Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(20)2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36293378

ABSTRACT

The SARS-CoV-2 virus can utilize host cell proteases to facilitate cell entry, whereby the Spike (S) protein is cleaved at two specific sites to enable membrane fusion. Furin, transmembrane protease serine 2 (TMPRSS2), and cathepsin L (CatL) are the major proteases implicated, and are thus targets for anti-viral therapy. The human serpin (serine protease inhibitor) alpha-1 antitrypsin (A1AT) shows inhibitory activity for TMPRSS2, and has previously been found to suppress cell infection with SARS-CoV-2. Here, we have generated modified serpin inhibitors with increased specificity for these cellular proteases. Using SerpinB3 (SCCA-1), a cross-class inhibitor of CatL, as a scaffold, we have designed and produced reactive centre loop (RCL) variants to more specifically target both furin and TMPRSS2. Two further variants were generated by substituting the RCL P7-P1 with the spike protein S1/S2 cleavage site from either SARS-CoV-2 alpha or delta (P681R) sequences. Altered inhibitory specificity of purified recombinant proteins was verified in protease assays, with attenuated CatL inhibition and gain of furin or TMPRSS2 inhibition, as predicted, and modified serpins were shown to block S protein cleavage in vitro. Furthermore, the serpin variants were able to inhibit S-pseudoparticle entry into A549-ACE2-TMPRSS2 cells and suppress SARS-CoV-2 replication in Vero E6 cells expressing TMPRSS2. The construct designed to inhibit TMPRSS2 (B3-TMP) was most potent. It was more effective than A1AT for TMPRSS2 enzyme inhibition (with an eighteen-fold improvement in the second order inhibition rate constant) and for blocking SARS-CoV-2 viral replication. These findings advance the potential for serpin RCL mutagenesis to generate new inhibitors, and may lead to novel anti-viral biological molecules.


Subject(s)
COVID-19 Drug Treatment , Serpins , Humans , SARS-CoV-2 , Furin/genetics , Furin/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Serpins/genetics , Serpins/pharmacology , Cathepsin L/metabolism , Angiotensin-Converting Enzyme 2 , Virus Internalization , Antiviral Agents/pharmacology , Mutagenesis , Recombinant Proteins , Serine , Serine Endopeptidases/genetics
2.
Biochim Biophys Acta Proteins Proteom ; 1865(2): 195-200, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27989643

ABSTRACT

SerpinI2/Pancpin/MEPI is a 46kDa member of the serpin (serine protease inhibitor) superfamily. It is downregulated in pancreatic and breast cancer, and associated with acinar cell apoptosis and pancreatic insufficiency when absent in mice. However, the target protease and protein properties of serpinI2 are previously uncharacterised. We have expressed and purified recombinant serpin I2 in E. coli. The protein exhibited thermal instability typical of inhibitory serpins, which was lost following RCL cleavage. SerpinI2 did not inhibit trypsin, but was found to inhibit pancreatic chymotrypsin and elastase with Kass values >105M-1s-1, and with stoichiometry of inhibition of 1.4 and 1.7 respectively. Mutagenesis of the predicted critical hinge region residue Ser344 abolished inhibitory activity, and a cleavage site C-terminal to Met358 was identified. The protein is also prone to polymerisation/aggregation at 45°C, a characteristic of serpins associated with disease. This study therefore reveals a function for serpinI2 and supports the hypothesis that this protein can protect pancreatic cells from prematurely activated zymogens.


Subject(s)
Chymotrypsin/antagonists & inhibitors , Pancreatic Elastase/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Serpins/pharmacology , Amino Acid Sequence , Cell Line , Escherichia coli/metabolism , Neoplasm Proteins/pharmacology , Recombinant Proteins/pharmacology , Sequence Homology, Amino Acid , Somatostatin-Secreting Cells/drug effects , Somatostatin-Secreting Cells/metabolism , Substrate Specificity , Trypsin/metabolism
3.
PLoS One ; 6(10): e26234, 2011.
Article in English | MEDLINE | ID: mdl-22028839

ABSTRACT

PEDF (Pigment epithelium-derived factor) is a non-inhibitory member of the serpin gene family (serpinF1) that displays neurotrophic and anti-angiogenic properties. PEDF contains a secretion signal sequence, but although originally regarded as a secreted extracellular protein, endogenous PEDF is found in the cytoplasm and nucleus of several mammalian cell types. In this study we employed a yeast two-hybrid interaction trap screen to identify transportin-SR2, a member of the importin-ß family of nuclear transport karyopherins, as a putative PEDF binding partner. The interaction was supported in vitro by GST-pulldown and co-immunoprecipitation. Following transfection of HEK293 cells with GFP-tagged PEDF the protein was predominantly localised to the nucleus, suggesting that active import of PEDF occurs. A motif (YxxYRVRS) shared by PEDF and the unrelated transportin-SR2 substrate, RNA binding motif protein 4b, was identified and we investigated its potential as a nuclear localization signal (NLS) sequence. Site-directed mutagenesis of this helix A motif in PEDF resulted in a GFP-tagged mutant protein being excluded from the nucleus, and mutation of two arginine residues (R67, R69) was sufficient to abolish nuclear import and PEDF interaction with transportin-SR2. These results suggest a novel NLS and mechanism for serpinF1 nuclear import, which may be critical for anti-angiogenic and neurotrophic function.


