Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
2.
Molecules ; 22(9)2017 Sep 08.
Article in English | MEDLINE | ID: mdl-28885589

ABSTRACT

Glutathione peroxidase (GPx) is an antioxidant protein containing selenium. Owing to the limitations of native GPx, considerable efforts have been made to develop GPx mimics. Here, a short 5-mer peptides (5P) was synthesized and characterized using matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Enzyme coupled assays were used to evaluate GPx activity. The cell viability and apoptosis of H22 cells were tested, and mice bearing H22 cell-derived tumors were used to determine the effects of 5P on tumor inhibition. In comparison with other enzyme models, 5P provided a suitable substrate with proper catalytic site positions, resulting in enhanced catalytic activity. In our mouse model, 5P showed excellent inhibition of tumor growth and improved immunity. In summary, our findings demonstrated the design and synthesis of the small 5P molecule, which inhibited tumor growth and improved immunity. Notably, 5P could inhibit tumor growth without affecting normal growth. Based on these advantages, the novel mimic may have several clinical applications.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Carcinoma, Hepatocellular/drug therapy , Immunity, Innate/drug effects , Liver Neoplasms/drug therapy , Oligopeptides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antioxidants/chemical synthesis , Antioxidants/metabolism , Apoptosis/drug effects , Biocatalysis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Design , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Glutathione Peroxidase/chemistry , Glutathione Peroxidase/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Molecular Mimicry , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Organ Specificity , Phagocytosis/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Treatment Outcome , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/immunology
3.
PLoS One ; 8(5): e63966, 2013.
Article in English | MEDLINE | ID: mdl-23696862

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces cell death in various types of cancer cells but has little or no effects on normal cells. Unfortunately, not all cancer cells respond to TRAIL; therefore, TRAIL sensitizing agents are currently being explored. Here, we reported that 6-(4-N,N-dimethylaminophenyltelluro)-6-deoxy-ß-cyclodextrin (DTCD), a cyclodextrin-derived diorganyl telluride which has been identified as an excellent inhibitor of thioredoxin reductase (TrxR), could sensitize TRAIL resistant human ovarian cancer cells to undergo apoptosis. In vitro, DTCD enhanced TRAIL-induced cytotoxicity in human ovarian cancer cells through up-regulation of DR5. Luciferase analysis and CHIP assays showed that DTCD increased DR5 promoter activity via Sp1 activation. Additionally, DTCD stimulated extracellular signal-regulated kinase (ERK) activation, while the ERK inhibitor PD98059 blocked DTCD-induced DR5 expression and suppressed binding of Sp1 to the DR5 promoter. We further demonstrated that DTCD could induce the release of ASK1 from its complex with Trx-1, and recovered its kinase activity. Meanwhile, suppression of ASK1 by RNA interference led to decreased ERK phosphorylation induced by DTCD. The underlying mechanisms reveal that Trx-1 is heavily oxidized in response to DTCD treatment, in accordance with the fact that DTCD could inhibit the activity of TrxR that reduces oxidized Trx-1. Moreover, using an A2780 xenograft model, DTCD plus TRAIL significantly inhibited the growth of tumor in vivo. Our results suggest that Trx/TrxR system inhibition may play a critical role in apoptosis by combined treatment with DTCD and TRAIL, and raise the possibility that their combination may be a promising strategy for ovarian carcinoma treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclodextrins/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Sp1 Transcription Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Female , Flavonoids/pharmacology , Humans , Phosphorylation/drug effects , Protein Binding/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/drug effects , Thioredoxins/metabolism
4.
Amino Acids ; 44(3): 1009-19, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23224825

