Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Lab Invest ; 91(11): 1572-83, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21826057

ABSTRACT

Glutathione transferase kappa (GSTK1-1) is a highly conserved, mitochondrial enzyme potentially involved in redox reactions. GSTK1-1-deficient mice were generated to further study the enzyme's biological role. Reduced and total glutathione levels in liver and kidney were unchanged by GSTK1-1 deficiency and NADPH quinone oxidoreductase 1 expression was not elevated indicating that there is no general underlying oxidative stress in Gstk1(-/-) mice. Electron microscopy of liver and kidney showed no changes in mitochondrial morphology with GSTK1-1 deficiency. The death of a number of Gstk1(-/-) males with urinary tract problems prompted close examination of the kidneys. Electron microscopy revealed glomerular basement membrane changes at 3 months, accompanied by detectable microalbuminuria in male mice (albumin:creatinine ratio of 2.66±0.83 vs 1.13±0.20 mg/mmol for Gstk1(-/-) and wild-type (WT), respectively, P=0.001). This was followed by significant foot process effacement (40-55% vs 10% for Gstk1(-/-) and WT, respectively) at 6 months of age in all Gstk1(-/-) mice examined. Kidney tubules were ultrastructurally normal. Compared with human disease, the Gstk1(-/-) kidneys show changes seen in glomerulopathies causing nephrotic syndrome. Gstk1(-/-) mice may offer insights into the early development of glomerular nephropathies.


Subject(s)
Glomerulonephritis/etiology , Glomerulonephritis/pathology , Glutathione Transferase/deficiency , Albuminuria/etiology , Animals , Blood Chemical Analysis , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Kidney/ultrastructure , Liver/ultrastructure , Male , Mice , Mice, Knockout , Microscopy, Electron , Oxidative Stress/physiology , Urinalysis
2.
Genomics ; 95(5): 299-305, 2010 May.
Article in English | MEDLINE | ID: mdl-20193754

ABSTRACT

The level of glutathione transferase Kappa (GSTK1-1) has been correlated with obesity (Liu et.al. 2008 PNAS 105: 18302-7) and a polymorphism in the hGSTK1 promoter has been associated with insulin secretion and fat deposition (Gao et al 2009 Endocr J 56: 487-94). We searched for additional polymorphisms that may influence GSTK1-1 function or expression. Two SNPs were identified in the 5' non-coding region. A SNP at -1308 that occurs in Chinese subjects is predicted to eliminate a FXR/RXR transcription factor-binding site and causes a 55% increase in transcription rate in HepG2 cells and a 59% decrease in HEK293 cells. These data suggest that the impact of this polymorphism is complex and tissue specific. A SNP at -1032 alters a methylation site and represses transcription by 38%. These observations provide the first functional insight into genetic factors that regulate hGSTK1 expression and may directly influence insulin secretion and fat deposition.


Subject(s)
Gene Expression Regulation , Glutathione Transferase/biosynthesis , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Female , Glutathione Transferase/genetics , Hep G2 Cells , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Male , Obesity/genetics , Obesity/metabolism
3.
J Biol Chem ; 285(13): 9642-9648, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20110353

ABSTRACT

Gamma-glutamylamine cyclotransferase (GGACT) is an enzyme that converts gamma-glutamylamines to free amines and 5-oxoproline. GGACT shows high activity toward gamma-glutamyl-epsilon-lysine, derived from the breakdown of fibrin and other proteins cross-linked by transglutaminases. The enzyme adopts the newly identified cyclotransferase fold, observed in gamma-glutamylcyclotransferase (GGCT), an enzyme with activity toward gamma-glutamyl-alpha-amino acids (Oakley, A. J., Yamada, T., Liu, D., Coggan, M., Clark, A. G., and Board, P. G. (2008) J. Biol. Chem. 283, 22031-22042). Despite the absence of significant sequence identity, several residues are conserved in the active sites of GGCT and GGACT, including a putative catalytic acid/base residue (GGACT Glu(82)). The structure of GGACT in complex with the reaction product 5-oxoproline provides evidence for a common catalytic mechanism in both enzymes. The proposed mechanism, combined with the three-dimensional structures, also explains the different substrate specificities of these enzymes. Despite significant sequence divergence, there are at least three subfamilies in prokaryotes and eukaryotes that have conserved the GGCT fold and GGCT enzymatic activity.


