift to the right of the dose-response curve to guanfacine on aortic strips and the affinity of the α-adrenoceptor for guanfacine was lowered without any accompanying changes in the affinity value for phenylephrine. However the relative efficacies of phenylephrine or guanfacine were not influenced by imipramine in either preparation. The ability of phenylephrine to displace [ 3 H]prazosin from its specific binding sites was significantly enhanced after chronic imipramine treatment. These results may indicate that following chronic imipramine treatment the α 1 -adrenoceptors of both central and peripheral tissues responded with supersensitivity to an α 1 -preferential agonist, and the α 2 -adrenoceptors with reduced sensitivity to an α 1 -preferential agonist.


FEBS Letters | 2007

High glucose enhances MMP-2 production in adventitial fibroblasts via Akt1-dependent NF-κB pathway

Seung Jin Lee; Sun Sik Bae; Koan Hoi Kim; Won Suk Lee; Byung Yong Rhim; Ki Whan Hong; Chi Dae Kim

To understand the role of adventitial fibroblasts (AF) in diabetic vascular diseases, the importance of high glucose (HG, 25 mM) on matrix metalloproteinase‐2 (MMP‐2) production in AF was determined. HG enhanced mRNA, protein and gelatinolytic activity of MMP‐2. The enhanced MMP‐2 activity was significantly attenuated not only by a PI3K inhibitor but also by an Akt inhibitor. These HG‐induced MMP‐2 responses were markedly reduced in Akt1‐deficient (1KO) cells. The diminished HG‐induced MMP‐2 responses were completely restored by re‐expression of Akt1. Both the reporter activity and electrophoretic mobility shift assay for activator protein‐1 and nuclear factor‐kappa B (NF‐κB) were enhanced by HG, but NF‐κB were not increased in 1KO cells. Furthermore, HG‐induced MMP‐2 responses were markedly suppressed by NF‐κB decoy oligodeoxynucleotides. Based on these results, it is suggested that HG augments MMP‐2 production via PI3K/Akt1/NF‐κB pathway.


Journal of Neuroscience Research | 2014

Attenuation of β-amyloid-induced tauopathy via activation of CK2α/SIRT1: Targeting for cilostazol

Hye Rin Lee; Hwa Kyoung Shin; So Youn Park; Hye Young Kim; Won Suk Lee; Byung Yong Rhim; Ki Whan Hong; Chi Dae Kim

β‐Amyloid (Aβ) deposits and hyperphosphorylated tau aggregates are the chief hallmarks in the Alzheimers disease (AD) brains, but the strategies for controlling these pathological events remain elusive. We hypothesized that CK2‐coupled SIRT1 activation stimulated by cilostazol suppresses tau acetylation (Ac‐tau) and tau phosphorylation (P‐tau) by inhibiting activation of P300 and GSK3β. Aβ was endogenously overproduced in N2a cells expressing human APP Swedish mutation (N2aSwe) by exposure to medium containing 1% fetal bovine serum for 24 hr. Increased Aβ accumulation was accompanied by increased Ac‐tau and P‐tau levels. Concomitantly, these cells showed increased P300 and GSK3β P‐Tyr216 expression; their expressions were significantly reduced by treatment with cilostazol (3–30 μM) and resveratrol (20 μM). Moreover, decreased expression of SIRT1 and its activity by Aβ were significantly reversed by cilostazol as by resveratrol. In addition, cilostazol strongly stimulated CK2α phosphorylation and its activity, and then stimulated SIRT1 phosphorylation. These effects were confirmed by using the pharmacological inhibitors KT5720 (1 μM, PKA inhibitor), TBCA (20 μM, inhibitor of CK2), and sirtinol (20 μM, SIRT1 inhibitor) as well as by SIRT1 gene silencing and overexpression techniques. In conclusion, increased cAMP‐dependent protein kinase‐linked CK2/SIRT1 expression by cilostazol can be a therapeutic strategy to suppress the tau‐related neurodegeneration in the AD brain.

Collaboration


Dive into the Byung Yong Rhim's collaboration.

Top Co-Authors

Avatar

Ki Whan Hong

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Won Suk Lee

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Chi Dae Kim

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

So Youn Park

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Hwa Kyoung Shin

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Jeong Hyun Lee

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Hye Rin Lee

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Yung Woo Shin

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Hye Young Kim

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Ki Young Kim

Pusan National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge