20 results on '"Chang, Tong-Shin"'
Search Results
2. Inhibitory effect of brazilin on osteoclast differentiation and its mechanism of action
- Author
-
Kim, Jinhee, Lee, Hyo Keun, Chang, Tong-Shin, Kang, Ki Sung, and Hwang, Gwi Seo
- Published
- 2015
- Full Text
- View/download PDF
3. Methods for detection and measurement of hydrogen peroxide inside and outside of cells.
- Author
-
Rhee, Sue, Chang, Tong-Shin, Jeong, Woojin, and Kang, Dongmin
- Abstract
Hydrogen peroxide (HO) is an incompletely reduced metabolite of oxygen that has a diverse array of physiological and pathological effects within living cells depending on the extent, timing, and location of its production. Characterization of the cellular functions of HO requires measurement of its concentration selectively in the presence of other oxygen metabolites and with spatial and temporal fidelity in live cells. For the measurement of HO in biological fluids, several sensitive methods based on horseradish peroxidase and artificial substrates (such as Amplex Red and 3,5,3'5'-tetramethylbenzidine) or on ferrous oxidation in the presence of xylenol orange (FOX) have been developed. For measurement of intracellular HO, methods based on dihydro compounds such as 2',7'-dichlorodihydrofluorescein that fluoresce on oxidation are used widely because of their sensitivity and simplicity. However, such probes react with a variety of cellular oxidants including nitric oxide, peroxynitrite, and hypochloride in addition to HO. Deprotection reaction-based probes (PG1 and PC1) that fluoresce on HO-specific removal of a boronate group rather than on nonspecific oxidation have recently been developed for selective measurement of HO in cells. Furthermore, a new class of organelle-targetable fluorescent probes has been devised by joining PG1 to a substrate of SNAP-tag. Given that SNAP-tag can be genetically targeted to various subcellular organelles, localized accumulation of HO can be monitored with the use of SNAP-tag bioconjugation chemistry. However, given that both dihydro- and deprotection-based probes react irreversibly with HO, they cannot be used to monitor transient changes in HO concentration. This drawback has been overcome with the development of redox-sensitive green fluorescent protein (roGFP) probes, which are prepared by the introduction of two redox-sensitive cysteine residues into green fluorescent protein; the oxidation of these residues to form a disulfide results in a conformational change of the protein and altered fluorogenic properties. Such genetically encoded probes react reversibly with HO and can be targeted to various compartments of the cell, but they are not selective for HO because disulfide formation in roGFP is promoted by various cellular oxidants. A new type of HO-selective, genetically encoded, and reversible fluorescent probe, named HyPer, was recently prepared by insertion of a circularly permuted yellow fluorescent protein (cpYFP) into the bacterial peroxide sensor protein OxyR. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
4. Antiplatelet activity of BRX-018, (6aS,cis)-Malonic acid 3-acetoxy-6a9-bis-(2-methoxycarbonyl-acetoxy)-6,6a,7,11b-tetrahydro-indeno[2,1-c]chromen-10-yl ester methylester
- Author
-
Lee, Gwi-Yeop, Chang, Tong-Shin, Lee, Ki-Seon, Khil, Lee-Yong, Kim, Deukjoon, Chung, Jin-Ho, Kim, Young-Chul, Lee, Byung-Hoon, Moon, Chang-Hyun, and Moon, Chang-Kiu
- Subjects
- *
PHOSPHOLIPASES , *ESTERASES , *BLOOD platelet aggregation , *MALONIC acid - Abstract
Abstract: Brazilin (7,11b-dihydrobenz[b]indeno[1,2-d]pyran-3,6a,9,10 (6H)-tetrol), the major component of Caesalpinia sappan L., was reported to show antiplatelet activity through the inhibition of phospholipase A2 (PLA2) activity and the increase in intracellular free Ca2+ concentration ([Ca2+] i ). To search more potential antiplatelet agent, brazilin derivatives were synthesized and examined for their effects on the platelet aggregation. Among those compounds, BRX-018, (6aS,cis)-Malonic acid 3-acetoxy-6a9-bis-(2-methoxycarbonyl-acetoxy)-6,6a,7,11b-tetrahydro-indeno[2,1-c]chromen-10-yl ester methylester, was confirmed as one of the potential antiplatelet agents. In the present study, we investigated the antiplatelet mechanism of BRX-018. BRX-018 inhibited the thrombin-, collagen-, and ADP-induced rat platelet aggregation in a concentration-dependent manner, with IC50 values of 35, 15, and 25 μM, respectively. BRX-018 also inhibited thrombin-induced dense granule secretion, thromboxane A2 (TXA2) synthesis, and [Ca2+] i elevation in platelets. BRX-018 was also found to inhibit A23187-induced [Ca2+] i and aggregation in the presence of apyrase (ADP scavenger) but not in the presence of both apyrase and indomethacin (a specific inhibitor of cyclooxygenase, COX). Although BRX-018 significantly inhibited arachidonic acid (AA)-induced aggregation and TXA2 synthesis, it had no significant inhibitory effect on cyclooxygenase activity in vitro. In contrast, BRX-018 inhibited the activity of purified PLA2. Dixon plot showed that this inhibition was mixed type with an inhibition constant of K i=23 μM. Taken together, the present study suggests that BRX-018 may be a promising antiplatelet agent and that its antiplatelet activity may be based on the inhibitory mechanisms on TXA2 synthesis in stimulated platelets. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
5. Characterization of Mammalian Sulfiredoxin and Its Reactivation of Hyperoxidized Peroxiredoxin through Reduction of Cysteine Sulfinic Acid in the Active Site to Cysteine.
- Author
-
Chang, Tong-Shin, Jeong, Woojin, Woo, Hyun Ae, Lee, Sun Mi, Park, Sunjoo, and Rhee, Sue Goo
- Subjects
- *
PEROXIDASE , *ENZYMES , *PROTEINS , *METALLOENZYMES , *SURFACE chemistry , *GLUTATHIONE , *PARTICLES (Nuclear physics) , *NUCLEIC acids , *RNA - Abstract
Peroxiredoxins (Prxs) are a family of peroxidases that reduce hydroperoxides. The cysteine residue in the active site of certain eukaryotic Prx enzymes undergoes reversible oxidation to sulfinic acid (Cys-SO2H) during catalysis, and sulfiredoxin (Srx) has been identified as responsible for reversal of the resulting enzyme inactivation in yeast. We have now characterized mammalian orthologs of yeast Srx with an assay based on monitoring of the reduction of sulfinic Prx by immunoblot analysis with antibodies specific for the sulfinic state. Sulfinic reduction by mammalian Srx was found to be a slow process (kcat = 0.18/min) that requires ATP hydrolysis. ATP could be efficiently replaced by GTP, dATP, or dGTP but not by CTP, UTP, dCTP, or dTTP. Both glutathione and thioredoxin are potential physiological electron donors for the Srx reaction, given that their Km values (1.8 nM and 1.2 µM, respectively) are in the range of their intracellular concentrations, and the Vmax values obtained with the two reductants were similar. Although its pKa is relatively low (∼7.3), the active site cysteine of Srx remained reduced even when the active site cysteine of most Prx molecules became oxidized. Finally, depletion of human Srx by RNA interference suggested that Srx is largely responsible for reduction of the Cys-SO2H of Prx in A549 human cells. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
6. The RING-H2–finger protein APC11 as a target of hydrogen peroxide
- Author
-
Chang, Tong-Shin, Jeong, Woojin, Lee, Duck-Yeon, Cho, Chun-Seok, and Rhee, Sue Goo
- Subjects
- *
CELL cycle , *BLOOD agglutination , *ZONE electrophoresis , *KARYOKINESIS - Abstract
The anaphase-promoting complex (APC) is a ubiquitin-protein ligase (E3) that targets cell cycle regulators such as cyclin B and securin for degradation. The APC11 subunit functions as the catalytic core of this complex and mediates the transfer of ubiquitin from a ubiquitin-conjugating enzyme (E2) to the substrate. APC11 contains a RING-H2–finger domain, which includes one histidine and seven cysteine residues that coordinate two Zn2+ ions. We now show that exposure of purified APC11 to H2O2 (0.1 to 1 mM) induced the release of bound zinc as a result of the oxidation of cysteine residues. It also impaired the physical interaction between APC11 and the E2 enzyme Ubc4 as well as inhibited the ubiquitination of cyclin B1 by APC11. The release of HeLa cells from metaphase arrest in the presence of exogenous H2O2 inhibited the ubiquitination of cyclin B1 as well as the degradation of cyclin B1 and securin that were apparent in the absence of H2O2. The presence of H2O2 also blocked the co-immunoprecipitation of Ubc4 with APC11 and delayed the exit of cells from mitosis. Inhibition of APC11 function by H2O2 thus likely contributes to the delay in cell cycle progression through mitosis that is characteristic of cells subjected to oxidative stress. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
7. Cytosolic Peroxiredoxin Attenuates the Activation of JNK and p38 but Potentiates That of ERK in HeLa Cells Stimulated with Tumor Necrosis Factor-α.
- Author
-
Kang, Sang Won, Chang, Tong-Shin, Lee, Tae-Hoon, Kim, Eun Soo, Yu, Dae-Yeul, and Rhee, Seu Goo
- Subjects
- *
PROTEIN kinases , *MITOGENS , *HELA cells , *TUMOR necrosis factors , *REACTIVE oxygen species , *PEROXIDASE - Abstract
Tumor necrosis factor-α (TNF-α) induces the activation of all three types of mitogen-activated protein kinase (MAPK): c-Jun NH[sub 2]-terminal kinase (JNK), p38, and extracellular signal-regulated kinase (ERK). This cytokine also induces the production of several types of reactive oxygen species, including H[sub 2]O[sub 2]. With the use both of HeLa cells expressing wild-type or dominant negative forms of the cytosolic peroxidase peroxiredoxin II and of mouse embryonic fibroblasts deficient in this protein, we evaluated the roles of H202 in the activation of MAPKs by TNF-α. In vitro kinase assays as well as immunoblot analysis with antibodies specific for activated MAPKs indicated that H[sub 2]O[sub 2] produced in response to TNF-α potentiates the activation of JNK and p38 induced by this cytokine but inhibits that of ERK. Our results also suggest that cytosolic peroxiredoxins are important regulators of TNF signaling pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
8. A Crucial Role of SHP-1 Oxidation in RANKL-Induced Osteoclastogenesis
- Author
-
Baek, Jin Young and Chang, Tong Shin
- Published
- 2011
- Full Text
- View/download PDF
9. Collagen-Induced Generation of Hydrogen Peroxide Regulates Platelet Activation Through Oxidation of SHP-2
- Author
-
Jang, Jiyong and Chang, Tong Shin
- Published
- 2011
- Full Text
- View/download PDF
10. NADPH oxidase 1/4 dual inhibitor setanaxib suppresses platelet activation and thrombus formation.
- Author
-
Oh, Eun Bee, Shin, Hye Ji, Yu, Hyunseong, Jang, Joara, Park, Ji Won, and Chang, Tong-Shin
- Subjects
- *
BLOOD platelet activation , *NADPH oxidase , *REACTIVE oxygen species , *CARDIOVASCULAR diseases , *EXTRACELLULAR signal-regulated kinases , *BLOOD platelet aggregation - Abstract
The production of reactive oxygen species (ROS) by NADPH oxidase (NOX) is able to induce platelet activation, making NOX a promising target for antiplatelet therapy. In this study, we examined the effects of setanaxib, a dual NOX1/4 inhibitor, on human platelet function and ROS-related signaling pathways. In collagen-stimulated human platelets, aggregometry, assessment of ROS and Ca2+, immunoblotting, ELISA, flow cytometry, platelet adhesion assay, and assessment of mouse arterial thrombosis were performed in this study. Setanaxib inhibited both intracellular and extracellular ROS production in collagen-activated platelets. Additionally, setanaxib significantly inhibited collagen-induced platelet aggregation, P-selectin exposure from α-granule release, and ATP release from dense granules. Setanaxib blocked the specific tyrosine phosphorylation-mediated activation of Syk, LAT, Vav1, and Btk within collagen receptor signaling pathways, leading to reduced activation of PLCγ2, PKC, and Ca2+ mobilization. Setanaxib also inhibited collagen-induced activation of integrin αIIbβ3, which is linked to increased cGMP levels and VASP phosphorylation. Furthermore, setanaxib suppressed collagen-induced p38 MAPK activation, resulting in decreased phosphorylation of cytosolic PLA 2 and reduced TXA 2 generation. Setanaxib also inhibited ERK5 activation, affecting the exposure of procoagulant phosphatidylserine. Setanaxib reduced thrombus formation under shear conditions by preventing platelet adhesion to collagen. Finally, in vivo administration of setanaxib in animal models led to the inhibition of arterial thrombosis. This study is the first to show that setanaxib suppresses ROS generation, platelet activation, and collagen-induced thrombus formation, suggesting its potential use in treating thrombotic or cardiovascular diseases. • ROS generation by NOX plays a critical role in platelet activation. • Setanaxib is a selective NOX1/4 dual inhibitor. • Setanaxib inhibits ROS generation, platelet activation and thrombus formation. • Setanaxib, potentially therapeutic agent for treating thrombotic or cardiovascular diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Pretreatment with interferon-γ protects microglia from oxidative stress via up-regulation of Mn-SOD
- Author
-
Chen, Xia, Choi, In Young, Chang, Tong-Shin, Noh, You Hyun, Shin, Chan Young, Wu, Chun-Fu, Ko, Kwang Ho, and Kim, Won-Ki
- Subjects
- *
INTERFERONS , *MICROGLIA , *OXIDATIVE stress , *ENZYME regulation , *SUPEROXIDE dismutase , *ANTIOXIDANTS - Abstract
Abstract: Microglial cells, resident macrophage-like immune cells in the brain, are exposed to intense oxidative stress under various pathophysiological conditions. For self-defense against oxidative injuries, microglial cells must be equipped with antioxidative mechanisms. In this study, we investigated the regulation of antioxidant enzyme systems in microglial cells by interferon-γ (IFN-γ) and found that pretreatment with IFN-γ for 20 h protected microglial cells from the toxicity of various reactive species such as hydrogen peroxide (H2O2), superoxide anion, 4-hydroxy-2(E)-nonenal, and peroxynitrite. The cytoprotective effect of IFN-γ pretreatment was abolished by the protein synthesis inhibitor cycloheximide. In addition, treatment of microglial cells with both IFN-γ and H2O2 together did not protect them from the H2O2-evoked toxicity. These results imply that protein synthesis is required for the protection by IFN-γ. Among various antioxidant enzymes such as manganese or copper/zinc superoxide dismutase (Mn-SOD or Cu/Zn-SOD), catalase, and glutathione peroxidase (GPx), only Mn-SOD was up-regulated in IFN-γ-pretreated microglial cells. Transfection with siRNA of Mn-SOD abolished both up-regulation of Mn-SOD expression and protection from H2O2 toxicity by IFN-γ pretreatment. Furthermore, whereas the activities of Mn-SOD and catalase were up-regulated by IFN-γ pretreatment, those of Cu/Zn-SOD and GPx were not. These results indicate that IFN-γ pretreatment protects microglial cells from oxidative stress via selective up-regulation of the level of Mn-SOD and activity of Mn-SOD and catalase. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
12. 2-Cys peroxiredoxin function in intracellular signal transduction: therapeutic implications
- Author
-
Kang, Sang Won, Rhee, Sue Goo, Chang, Tong-Shin, Jeong, Woojin, and Choi, Min Hee
- Subjects
- *
CELLULAR signal transduction , *REACTIVE oxygen species , *CELL receptors , *PEROXIDASE , *THIOREDOXIN - Abstract
H2O2 is a reactive oxygen species that has drawn much interest because of its role as a second messenger in receptor-mediated signaling. Mammalian 2-Cys peroxiredoxins have been shown to eliminate efficiently the H2O2 generated in response to receptor stimulation. 2-Cys peroxiredoxins are members of a novel peroxidase family that catalyze the H2O2 reduction reaction in the presence of thioredoxin, thioredoxin reductase and NADPH. Several lines of evidence suggest that 2-Cys peroxiredoxins have dual roles as regulators of the H2O2 signal and as defenders of oxidative stress. In particular, 2-Cys peroxiredoxin appears to provide selective, specific and localized control of receptor-mediated signal transduction. Thus, the therapeutic potential of 2-Cys peroxiredoxins is clear for diseases, such as cancer and cardiovascular diseases, that involve reactive oxygen species. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
13. Sulfiredoxin inhibitor induces preferential death of cancer cells through reactive oxygen species-mediated mitochondrial damage.
- Author
-
Kim, Hojin, Lee, Gong-Rak, Kim, Jiwon, Baek, Jin Young, Jo, You-Jin, Hong, Seong-Eun, Kim, Sung Hoon, Lee, Jiae, Lee, Hye In, Park, Song-Kyu, Kim, Hwan Mook, Lee, Hwa Jeong, Chang, Tong-Shin, Rhee, Sue Goo, Lee, Ju-Seog, and Jeong, Woojin
- Subjects
- *
SULFIREDOXIN , *CANCER cells , *CELL death , *REACTIVE oxygen species , *MITOCHONDRIAL pathology , *OXIDATIVE stress - Abstract
Recent studies have shown that many types of cancer cells have increased levels of reactive oxygen species (ROS) and enhance antioxidant capacity as an adaptation to intrinsic oxidative stress, suggesting that cancer cells are more vulnerable to oxidative insults and are more dependent on antioxidant systems compared with normal cells. Thus, disruption of redox homeostasis caused by a decline in antioxidant capacity may provide a method for the selective death of cancer cells. Here we show that ROS-mediated selective death of tumor cells can be caused by inhibiting sulfiredoxin (Srx), which reduces hyperoxidized peroxiredoxins, leading to their reactivation. Srx inhibitor increased the accumulation of sulfinic peroxiredoxins and ROS, which led to oxidative mitochondrial damage and caspase activation, resulting in the death of A549 human lung adenocarcinoma cells. Srx depletion also inhibited the growth of A549 cells like Srx inhibition, and the cytotoxic effects of Srx inhibitor were considerably reversed by Srx overexpression or antioxidants such as N-acetyl cysteine and butylated hydroxyanisol. Moreover, Srx inhibitor rendered tumorigenic ovarian cells more susceptible to ROS-mediated death compared with nontumorigenic cells and significantly suppressed the growth of A549 xenografts without acute toxicity. Our results suggest that Srx might serve as a novel therapeutic target for cancer treatment based on ROS-mediated cell death. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
14. Resveratrol inhibits collagen-induced platelet stimulation through suppressing NADPH oxidase and oxidative inactivation of SH2 domain-containing protein tyrosine phosphatase-2.
- Author
-
Jang, Ji Yong, Min, Ji Hyun, Wang, Su Bin, Chae, Yun Hee, Baek, Jin Young, Kim, Myunghee, Ryu, Jae-Sang, and Chang, Tong-Shin
- Subjects
- *
RESVERATROL , *COLLAGEN , *BLOOD platelets , *NADPH oxidase , *PHOSPHATASES , *REACTIVE oxygen species - Abstract
Reactive oxygen species (ROS) produced upon collagen stimulation are implicated in propagating various platelet-activating pathways. Among ROS-producing enzymes, NADPH oxidase (NOX) is largely responsible for collagen receptor-dependent ROS production. Therefore, NOX has been proposed as a novel target for the development of antiplatelet agent. We here investigate whether resveratrol inhibits collagen-induced NOX activation and further examine the effects of resveratrol on ROS-dependent signaling pathways in collagen-stimulated platelets. Collagen-induced superoxide anion production in platelets was inhibited by resveratrol. Resveratrol suppressed collagen-induced phosphorylation of p47 phox , a major regulatory subunit of NOX. Correlated with the inhibitory effects on NOX, resveratrol protected SH2 domain-containing protein tyrosine phosphatase-2 (SHP-2) from ROS-mediated inactivation and subsequently attenuated the specific tyrosine phosphorylation of key components (spleen tyrosine kinase, Vav1, Bruton’s tyrosine kinase, and phospholipase Cγ2) for collagen receptor signaling cascades. Resveratrol also inhibited downstream responses such as cytosolic calcium elevation, P-selectin surface exposure, and integrin-αIIbβ3 activation. Furthermore, resveratrol inhibited platelet aggregation and adhesion in response to collagen. The antiplatelet effects of resveratrol through the inhibition of NOX-derived ROS production and subsequent oxidative inactivation of SHP-2 suggest that resveratrol is a potential compound for prevention and treatment of thrombovascular diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
15. Kaempferol suppresses collagen-induced platelet activation by inhibiting NADPH oxidase and protecting SHP-2 from oxidative inactivation.
- Author
-
Wang, Su Bin, Jang, Ji Yong, Chae, Yun Hee, Min, Ji Hyun, Baek, Jin Young, Kim, Myunghee, Park, Yunjeong, Hwang, Gwi Seo, Ryu, Jae-Sang, and Chang, Tong-Shin
- Subjects
- *
NADPH oxidase , *OXIDATIVE stress , *REACTIVE oxygen species , *PROTEIN-tyrosine phosphatase , *OXIDATION-reduction reaction - Abstract
Reactive oxygen species (ROS) generated upon collagen stimulation act as second messengers to propagate various platelet-activating events. Among the ROS-generating enzymes, NADPH oxidase (NOX) plays a prominent role in platelet activation. Thus, NOX has been suggested as a novel target for anti-platelet drug development. Although kaempferol has been identified as a NOX inhibitor, the influence of kaempferol on the activation of platelets and the underlying mechanism have never been investigated. Here, we studied the effects of kaempferol on NOX activation, ROS-dependent signaling pathways, and functional responses in collagen-stimulated platelets. Superoxide anion generation stimulated by collagen was significantly inhibited by kaempferol in a concentration-dependent manner. More importantly, kaempferol directly bound p47 phox , a major regulatory subunit of NOX, and significantly inhibited collagen-induced phosphorylation of p47 phox and NOX activation. In accordance with the inhibition of NOX, ROS-dependent inactivation of SH2 domain-containing protein tyrosine phosphatase-2 (SHP-2) was potently protected by kaempferol. Subsequently, the specific tyrosine phosphorylation of key components (Syk, Vav1, Btk, and PLCγ2) of collagen receptor signaling pathways was suppressed by kaempferol. Kaempferol also attenuated downstream responses, including cytosolic calcium elevation, P-selectin surface exposure, and integrin-α IIb β 3 activation. Ultimately, kaempferol inhibited platelet aggregation and adhesion in response to collagen in vitro and prolonged in vivo thrombotic response in carotid arteries of mice. This study shows that kaempferol impairs collagen-induced platelet activation through inhibition of NOX-derived ROS production and subsequent oxidative inactivation of SHP-2. This effect suggests that kaempferol has therapeutic potential for the prevention and treatment of thrombovascular diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
16. Sulfiredoxin Translocation into Mitochondria Plays a Crucial Role in Reducing Hyperoxidized Peroxiredoxin III.
- Author
-
Noh, You Hyun, Baek, Jinn Young, Jeong, Woojin, Rhee, Sue Goo, and Chang, Tong-Shin
- Subjects
- *
CHROMOSOMAL translocation , *REACTIVE oxygen species , *MITOCHONDRIA , *APOPTOSIS , *SULFINIC acids , *CYSTEINE proteinases - Abstract
The mitochondria are the major intracellular source of reactive oxygen species (ROS), which are generated during cellular respiration. The role of peroxiredoxin (Prx) III, a 2-Cys Prx family member, in the scavenging of mitochondrial H2O2 has recently been emphasized. While eliminating H2O2, Prx can become overoxidized and inactivated by modifying the active cysteine into cysteine sulfinic acid (Cys-SO2H). When 2-Cys Prxs are inactivated in vitro, sulfiredoxin (Srx) reduces the cysteine sulfinic acid to cysteines. However, whereas Srx is localized in the cytoplasm, Prx III is present exclusively in the mitochondria. Although Srx reduces sulfinic Prx III in vitro, it remains unclear whether the reduction of Prx III in cells is actually mediated by Srx. Our gain- and loss-of-function experiments show that Srx is responsible for reducing not only sulfinic cytosolic Prxs (I and II) but also sulfinic mitochondrial Prx III. We further demonstrate that Srx translocates from the cytosol to mitochondria in response to oxidative stress. Overexpression of mitochondrion-targeted Srx promotes the regeneration of sulfinic Prx III and results in cellular resistance to apoptosis, with enhanced elimination of mitochondrial H2O2 and decreased rates of mitochondrial membrane potential collapse. These results indicate that Srx plays a crucial role in the reactivation of sulfinic mitochondrial Prx III and that its mitochondnal translocation is critical in maintaining the balance between mitochondnial H2O2 production and elimination. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
17. Effects of DK-002, a synthesized (6aS,cis)-9,10-Dimethoxy-7,11b-dihydro-indeno[2,1-c]chromene-3,6a-diol, on platelet activity
- Author
-
Lee, Ki-Seon, Khil, Lee-Yong, Chae, Sang-Ho, Kim, Deukjoon, Lee, Byung-Hoon, Hwang, Gwi-Seo, Moon, Chang-Hyun, Chang, Tong-Shin, and Moon, Chang-Kiu
- Subjects
- *
BLOOD platelet aggregation , *INDOMETHACIN , *PHOSPHOCREATINE , *PROSTAGLANDINS E - Abstract
Abstract: In the present study, the mechanism of antiplatelet activity of DK-002, a synthesized (6aS,cis)-9,10-Dimethoxy-7,11b-dihydro-indeno[2,1-c]chromene-3,6a-diol, was investigated. DK-002 inhibited the thrombin, collagen, and ADP-induced rat platelet aggregation in a concentration-dependent manner, with IC50 values of 120, 27, and 47 μM, respectively. DK-002 also inhibited thrombin-induced dense granule secretion, thromboxane A2 synthesis, and [Ca2+]i elevation in platelets. DK-002 did not show any significant effect on ADP-induced inhibition of cyclic AMP elevation by prostaglandin E1, but DK-002 was confirmed to inhibit ADP-induced [Ca2+]i elevation and shape change. DK-002 inhibited 4-bromo-A23187-induced [Ca2+]i elevation in the presence of creatine phosphate/creatine phosphokinase (CP/CPK, a ADP scavenging system) and indomethacin (a specific inhibitor of cyclooxygenase). DK-002 also inhibited Ca2+ mobilization in thrombin- or 4-bromo-A23187-stimulated platelets through its inhibitory effects on both Ca2+ release from intracellular stores and Ca2+ influx, in the presence of CP/CPK and indomethacin. Taken together, the present study shows that DK-002 has inhibitory effects on stimulation of platelets, and suggests that its antiplatelet activity might be related to the inhibitory mechanism on Ca2+ mobilization in stimulated platelets. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
18. Intracellular messenger function of hydrogen peroxide and its regulation by peroxiredoxins
- Author
-
Rhee, Sue Goo, Kang, Sang Won, Jeong, Woojin, Chang, Tong-Shin, Yang, Kap-Seok, and Woo, Hyun Ae
- Subjects
- *
HYDROGEN peroxide , *PROTEINS , *GROWTH factors , *CYTOKINES , *TYROSINE - Abstract
Hydrogen peroxide (H2O2) accumulates transiently in various cell types stimulated with peptide growth factors and participates in receptor signaling by oxidizing the essential cysteine residues of protein tyrosine phosphatases and the lipid phosphatase PTEN. The reversible inactivation of these phosphatases by H2O2 is likely required to prevent futile cycles of phosphorylation–dephosphorylation of proteins and phosphoinositides. The accumulation of H2O2 is possible even in the presence of large amounts of the antioxidant enzymes peroxiredoxin I and II in the cytosol, probably because of a built-in mechanism of peroxiredoxin inactivation that is mediated by H2O2 and reversed by an ATP-dependent reduction reaction catalyzed by sulfiredoxin. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
19. Tumor necrosis factor α-induced apoptosis in astrocytes is prevented by the activation of P2Y6, but not P2Y4 nucleotide receptors
- Author
-
Kim, Seong G., Soltysiak, Kelly A., Gao, Zhan-Guo, Chang, Tong-Shin, Chung, Eunju, and Jacobson, Kenneth A.