ABSTRACT

By combining computational design and site-directed mutagenesis, we have engineered a new catalytic ability into the antibody scFv2F3 by installing a catalytic triad (Trp(29)-Sec(52)-Gln(72)). The resulting abzyme, Se-scFv2F3, exhibits a high glutathione peroxidase (GPx) activity, approaching the native enzyme activity. Activity assays and a systematic computational study were performed to investigate the effect of successive replacement of residues at positions 29, 52, and 72. The results revealed that an active site Ser(52)/Sec substitution is critical for the GPx activity of Se-scFv2F3. In addition, Phe(29)/Trp-Val(72)/Gln mutations enhance the reaction rate via functional cooperation with Sec(52). Molecular dynamics simulations showed that the designed catalytic triad is very stable and the conformational flexibility caused by Tyr(101) occurs mainly in the loop of complementarity determining region 3. The docking studies illustrated the importance of this loop that favors the conformational shift of Tyr(54), Asn(55), and Gly(56) to stabilize substrate binding. Molecular dynamics free energy and molecular mechanics-Poisson Boltzmann surface area calculations estimated the pK(a) shifts of the catalytic residue and the binding free energies of docked complexes, suggesting that dipole-dipole interactions among Trp(29)-Sec(52)-Gln(72) lead to the change of free energy that promotes the residual catalytic activity and the substrate-binding capacity. The calculated results agree well with the experimental data, which should help to clarify why Se-scFv2F3 exhibits high catalytic efficiency.


Subject(s)
Glutathione Peroxidase/chemistry , Glutathione Peroxidase/metabolism , Mutation , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Amino Acid Sequence , Antibodies, Catalytic/chemistry , Antibodies, Catalytic/genetics , Antibodies, Catalytic/metabolism , Catalytic Domain , Glutathione Peroxidase/genetics , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Single-Chain Antibodies/genetics , Thermodynamics
5.
Australas J Dermatol ; 54(2): 120-5, 2013 May.
Article in English | MEDLINE | ID: mdl-23094659

ABSTRACT

BACKGROUND: The generation of harmful reactive oxygen species (ROS) induced by UVB irradiation could induce cell apoptosis and change the cell cycle. 6A,6A'-dicyclohexylamine-6B,6B'-diselenide-bis-ß-cyclodextrin (6-CySeCD) is a novel glutathione peroxidase (GPx; EC 1.11.1.9) mimic. The aim of this study was to investigate the anti-oxidative effects of 6-CySeCD in cultured immortalised human keratinocyte cells (HaCaT). METHODS: HaCaT cells were treated with 30 mJ/cm(2) UVB to establish a damage model. The cultured HaCaT cells were randomly assigned to the control, UVB and treatment groups. The treatment group was incubated with 20 µmol/L of GPx mimics before UVB irradiation. Cell viability was detected by (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, the level of lipid peroxidation was determined by the formation of malondialdehyde (MDA), DNA fragmentation was observed using agarose gel electrophoresis and the levels of intracellular ROS and cell cycle progression were measured by flow cytometry. RESULTS: The levels of cytotoxicity, intracellular ROS, lipid peroxidation and oxidative DNA damage significantly increased after UVB irradiation in the HaCaT cells. UVB irradiation caused pre-G1 -phase arrest in HaCaT cells and significantly reduced the number of HaCaT cells in the S phase. The GPx mimics 6-CySeCD and 2-phenyl-l,2-benzisoselenazol-3(2H)-one (ebselen) significantly blocked UVB-induced apoptosis and changed the cell cycle of the HaCaT cells. The blocked effect of pretreatment 6-CySeCD in UVB-irradiated HaCaT cells was better than that of pretreatment with ebselen. CONCLUSION: 6-CySeCD can relieve the damage induced by UVB irradiation in HaCaT cells.


Subject(s)
Keratinocytes/drug effects , Keratinocytes/radiation effects , Organoselenium Compounds/pharmacology , Radiation-Protective Agents/pharmacology , beta-Cyclodextrins/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Azoles/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/radiation effects , Humans , Isoindoles , Keratinocytes/metabolism , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , Reactive Oxygen Species/metabolism , S Phase Cell Cycle Checkpoints/drug effects , S Phase Cell Cycle Checkpoints/radiation effects
6.
Int J Biochem Cell Biol ; 43(12): 1802-11, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21911079