Subject(s)
Dipeptides/chemistry , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , gamma-Glutamylcyclotransferase/chemistry , gamma-Glutamylcyclotransferase/genetics , Amino Acid Sequence , Catalysis , Catalytic Domain , Cell Line, Tumor , Cloning, Molecular , Cross-Linking Reagents/chemistry , Crystallography, X-Ray/methods , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Pyrrolidonecarboxylic Acid/chemistry , Sequence Homology, Amino Acid
4.
J Biol Chem ; 283(32): 22031-42, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18515354

ABSTRACT

The hypothetical protein C7orf24 has been implicated as a cancer marker with a potential role in cell proliferation. We have identified C7orf24 as gamma-glutamyl cyclotransferase (GGCT) that catalyzes the formation of 5-oxoproline (pyroglutamic acid) from gamma-glutamyl dipeptides and potentially plays a significant role in glutathione homeostasis. In the present study we have identified the first cDNA clones encoding a gamma-glutamyl cyclotransferase. The GGCT gene is located on chromosome 7p14-15 and consists of four exons that span 8 kb. The primary sequence is 188 amino acids in length and is unlike any protein of known function. We crystallized functional recombinant gamma-glutamyl cyclotransferase and determined its structure at 1.7 A resolution. The enzyme is a dimer of 20,994-Da subunits. The topology of GGCT is unrelated to other enzymes associated with cyclotransferase-like activity. The fold was originally classified as "BtrG-like," a small family that only includes structures of hypothetical proteins from Mus musculus, Escherichia coli, Pyrococcus horikoshii, and Arabidopsis thaliana. Since this is the first member of this family with a defined function, we propose to refer to this structure as the gamma-glutamyl cyclotransferase fold. We have identified a potential active site pocket that contains a highly conserved glutamic acid (Glu(98)) and propose that it acts as a general acid/base in the reaction mechanism. Mutation of Glu(98) to Ala or Gln completely inactivates the enzyme without altering the overall fold.


Subject(s)
Models, Molecular , gamma-Glutamylcyclotransferase/chemistry , gamma-Glutamylcyclotransferase/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/genetics , Gene Expression Regulation, Enzymologic , Gene Order , Humans , Mice , Molecular Sequence Data , Mutation , Open Reading Frames , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Homology, Nucleic Acid , gamma-Glutamylcyclotransferase/metabolism
5.
Int J Biochem Cell Biol ; 40(11): 2553-9, 2008.
Article in English | MEDLINE | ID: mdl-18571971

ABSTRACT

The Omega class glutathione transferase GSTO1-1 can catalyze the reduction of pentavalent methylated arsenic species and is responsible for the biotransfomation of potentially toxic alpha-haloketones. We investigated the cause of GSTO1-1 deficiency in the T-47D breast cancer cell line and found that the cell line is hemizygous for a polymorphic allele that encodes the deletion of Glu155. Northern and Western blots show that T-47D cells contain GSTO1 mRNA but no GSTO1-1 protein suggesting that the deletion of Glu155 causes GSTO1-1 deficiency in vivo. In further support of this contention we found that lymphoblastoid cell lines from subjects who are heterozygous for the deletion of Glu155 have only 60% of normal activity with the GSTO1-1 specific substrate 4-nitrophenacyl glutathione. Pulse-chase studies showed that the deletion of Glu155 causes increased turnover of GSTO1-1 in T47-D cells. These data establish the fact that the polymorphic deletion of Glu155 can cause GSTO1-1 deficiency in vivo. GSTO1-1 expression is elevated in some cell lines that are resistant to the cytotoxic cancer drugs adriamycin, etoposide and cisplatinum but its specific contribution to multi drug resistance has not been evaluated. In this study GSTO1-1 deficient T47-D cells were used to determine if GSTO1-1 contributes directly to arsenic and drug resistance. We established stable expression of normal GSTO1-1 in T-47D cells and found that this did not alter sensitivity to arsenic trioxide, cisplatinum daunorubicin or etoposide.


Subject(s)
Antineoplastic Agents/metabolism , Arsenicals/metabolism , Cytotoxins/metabolism , Drug Resistance, Neoplasm , Glutamic Acid/metabolism , Glutathione Transferase , Oxides/metabolism , Arsenic Trioxide , Cell Line, Tumor , Drug Screening Assays, Antitumor , Genotype , Glutathione Transferase/deficiency , Glutathione Transferase/genetics , Humans , Polymorphism, Genetic
6.
Int J Biochem Cell Biol ; 40(8): 1616-28, 2008.
Article in English | MEDLINE | ID: mdl-18308613

ABSTRACT

We show that a glutathione transferase (GST) protein, which is recognised by an antibody against the muscle-specific human GSTM2-2 (hGSTM2-2), is associated with the lumen of the sarcoplasmic reticulum (SR) of cardiac muscle, but not skeletal muscle. We further show that hGSTM2-2 modifies both cardiac and skeletal ryanodine receptor (RyR) activity when it binds to the luminal domain of the RyR channel complex. The properties of hGSTM2-2 were compared with those of the calsequestrin (CSQ), a Ca(2+) binding protein also present in the lumen of the SR which, like GSTM2-2, contains a thioredoxin-fold structure and modifies RyR activity (Wei, L., Varsanyi, M., Dulhunty, A. F., Beard, N. A. (2006). The Biophysical Journal, 91, 1288-1301). The glutathione transferase activity of hGSTM2-2 is strong, while CSQ is essentially inactive. Conversely CSQ is a strong Ca(2+) binder, but hGSTM2-2 is not. The effects of luminal hGSTM2-2 on RyR activity differ from those of CSQ in that hGSTM2-2 activates RyRs by increasing their open probability and conductance and the effects are independent of luminal Ca(2+) concentration. The results suggest that GSTM2-2 can interact with specific luminal sites on the RyR complex and that the interaction is likely to be within the pore of the RyR channel. The differences between the effects of CSQ and hGSTM2-2 suggest that the thioredoxin fold is not a major determinant of the luminal actions of either protein. The results indicate that GSTM2-2 is a novel luminal regulator of the RyR channels in the heart.


Subject(s)
Glutathione Transferase/physiology , Myocardium/enzymology , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/enzymology , Animals , Humans , Muscle, Skeletal/enzymology , Protein Folding , Protein Structure, Tertiary , Rabbits , Recombinant Proteins/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Sheep , Thioredoxins/metabolism
7.
Pharmacogenet Genomics ; 18(1): 1-10, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18216717

ABSTRACT

OBJECTIVES AND METHODS: The aim of this study was to investigate genetic variation in glutathione transferase omega 1 (GSTO1-1) in Atacameños, an indigenous population from Chile that has been exposed to environmental arsenic for many generations. GSTO1-1 is thought to catalyse the rate-limiting step in the biotransformation of arsenic in humans and may modulate the response of cancer patients to arsenic trioxide therapy. Allele frequencies were determined by PCR-based methods and a polymorphic variant (GSTO1-1 Val236) was expressed in Escherichia coli and functionally characterized. Urinary arsenic profiles were determined by inductive coupled plasma/mass spectrometry. RESULTS: A novel allele resulting in an Ala236Val substitution that has not been functionally characterized was detected in Atacameños and Chilean participants at a frequency of 0.033 and 0.009, respectively. The Val236 isoenzyme has diminished specific activity (10-20%) with a range of substrates. This loss of activity appears to result from a decrease in the kcat. The Val236 variant is also unstable and rapidly loses activity during purification or when heated at 45 degrees C. The percent of inorganic arsenic in the urine of 205 Chilean participants showed a bimodal distribution that was not associated with the Ala140Asp, Glu155del or Ala236Val polymorphisms in GSTO1-1. CONCLUSION: It is likely that heterozygotes inheriting the Val236 variant subunit would have a partial deficiency of GSTO1-1 activity. Despite their effects on enzyme function the known variants of GSTO1-1 do not appear to explain the observed variability in the excretion of inorganic arsenic.


Subject(s)
Arsenic/toxicity , Environmental Exposure , Glutathione Transferase/genetics , Polymorphism, Genetic , Arsenic/urine , Base Sequence , Chromatography, High Pressure Liquid , DNA Primers , Humans , Mass Spectrometry , Polymerase Chain Reaction
8.
Anal Biochem ; 374(1): 25-30, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18028863

ABSTRACT

Glutathione transferase omega 1-1 (GSTO1-1) catalyzes the biotransformation of arsenic and is implicated as a factor influencing the age-at-onset of Alzheimer's disease and the posttranslational activation of interleukin 1beta (IL-1beta). Investigation of the biological role of GSTO1-1 variants has been hampered by the lack of a specific assay for GSTO1-1 activity in tissue samples that contain other GSTs and other enzymes with similar catalytic specificities. Previous studies (P. G. Board and M. W. Anders, Chem. Res. Toxicol. 20 (2007) 149-154) have shown that GSTO1-1 catalyzes the reduction of S-(phenacyl)glutathiones to acetophenones. A new substrate, S-(4-nitrophenacyl)glutathione (4NPG), has been prepared and found to have a high turnover with GSTO1-1 but negligible activity with GSTO2-2 and other members of the glutathione transferase superfamily. A spectrophotometric assay with 4NPG as a substrate has been used to determine GSTO1-1 activity in several human breast cancer cell lines and in mouse liver and brain tissues.


Subject(s)
Glutathione Transferase/metabolism , Glutathione/analogs & derivatives , Animals , Breast Neoplasms/enzymology , Cell Line, Tumor , Glutathione/metabolism , Humans , Mice , Spectrophotometry, Ultraviolet
9.
Article in English | MEDLINE | ID: mdl-18007051

ABSTRACT

The chloride intracellular channel (CLIC) family of proteins are unusual in that they can exist in either an integral membrane-channel form or a soluble form. Here, the expression, purification, crystallization and preliminary diffraction analysis of CLIC2, one of the least-studied members of this family, are reported. Human CLIC2 was crystallized in two different forms, both in the presence of reduced glutathione and both of which diffracted to better than 1.9 A resolution. Crystal form A displayed P2(1)2(1)2(1) symmetry, with unit-cell parameters a = 44.0, b = 74.7, c = 79.8 A. Crystal form B displayed P2(1) symmetry, with unit-cell parameters a = 36.0, b = 66.9, c = 44.1 A. Structure determination will shed more light on the structure and function of this enigmatic family of proteins.


Subject(s)
Chloride Channels/chemistry , Chloride Channels/biosynthesis , Chloride Channels/isolation & purification , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Humans
10.
J Mol Biol ; 374(3): 719-31, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17945253

ABSTRACT

Chloride intracellular channel (CLIC) proteins possess the remarkable property of being able to convert from a water-soluble state to a membrane channel state. We determined the three-dimensional structure of human CLIC2 in its water-soluble form by X-ray crystallography at 1.8-A resolution from two crystal forms. In contrast to the previously characterized CLIC1 protein, which forms a possibly functionally important disulfide-induced dimer under oxidizing conditions, we show that CLIC2 possesses an intramolecular disulfide and that the protein remains monomeric irrespective of redox conditions. Site-directed mutagenesis studies show that removal of the intramolecular disulfide or introduction of cysteine residues in CLIC2, equivalent to those that form the intramolecular disulfide in CLIC1, does not cause dimer formation under oxidizing conditions. We also show that CLIC2 forms pH-dependent chloride channels in vitro with higher channel activity at low pH levels and that the channels are subject to redox regulation. In both crystal forms, we observed an extended loop region from the C-terminal domain, called the foot loop, inserting itself into an interdomain crevice of a neighboring molecule. The equivalent region in the structurally related glutathione transferase superfamily corresponds to the active site. This so-called foot-in-mouth interaction suggests that CLIC2 might recognize other proteins such as the ryanodine receptor through a similar interaction.


Subject(s)
Chloride Channels/chemistry , Chloride Channels/genetics , Chlorides/chemistry , Chromatography, Gel , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Glutathione/chemistry , Humans , Hydrogen-Ion Concentration , Ion Transport , Models, Molecular , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Conformation , Solubility , Water
11.
Biochem J ; 390(Pt 1): 333-43, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15916532

ABSTRACT

The recently discovered CLIC-2 protein (where CLIC stands for chloride intracellular channel), which belongs to the ubiquitous glutathione transferase structural family and is expressed in the myocardium, is a regulator of native cardiac RyR2 (ryanodine receptor 2) channels. Here we show that recombinant CLIC-2 increases [3H]ryanodine binding to native and purified RyR channels, enhances substate activity in individual channels, increases the number of rare coupled gating events between associated RyRs, and reduces activation of the channels by their primary endogenous cytoplasmic ligands, ATP and Ca2+. CLIC-2 (0.2-10 microM) added to the cytoplasmic side of RyR2 channels in lipid bilayers depressed activity in a reversible, voltage-independent, manner in the presence of activating (10-100 microM) or sub-activating (100 nM) cytoplasmic Ca2+ concentrations. Although the number of channel openings to all levels was reduced, the fraction and duration of openings to substate levels were increased after exposure to CLIC-2. CLIC-2 reduced increases in activity induced by ATP or adenosine 5'-[beta,gamma-imido]triphosphate. Depression of channel activity by CLIC-2 was greater in the presence of 100 microM cytoplasmic Ca2+ than with 100 nM or 10 microM Ca2+. Further, CLIC-2 prevented the usual approximately 50-fold increase in activity when the cytoplasmic Ca2+ concentration was increased from 100 nM to 100 microM. The results show that CLIC-2 interacts with the RyR protein by a mechanism that does not require oxidation, but is influenced by a conserved Cys residue at position 30. CLIC-2 is one of only a few cytosolic inhibitors of cardiac RyR2 channels, and may suppress their activity during diastole and during stress. CLIC-2 provides a unique probe for substate activity, coupled gating and ligand-induced activation of cardiac RyR channels.


Subject(s)
Adenosine Triphosphate/physiology , Calcium Signaling/physiology , Chloride Channels/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Chloride Channels/chemistry , Cytoplasm/physiology , Escherichia coli/metabolism , Humans , Membrane Potentials , Myocardium/cytology , Organisms, Genetically Modified , Protein Binding , Ryanodine/metabolism , Sarcoplasmic Reticulum/physiology , Sheep
12.
Methods Enzymol ; 401: 78-99, 2005.
Article in English | MEDLINE | ID: mdl-16399380