- Subjects
- *
APOPTOSIS , *TUMOR necrosis factors , *G proteins - Abstract
The physiological role of the uracil nucleotide-preferring P2Y6 and P2Y4 receptors is still unclear, although they are widely distributed in various tissues. In an effort to identify their biological functions, we found that activation by UDP of the rat P2Y6 receptor expressed in 1321N1 human astrocytes significantly reduced cell death induced by tumor necrosis factor α (TNFα). This effect of UDP was not observed in non-transfected 1321N1 cells. Activation of the human P2Y4 receptor expressed in 1321N1 cells by UTP did not elicit this protective effect, although both receptors were coupled to phospholipase C. The activation of P2Y6 receptors prevented the activation of both caspase-3 and caspase-8 resulting from TNFα exposure. Even a brief (10-min) incubation with UDP protected the cells against TNFα-induced apoptosis. Interestingly, UDP did not protect the P2Y6-1321N1 cells from death induced by other methods, i.e. oxidative stress induced by hydrogen peroxide and chemical ischemia. Therefore, it is suggested that P2Y6 receptors interact rapidly with the TNFα-related intracellular signals to prevent apoptotic cell death. This is the first study to describe the cellular protective role of P2Y6 nucleotide receptor activation. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
20. Identification of 6′-β-fluoro-homoaristeromycin as a potent inhibitor of chikungunya virus replication.
- Author
-
Shin, Young Sup, Jarhad, Dnyandev B., Jang, Min Hwan, Kovacikova, Kristina, Kim, Gyudong, Yoon, Ji-seong, Kim, Hong-Rae, Hyun, Young Eum, Tipnis, Amol S., Chang, Tong-Shin, van Hemert, Martijn J., and Jeong, Lak Shin
- Subjects
- *
CORONAVIRUSES , *CHIKUNGUNYA virus , *MERS coronavirus , *SARS disease , *VIRUS inhibitors , *VIRAL replication - Abstract
We have reported on aristeromycin (1) and 6′-fluorinated-aristeromycin analogues (2), which are active against RNA viruses such as Middle East respiratory syndrome coronavirus (MERS-CoV), severe acute respiratory syndrome coronavirus (SARS-CoV), Zika virus (ZIKV), and Chikungunya virus (CHIKV). However, these exhibit substantial cytotoxicity. As this cytotoxicity may be attributed to 5′-phosphorylation, we designed and synthesized one-carbon homologated 6′-fluorinated-aristeromycin analogues. This modification prevents 5′-phosphorlyation by cellular kinases, whereas the inhibitory activity towards S -adenosyl- l -homocysteine (SAH) hydrolase will be retained. The enantiomerically pure 6 ′ -fluorinated-5 ′ -homoaristeromycin analogues 3a - e were synthesized via the electrophilic fluorination of the silyl enol ether with Selectfluor, using a base-build up approach as the key steps. All synthesized compounds exhibited potent inhibitory activity towards SAH hydrolase, among which 6 ′ -β-fluoroadenosine analogue 3a was the most potent (IC 50 = 0.36 μM). Among the compounds tested, 6 ′ -β-fluoro-homoaristeromycin 3a showed potent antiviral activity (EC 50 = 0.12 μM) against the CHIKV, without noticeable cytotoxicity up to 250 μM. Only 3a displayed anti-CHIKV activity, whereas both 3a and 3b inhibited SAH hydrolase with similar IC 50 values (0.36 and 0.37 μM, respectively), which suggested that 3a 's antiviral activity did not merely depend on the inhibition of SAH hydrolase. This is further supported by the fact that the antiviral effect was specific for CHIKV and some other alphaviruses and none of the homologated analogues inhibited other RNA viruses, such as SARS-CoV, MERS-CoV, and ZIKV. The potent inhibition and high selectivity index make 6 ′ -β-fluoro-homoaristeromycin (3a) a promising new template for the development of antivirals against CHIKV, a serious re-emerging pathogen that has infected millions of people over the past 15 years. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.