ABSTRACT

Reactive oxygen species (ROS) are involved in cell growth, differentiation, and death. Excessive amounts of ROS (e.g., O(2)(-), H(2)O(2), and HO) play a role in aging as well as in many human diseases. Superoxide dismutase (SOD) and glutathione peroxidase (GPx) are critical antioxidant enzymes in living organisms. SOD catalyzes the dismutation of O(2)(-) to H(2)O(2), and GPx catalyzes the reduction of H(2)O(2) and other harmful peroxides by glutathione (GSH). They not only function in catalytic processes but also protect each other, resulting in more efficient removal of ROS, protection of cells against injury, and maintenance of the normal metabolism of ROS. To imitate the synergism of SOD and GPx, a 65-mer peptide (65P), containing sequences that form the domains of the active center of SOD and the catalytic triad of GPx upon the incorporation of some metals, was designed on the basis of native enzyme structural models; 65P was expressed in the cysteine auxotrophic expression system to obtain Se-65P. Se-65P was converted into Se-CuZn-65P by incorporating Cu(2+) and Zn(2+). Se-CuZn-65P exhibited high SOD and GPx activities because it has a delicate dual-activity center. The synergism of the enzyme mimic was evaluated by using an in vitro model and a xanthine/xanthine oxidase/Fe(2+)-induced mitochondrial damage model system. We anticipate that the peptide enzyme mimic with synergism is promising for the treatment of human diseases and has potential applications in medicine as a potent antioxidant.


Subject(s)
Glutathione Peroxidase/metabolism , Peptides/chemistry , Superoxide Dismutase/metabolism , Catalytic Domain , Circular Dichroism , Glutathione Peroxidase/chemistry , Mitochondria/metabolism , Models, Molecular , Molecular Mimicry , Peptides/metabolism , Protein Conformation , Reactive Oxygen Species/metabolism , Superoxide Dismutase/chemistry
7.
Eur J Cancer ; 47(12): 1890-907, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21565489

ABSTRACT

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) exhibits potent antitumour activity via membrane receptors on cancer cells without deleterious side-effects for normal tissue. Unfortunately, like many other cancer types, breast cancer cells develop resistance to TRAIL; therefore, TRAIL-sensitising agents are currently being explored. In this study, we report that seleno-cyclodextrin (2-selenium-bridged ß-cyclodextrin, 2-SeCD), a seleno-organic compound with glutathione peroxidase (GPx)-mimetic activity, sensitises TRAIL-resistant human breast cancer cells and xenograft tumours to undergo apoptosis. In vitro, 2-SeCD reduces the viability of cancer cells by inducing cell cycle arrest in G(2)/M phase. Furthermore, 2-SeCD efficiently sensitises MDA-MB-468 and T47D cells but not untransformed human mammary epithelial cells to TRAIL-mediated apoptosis, as evidenced by enhanced caspase activity and poly-ADP-ribose-polymerase (PARP) cleavage. From a mechanistic standpoint, we show that 2-SeCD induces the expression of TRAIL receptors DR5 but not DR4 on both mRNA and protein levels in a dose-dependent manner. Moreover, 2-SeCD treatment also suppresses TRAIL-induced nuclear factor-κB (NF-κB) pro-survival pathways by preventing cytosolic IκBα degradation and p65 nuclear translocation. Consequently, the combined administration suppresses anti-apoptotic proteins transcriptionally regulated by NF-κB. In vivo, 2-SeCD and TRAIL are well tolerated in mice, and their combination significantly inhibits the growth of MDA-MB-468 xenografts and promotes apoptosis. Up-regulation of DR5 and down-regulation of NF-κB by dual treatment were also observed in tumour tissues. Overall, 2-SeCD sensitises resistant breast cancer cells to TRAIL-based apoptosis in vitro and in vivo. These findings provide strong evidence for the therapeutic potential of this combination against breast cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , NF-kappa B/antagonists & inhibitors , Organoselenium Compounds/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , beta-Cyclodextrins/pharmacology , Animals , Blotting, Western , Breast Neoplasms/pathology , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Luciferases/metabolism , Mice , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/metabolism , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , beta-Galactosidase/metabolism
8.
J Inorg Biochem ; 105(2): 283-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21194629

ABSTRACT

Glutathione peroxidase (GPX), superoxide dismutase (SOD) and catalase (CAT) play crucial roles in the metabolism and homeostasis of reactive oxygen species (ROS) in living organisms. From examination of the steady state and pre-steady state kinetic behavior of natural GPX it was found that, in contrast to accepted theories, the affinity of the enzyme for H(2)O(2) rather than reduced glutathione (GSH) most significantly affects its kinetic behavior. Consequently, an enzyme mimic was produced with a similar affinity for the substrate H(2)O(2). A salicylaldehyde Schiff base containing a dimanganese centre was selected as a precursor, because it has high H(2)O(2)-binding affinity for such a relatively small molecule and similar catalytic activity to that of SOD and CAT. Selenium was also incorporated into the catalytic center to provide activity similar to that of GPX, and thus trifunctional enzymatic activity. The K(mH2O2) of the mimic (7.32×10(-2) mM) was found quite close to that of natural enzyme (1.0×10(-2) mM), indicating that the affinity of the mimic to H(2)O(2) was successfully increased to approach natural GPX. The steady state kinetic performance of the enzyme mimic showed that the ratio between k(cat)/K(mGSH) and k(cat)/ K(mH2O2) was quite similar to that of native GPX, indicating that the Mn(III)(2)(L-Se-SO(3)Na) had the same selectivity for both substrates GSH and H(2)O(2) as native GPX, which put it among the best existing GPX mimics. Moreover, the new mimic was confirmed to strongly inhibit lipid peroxidation and mitochondrial swelling, probably due to the synergism between the three antioxidant enzymatic activities.


Subject(s)
Catalase/chemistry , Coordination Complexes/chemical synthesis , Glutathione Peroxidase/chemistry , Manganese , Superoxide Dismutase/chemistry , Animals , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Humans , Hydrogen Peroxide/chemistry , In Vitro Techniques , Lipid Peroxidation/drug effects , Mice , Mitochondrial Swelling/drug effects , Molecular Mimicry , Rabbits
9.
Carcinogenesis ; 32(2): 154-67, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21081474

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exhibits potent antitumor activity via membrane receptors on cancer cells without deleterious side effects for normal tissue. Unfortunately, breast cancer cells, as many other cancer types, develop resistance to TRAIL; therefore, TRAIL sensitizing agents are currently being explored. 2-Tellurium-bridged ß-cyclodextrin (2-TeCD) is a synthetic organotellurium compound, with both glutathione peroxidase-like catalytic ability and thioredoxin reductase inhibitor activity. In the present study, we reported that 2-TeCD sensitized TRAIL-resistant human breast cancer cells and xenograft tumors to undergo apoptosis. In vitro, 2-TeCD efficiently sensitized MDA-MB-468 and T47D cells, but not untransformed human mammary epithelial cells, to TRAIL-mediated apoptosis, as evidenced by enhanced caspase activity and poly (adenosine diphosphate-ribose) polymerase cleavage. From a mechanistic standpoint, we showed that 2-TeCD treatment of breast cancer cells significantly upregulated the messenger RNA and protein levels of TRAIL receptor, death receptor (DR) 5, in a transcription factor Sp1-dependent manner. 2-TeCD treatment also suppressed TRAIL-induced nuclear factor-κB (NF-κB) prosurvival pathways by preventing cytosolic IκBα degradation, as well as p65 nuclear translocation. Consequently, the combined administration suppressed anti-apoptotic molecules that are transcriptionally regulated by NF-κB. In vivo, 2-TeCD and TRAIL were well tolerated in mice and their combination significantly inhibited growth of MDA-MB-468 xenografts and promoted apoptosis. Upregulation of DR5 and downregulation of NF-κB by the dual treatment were also observed in tumor tissues. Overall, 2-TeCD sensitizes resistant breast cancer cells to TRAIL-based apoptosis in vitro and in vivo. These findings provide strong evidence for the therapeutic potential of this combination against breast cancers.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cyclodextrins/pharmacology , Enzyme Inhibitors/pharmacology , NF-kappa B/antagonists & inhibitors , Organometallic Compounds/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/physiology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Breast Neoplasms/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Sp1 Transcription Factor/physiology
10.
Mini Rev Med Chem ; 10(4): 342-56, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20470248