ABSTRACT

The Omega class of cytosolic glutathione transferases was initially recognized by bioinformatic analysis of human sequence databases, and orthologous sequences were subsequently discovered in mouse, rat, pig, Caenorhabditis elegans, Schistosoma mansoni, and Drosophila melanogaster. In humans and mice, two GSTO genes have been recognized and their genetic structures and expression patterns identified. In both species, GSTO1 mRNA is expressed in liver and heart as well as a range of other tissues. GSTO2 is expressed predominantly in the testis, although moderate levels of expression are seen in other tissues. Extensive immunohistochemistry of rat and human tissue sections has demonstrated cellular and subcellular specificity in the expression of GSTO1-1. The crystal structure of recombinant human GSTO1-1 has been determined, and it adopts the canonical GST fold. A cysteine residue in place of the catalytic tyrosine or serine residues found in other GSTs was shown to form a mixed disulfide with glutathione. Omega class GSTs have dehydroascorbate reductase and thioltransferase activities and also catalyze the reduction of monomethylarsonate, an intermediate in the pathway of arsenic biotransformation. Other diverse actions of human GSTO1-1 include modulation of ryanodine receptors and interaction with cytokine release inhibitory drugs. In addition, GSTO1 has been linked to the age at onset of both Alzheimer's and Parkinson's diseases. Several polymorphisms have been identified in the coding regions of the human GSTO1 and GSTO2 genes. Our laboratory has expressed recombinant human GSTO1-1 and GSTO2-2 proteins, as well as a number of polymorphic variants. The expression and purification of these proteins and determination of their enzymatic activity is described.


Subject(s)
Glutathione Transferase , Isoenzymes , Amino Acid Sequence , Animals , Glutaredoxins , Glutathione Transferase/chemistry , Glutathione Transferase/classification , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/classification , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Molecular Sequence Data , Multigene Family , Polymorphism, Genetic , Protein Conformation , Protein Disulfide Reductase (Glutathione)/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Tissue Distribution
13.
Pharmacogenetics ; 14(10): 657-63, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15454730

ABSTRACT

The alpha class glutathione transferase GSTA3-3 is involved in steroid biosynthesis and the metabolism of some xenobiotics. A bioinformatics approach was utilized to identify novel coding region polymorphisms in the glutathione transferase A3 gene (GSTA3). We describe an I71L polymorphism in GSTA3 that occurs at a low frequency in African populations. The activity of the leucine containing isoform was significantly reduced in a range of glutathione-conjugating reactions due to a diminished affinity for reduced glutathione, indicating that this allele could be implicated in disease caused by oxidative stress in steroidogenic tissue. By contrast, the delta(5)-androsten-3,17-dione isomerase activity of GSTA3-3 was not affected by this substitution, indicating that there is no direct effect on steroid synthesis. However, the L71 isoform displayed diminished stability at 45 degrees C. If this relative instability is mirrored in vivo, testosterone and progesterone synthesis may be affected in individuals carrying this allele.


Subject(s)
Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Polymorphism, Genetic , Africa , Amino Acid Substitution , Base Sequence , DNA, Complementary/genetics , Enzyme Stability , Gene Frequency , Genotype , Glutathione Transferase/chemistry , Humans , In Vitro Techniques , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Models, Molecular , Oxidative Stress , Steroids/biosynthesis , Xenobiotics/metabolism
14.
Int J Biochem Cell Biol ; 36(8): 1599-612, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15147738

ABSTRACT

We have examined the biochemical and functional properties of the recently identified, uncharacterised CLIC-2 protein. Sequence alignments showed that CLIC-2 has a high degree of sequence similarity with CLIC-1 and some similarity to the omega class of glutathione transferases (GSTO). A homology model of CLIC-2 based on the crystal structure of CLIC-1 suggests that CLIC-2 belongs to the GST structural family but, unlike the GSTs, CLIC-2 exists as a monomer. It also has an unusual enzyme activity profile. While the CXXC active site motif is conserved between CLIC-2 and the glutaredoxins, no thiol transferase activity was detected. In contrast, low glutathione peroxidase activity was recorded. CLIC-2 was found to be widely distributed in tissues including heart and skeletal muscle. Functional studies showed that CLIC-2 inhibited cardiac ryanodine receptor Ca2+ release channels in lipid bilayers when added to the cytoplasmic side of the channels and inhibited Ca2+ release from cardiac sarcoplasmic reticulum vesicles. The inhibition of RyR channels was reversed by removing CLIC-2 from the solution or by adding an anti-CLIC-2 antibody. The results suggest that one function of CLIC-2 might be to limit Ca2+ release from internal stores in cells.