ABSTRACT

The antioxidant enzymes, such as superoxide dismutase, catalase, glutathione peroxidase, and glutathione S-transferase contribute dominatingly to enhance cellular antioxidant defense against oxidative stress. They act cooperatively to scavenge reactive oxygen species, and not one of them can singlehandedly clear all forms of reactive oxygen species. On the basis of the structural understanding for these natural enzymes, many mimics with multifunctional activities had been obtained by chemical synthesis, biosynthesis, and protein fusion techniques. Some of them display remarkable antioxidant cooperative effect in living model which possess potential application in medicine as potent antioxidants. This review summarizes aspects of some multifunctional mimics which have been reported so far.


Subject(s)
Antioxidants/metabolism , Biomimetic Materials/pharmacology , Enzymes/metabolism , Animals , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Drug Synergism , Humans , Molecular Weight , Proteins/chemical synthesis , Proteins/chemistry , Proteins/genetics , Proteins/pharmacology
11.
J Microbiol Biotechnol ; 20(1): 88-93, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20134238

ABSTRACT

Superoxide dismutase (SOD), glutathione peroxidase (GPX) and catalase (CAT) play crucial roles in balancing the production and decomposition of reactive oxygen species (ROS) in living organisms. These enzymes act cooperatively and synergistically to scavenge ROS. In order to imitate the synergism of these enzymes, we designed and synthesized a novel 32-mer peptide (32P) on the basis of the previous 15-mer peptide with GPX activity and a 17-mer peptide with SOD activity. Upon the selenation and chelation of copper, the 32-mer peptide is converted to a new Se- and Cu-containing 32-mer peptide (Se-Cu-32P) and displays both SOD and GPX activities and its kinetics was studied. Moreover, the novel peptide was demonstrated to be able to better protect vero cells from the injury induced by xanthine oxidase (XOD)/xanthine/Fe2+ damage system than its parents. Thus, this bifunctional enzyme imitated the synergism of SOD and GPX and could be a better candidate of therapeutic medicine.


Subject(s)
Glutathione Peroxidase/chemistry , Peptides/chemistry , Superoxide Dismutase/chemistry , Animals , Chlorocebus aethiops , Copper/chemistry , Glutathione Peroxidase/chemical synthesis , Glutathione Peroxidase/pharmacology , Kinetics , Oxidative Stress/drug effects , Peptides/chemical synthesis , Peptides/pharmacology , Selenium/chemistry , Superoxide Dismutase/chemical synthesis , Superoxide Dismutase/pharmacology , Vero Cells
12.
Environ Toxicol Pharmacol ; 30(1): 1-4, 2010 Jul.
Article in English | MEDLINE | ID: mdl-21787621

ABSTRACT

Apoptosis is known to occur after ultraviolet-B (UV-B) radiation. It was found that UV-B could induce cell apoptosis and change cell cycle progression. After exposure to 100J/m(2) of UV-B, pre-G1 phase thymocytes were increased significantly and S phase thymocytes were decreased significantly. UV-B could also induce lipid peroxidation of thymocytes to have their MDA amount increased. These phenomena could be explained by production of reactive oxygen species (ROS), which were induced by UV-B radiation. In this study, we examined the protective effect of dicyclodextrinyl ditelluride (2-TeCD), the glutathione peroxidase (GPX, EC 1.11.1.9) mimics, on thymocytes apoptosis induced by UV-B radiation. The experimental results showed that 2-TeCD protects thymocytes from apoptosis. Moreover, 2-TeCD inhibits lipid peroxidation of thymocytes and displayed great antioxidant ability. Furthermore, 2-TeCD blocks the accumulation of wild-type-p53 (wt-p53) tumor-suppressor gene product caused by UV-B radiation.

13.
Hepatol Res ; 39(11): 1125-36, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19624763

ABSTRACT

AIM: The reperfusion following liver ischemia results in the damage and apoptosis of hepatocytes. The aim of this study was to investigate the possible effects and mechanism of a new synthesized glutathione peroxidase (GPX) mimic, 2-selenium-bridged beta-cyclodextrin (2-SeCD), on rat liver ischemia-reperfusion (I/R) injury. METHODS: Male Wistar rats (n = 32) were randomly divided into four groups: I. sham-operated group, II. I/R group, III. I/R +2-SeCD group, IV. I/R + Ebselen group. Hepatic I/R was administered by 90 min of ischemia and 12 h of reperfusion. Liver tissues were collected at the end of reperfusion period for measurement of various biochemical parameters. RESULTS: The serum aspartate aminotransferase (AST), alanine aminotransferase (ALT) activity and tissue malondialdehyde, myeloperoxidase levels were increased in I/R group, while the increase was significantly reduced by 2-SeCD treatment. The glutathione level, depressed by I/R, was elevated back to normal levels by treatment with 2-SeCD. Severe hepatic damage were observed by light and transmission electron microscopy whilst pretreatment with 2-SeCD resulted in tissue and cellular preservation. Furthermore, 2-SeCD reduced cytochrome c release from mitochondria and subsequent DNA fragmentation by regulating Bcl-2/Bax expression ratio. RESULTS suggested that 2-SeCD was more effective than ebselen in the reversal of the alteration in tissue structural and biochemical parameters caused by I/R injury. CONCLUSION: 2-selenium-bridged beta-cyclodextrin playes an important role in the protection of liver against I/R injury and this treatment may be a novel pharmacological agent for liver surgery.

14.
Int Immunopharmacol ; 9(9): 1087-91, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19439196

ABSTRACT

Adhesion molecules play an important role in the pathogenesis of atherogenesis. They are expressed on endothelial cells surface in response to various inflammatory stimuli. In this paper, we examined the effect of 2-tellurium-bridged beta-cyclodextrin (2-TeCD), a GPx mimic, on the expression of adhesion molecules in human umbilical vein endothelial cells (HUVECs) under tumor necrosis factor-alpha (TNF-alpha) stimulation. Experimental results indicated that 2-TeCD suppressed the TNF-alpha-induced the expression of vascular adhesion molecule-1 (VCAM-1) and intercellular cell adhesion molecule-1 (ICAM-1) on HUVECs surface in a dose-dependent manner. 2-TeCD also reduced the level of mRNA expression of VCAM-1 and ICAM-1. Furthermore, 2-TeCD inhibited THP-1 monocyte adhesion to HUVECs stimulated by TNF-alpha. Nuclear factor-kappaB (NF-kappaB) could regulate transcription of VCAM-1 and ICAM-1 genes. Western blot analysis showed that 2-TeCD inhibited the translocation of the p65 subunit of NF-kappaB into the nucleus. In short, these results indicated that 2-TeCD inhibits TNF-alpha-stimulated VCAM-1 and ICAM-1 expression in HUVECs partly due to suppressing translocation of NF-kappaB.


Subject(s)
Cell Nucleus/metabolism , Cyclodextrins/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/biosynthesis , Organometallic Compounds/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Active Transport, Cell Nucleus , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Line , Cyclodextrins/chemistry , Drug Antagonism , Endothelial Cells/immunology , Endothelial Cells/pathology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Glutathione Peroxidase/chemistry , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Organometallic Compounds/chemistry , Pregnancy , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Umbilical Veins/pathology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
15.
J Mol Recognit ; 22(4): 293-300, 2009.
Article in English | MEDLINE | ID: mdl-19277948

ABSTRACT

Glutathione peroxidase (GPX) is one of the important members of the antioxidant enzyme family. It can catalyze the reduction of hydroperoxides with glutathione to protect cells against oxidative damage. In previous studies, we have prepared the human catalytic antibody Se-scFv-B3 (selenium-containing single-chain Fv fragment of clone B3) with GPX activity by incorporating a catalytic group Sec (selenocysteine) into the binding site using chemical mutation; however, its activity was not very satisfying. In order to try to improve its GPX activity, structural analysis of the scFv-B3 was carried out. A three-dimensional (3D) structure of scFv-B3 was constructed by means of homology modeling and binding site analysis was carried out. Computer-aided docking and energy minimization (EM) calculations of the antibody-GSH (glutathione) complex were also performed. From these simulations, Ala44 and Ala180 in the candidate binding sites were chosen to be mutated to serines respectively, which can be subsequently converted into the catalytic Sec group. The two mutated protein and wild type of the scFv were all expressed in soluble form in Escherichia coli Rosetta and purified by Ni(2+)-immobilized metal affinity chromatography (IMAC), then transformed to selenium-containing catalytic antibody with GPX activity by chemical modification of the reactive serine residues. The GPX activity of the mutated catalytic antibody Se-scFv-B3-A180S was significantly increased compared to the original Se-scFv-B3.