Subject(s)
Calcium Channel Blockers , Chloride Channels/physiology , Myocardium/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Binding Sites , Calcium/metabolism , Conserved Sequence , Cytoplasm/metabolism , Glutathione Transferase , Humans , Muscle, Skeletal/chemistry , Myocardium/metabolism , Sequence Alignment , Tissue Distribution
15.
Biochem J ; 374(Pt 3): 731-7, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12852784

ABSTRACT

hGSTZ1-1 (human glutathione transferase Zeta 1-1) catalyses a range of glutathione-dependent reactions and plays an important role in the metabolism of tyrosine via its maleylacetoacetate isomerase activity. The crystal structure and sequence alignment of hGSTZ1 with other GSTs (glutathione transferases) focused attention on three highly conserved residues (Ser-14, Ser-15, Cys-16) as candidates for an important role in catalysis. Progress in the investigation of these residues has been limited by the absence of a convenient assay for kinetic analysis. In this study we have developed a new spectrophotometric assay with a novel substrate [(+/-)-2-bromo-3-(4-nitrophenyl)propionic acid]. The assay has been used to rapidly assess the potential catalytic role of several residues in the active site. Despite its less favourable orientation in the crystal structure, Ser-14 was the only residue found to be essential for catalysis. It is proposed that a conformational change may favourably reposition the hydroxyl of Ser-14 during the catalytic cycle. The Cys16-->Ala (Cys-16 mutated to Ala) mutation caused a dramatic increase in the K(m) for glutathione, indicating that Cys-16 plays an important role in the binding and orientation of glutathione in the active site. Previous structural studies implicated Arg-175 in the orientation of alpha-halo acid substrates in the active site of hGSTZ1-1. Mutation of Arg-175 to Lys or Ala resulted in a significant lowering of the kcat in the Ala-175 variant. This result is consistent with the proposal that the charged side chain of Arg-175 forms a salt bridge with the carboxylate of the alpha-halo acid substrates.


Subject(s)
Amino Acids/chemistry , Glutathione Transferase/chemistry , cis-trans-Isomerases/chemistry , Amino Acids/genetics , Binding Sites/genetics , Gas Chromatography-Mass Spectrometry , Glutathione/analogs & derivatives , Glutathione/chemistry , Glutathione/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Glutathione Transferase/physiology , Humans , Mutagenesis, Site-Directed , Spectrophotometry/methods , cis-trans-Isomerases/genetics , cis-trans-Isomerases/metabolism , cis-trans-Isomerases/physiology
16.
Biochem J ; 366(Pt 1): 323-32, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12038961

ABSTRACT

A novel Theta class glutathione transferase (GST) isoenzyme from mouse termed mGSTT3 has been identified by analysis of the expressed sequence tag database. The gene encoding mGSTT3 is clustered with the mGSTT1 and mGSTT2 genes on chromosome 10 and has an exon/intron structure that is similar to that of the other Theta class genes. mGSTT3 is expressed strongly in the liver and to a decreasing extent in the kidney and testis. Recombinant mGSTT3-3 expressed in Escherichia coli had a substrate-specificity profile that differed significantly from that of GSTT1-1 and GSTT2-2 isoenzymes. A molecular model of mGSTT3 suggested that, in comparison with GSTT2, a decrease in volume of the hydrophobic substrate-binding site and the loss of the sulphate-binding pocket prevents its use of the GSTT2 substrate 1-menaphthyl sulphate.


Subject(s)
Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Blotting, Southern , Databases as Topic , Escherichia coli/enzymology , Exons , Glutathione Transferase/chemistry , Introns , Mice , Models, Genetic , Models, Molecular , Molecular Sequence Data , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...