Subject(s)
Antibodies, Catalytic/chemistry , Antibodies, Catalytic/metabolism , Glutathione Peroxidase/metabolism , Immunoglobulin Variable Region/metabolism , Mutagenesis, Site-Directed , Mutation/genetics , Selenium/metabolism , Amino Acid Sequence , Antibodies, Catalytic/genetics , Antibodies, Catalytic/isolation & purification , Binding Sites , Blotting, Western , Clone Cells , Computational Biology , Electrophoresis, Polyacrylamide Gel , Glutathione/chemistry , Glutathione Peroxidase/chemistry , Glutathione Peroxidase/genetics , Humans , Hydrogen Bonding , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/isolation & purification , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Selenocysteine/metabolism , Sequence Alignment , Thermodynamics
16.
Angew Chem Int Ed Engl ; 48(11): 2020-3, 2009.
Article in English | MEDLINE | ID: mdl-19199319

ABSTRACT

A rival to native peroxidase! An existing binding site for glutathione was combined with the catalytic residue tellurocysteine by using an auxotrophic expression system to create an engineered enzyme that functions as a glutathione peroxidase from the scaffold of a glutathione transferase (see picture). The catalytic activity of the telluroenzyme in the reduction of hydroperoxides by glutathione is comparable to that of native glutathione peroxidase.


Subject(s)
Cysteine/analogs & derivatives , Glutathione Peroxidase/metabolism , Glutathione Transferase/chemistry , Catalysis , Cysteine/chemical synthesis , Cysteine/chemistry , Glutathione Transferase/metabolism , Hydrogen Peroxide/chemistry , Oxidation-Reduction
17.
J Mol Recognit ; 21(5): 324-9, 2008.
Article in English | MEDLINE | ID: mdl-18574795

ABSTRACT

In order to generate catalytic antibodies with glutathione peroxidase (GPX) activity, we prepared GSH-S-2,4-dinitrophenyl t-butyl ester (GSH-S-DNPBu) as target antigen. Three clones (A11, B3, and D5) that bound specifically to the antigen were selected from the phage display antibody library (human synthetic VH + VL single-chain Fv fragment (scFv) library). Analysis of PCR products using gel electrophoresis and sequencing showed that only clone B3 beared intact scFv-encoding gene, which was cloned into the expression vector pPELB and expressed as soluble form (scFv-B3) in Escherichia coli Rosetta. The scFv-B3 was purified by Ni(2+)-immobilized metal affinity chromatography (IMAC). The yield of purified proteins was about 2.0-3.0 mg of proteins from 1 L culture. After the active site serines of scFv-B3 were converted into selenocysteines (Secs) with the chemical modification method, we obtained the human catalytic antibody (Se-scFv-B3) with GPX activity of 1288 U/micromol.


Subject(s)
Antibodies, Catalytic/metabolism , Glutathione Peroxidase/metabolism , Antibodies, Catalytic/chemistry , Antibodies, Catalytic/isolation & purification , Catalysis , Drug Evaluation, Preclinical , Glutathione/analogs & derivatives , Glutathione/immunology , Humans , Immunoglobulin Fragments/chemistry , Peptide Library , Selenocysteine/chemistry
18.
Biochim Biophys Acta ; 1780(6): 869-72, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18381079

ABSTRACT

Superoxide dismutase (SOD), glutathione peroxidase (GPX), glutathione S-transferase (GST) and glutathione reductase (GR) play crucial roles in balancing the production and decomposition of reactive oxygen species (ROS) in living organisms. These enzymes act cooperatively and synergistically to scavenge ROS, as not one of them can singlehandedly clear all forms of ROS. In order to imitate the synergy of the enzymes, we designed and generated a recombinant protein, which comprises of a Schistosoma japonicum GST (SjGST) and a bifunctional 35-mer peptide with SOD and GPX activities. The engineered protein demonstrated SOD, GPX and GST activities simultaneously. This trifunctional enzyme with SOD, GPX and GST activities is expected to be the best ROS scavenger.


Subject(s)
Glutathione Peroxidase/genetics , Glutathione Transferase/chemistry , Helminth Proteins/chemistry , Recombinant Fusion Proteins/chemistry , Schistosoma japonicum/enzymology , Superoxide Dismutase/chemistry , Animals , Glutathione Peroxidase/chemistry , Glutathione Transferase/genetics , Helminth Proteins/genetics , Reactive Oxygen Species/chemistry , Recombinant Fusion Proteins/genetics , Schistosoma japonicum/genetics , Superoxide Dismutase/genetics
19.
Int J Biochem Cell Biol ; 40(10): 2090-7, 2008.
Article in English | MEDLINE | ID: mdl-18373941

ABSTRACT

Glutathione peroxidase (GPX) is a critical antioxidant selenoenzyme in organisms that protects cells against oxidative damage by catalyzing the reduction of hydroperoxides by glutathione (GSH). Thus, some GPX mimics have been generated because of their potential therapeutic value. The generation of a semisynthetic selenoenzyme with peroxidase activity, which matches the catalytic efficiencies of naturally evolved GPX, has been a great challenge. Previously, we semisynthesized a GPX mimetic with high catalytic efficiency using a rat theta class glutathione transferase (rGST T2-2) as a scaffold, in which the highly specific GSH-binding site is adjacent to an active site serine residue that can be chemically modified to selenocysteine (Sec). In this study, we have taken advantage of a new scaffold, hGSTZ1-1, in which there are two serine residues in the active site, to achieve both high thiol selectivity and highly catalytic efficiency. The GPX activity of Se-hGSTZ1-1 is about 1.5 times that of rabbit liver GPX, indicating that the selenium content at the active site plays an important role in enhancement of catalytic performance. Kinetic studies revealed that the catalytic mechanism of Se-hGSTZ1-1 belong in a ping-pong mechanism similar to that of the natural GPX.


Subject(s)
Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , Selenium/metabolism , Catalysis/drug effects , Humans , Kinetics , Serine/metabolism , Spectrum Analysis , Tosyl Compounds/pharmacology
20.
Biomacromolecules ; 9(1): 363-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18163571

ABSTRACT

For imitating the active site of antioxidant selenoenzyme glutathione peroxidase (GPx), an artificial enzyme selenosubtilisin was employed as a scaffold for reconstructing substrate glutathione (GSH) specific binding sites by a bioimprinting strategy. GSH was first covalently linked to selenosubtilisin to form a covalent complex GSH-selenosubtilisin through a Se-S bond, then the GSH molecule was used as a template to cast a complementary binding site for substrate GSH recognition. The bioimprinting procedure consists of unfolding the conformation of selenosubtilisin and fixing the new conformation of the complex GSH-selenosubtilisin. Thus a new specificity for naturally occurring GPx substrate GSH was obtained. This bioimprinting procedure facilitates the catalytic selenium moiety of the imprinted selenosubtilisin to match the reactive thiol group of GSH in the GSH binding site, which contributes to acceleration of the intramolecular catalysis. These imprinted selenium-containing proteins exhibited remarkable rate enhancement for the reduction of H2O2 by GSH. The average GPx activity was found to be 462 U/micromol, and it was approximately 100 times that for unimprinted selenosubtilisin. Compared with ebselen, a well-known GPx mimic, an activity enhancement of 500-fold was observed. Detailed steady-state kinetic studies demonstrated that the novel selenoenzyme followed a ping-pong mechanism similar to the naturally occurring GPx.


Subject(s)
Glutathione Peroxidase/metabolism , Molecular Mimicry , Selenium/metabolism , Binding Sites , Catalysis , Electrophoresis, Polyacrylamide Gel , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...