Subject(s)
Cell Nucleus/metabolism , Eye Proteins/chemistry , Eye Proteins/metabolism , Nerve Growth Factors/chemistry , Nerve Growth Factors/metabolism , Nuclear Localization Signals , Serpins/chemistry , Serpins/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Amino Acid Motifs , Amino Acid Sequence , Eye Proteins/genetics , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nerve Growth Factors/genetics , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Secondary , Serpins/genetics , Two-Hybrid System Techniques
4.
PLoS One ; 6(8): e22645, 2011.
Article in English | MEDLINE | ID: mdl-21857942

ABSTRACT

Granzyme-mediated cell death is the major pathway for cytotoxic lymphocytes to kill virus-infected and tumor cells. In humans, five different granzymes (i.e. GrA, GrB, GrH, GrK, and GrM) are known that all induce cell death. Expression of intracellular serine protease inhibitors (serpins) is one of the mechanisms by which tumor cells evade cytotoxic lymphocyte-mediated killing. Intracellular expression of SERPINB9 by tumor cells renders them resistant to GrB-induced apoptosis. In contrast to GrB, however, no physiological intracellular inhibitors are known for the other four human granzymes. In the present study, we show that SERPINB4 formed a typical serpin-protease SDS-stable complex with both recombinant and native human GrM. Mutation of the P2-P1-P1' triplet in the SERPINB4 reactive center loop completely abolished complex formation with GrM and N-terminal sequencing revealed that GrM cleaves SERPINB4 after P1-Leu. SERPINB4 inhibited GrM activity with a stoichiometry of inhibition of 1.6 and an apparent second order rate constant of 1.3×10(4) M(-1) s(-1). SERPINB4 abolished cleavage of the macromolecular GrM substrates α-tubulin and nucleophosmin. Overexpression of SERPINB4 in tumor cells inhibited recombinant GrM-induced as well as NK cell-mediated cell death and this inhibition depended on the reactive center loop of the serpin. As SERPINB4 is highly expressed by squamous cell carcinomas, our results may represent a novel mechanism by which these tumor cells evade cytotoxic lymphocyte-induced GrM-mediated cell death.


Subject(s)
Antigens, Neoplasm/metabolism , Granzymes/metabolism , Intracellular Space/enzymology , Serpins/metabolism , Antigens, Neoplasm/genetics , Cell Death/immunology , Cytotoxicity, Immunologic , Granzymes/genetics , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Jurkat Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Kinetics , Mutation , Protein Binding , Recombinant Proteins/metabolism , Serpins/genetics , Substrate Specificity , Transfection
5.
J Biol Chem ; 285(6): 3722-3729, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19959474

ABSTRACT

The glycosaminoglycan heparin is known to possess antimetastatic activity in experimental models and preclinical studies, but there is still uncertainty over its mechanism of action in this respect. As an anticoagulant, heparin enhances inhibition of thrombin by the serpin antithrombin III, but a similar cofactor role has not been previously investigated for proteases linked to metastasis. The squamous cell carcinoma antigens (serpins B3 and B4) are tumor-associated proteins that can inhibit papain-like cysteine proteases, including cathepsins L, K, and S. In this study, we show that SCCA-1 (B3) and SCCA-2 (B4) can bind heparin as demonstrated by affinity chromatography, native PAGE gel shifts, and intrinsic fluorescence quenching. Binding was specific for heparin and heparan sulfate but not other glycosaminoglycans. The presence of heparin accelerated inhibition of cathepsin L by both serpins, and in the case of SCCA-1, heparin increased the second order inhibition rate constant from 5.4 x 10(5) to >10(8), indicating a rate enhancement of at least 180-fold. A templating mechanism was shown, consistent with ternary complex formation. Furthermore, SCCA-1 inhibition of cathepsin L-like proteolytic activity secreted from breast and melanoma cancer cell lines was significantly enhanced by heparin. This is the first example of glycosaminoglycan enhancement of B-clade serpin activity and the first report of heparin acting as a cofactor in serpin cross-class inhibition of cysteine proteases. Most importantly, this finding raises the possibility that the anticancer properties of heparin may be due, at least partly, to enhanced inhibition of prometastatic proteases.


Subject(s)
Antigens, Neoplasm/metabolism , Cathepsin L/metabolism , Heparin/metabolism , Serpins/metabolism , Anticoagulants/metabolism , Anticoagulants/pharmacology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/pharmacology , Catalysis/drug effects , Cathepsin G/metabolism , Cathepsin L/antagonists & inhibitors , Cell Line, Tumor , Chromatography, Affinity , Chymases/metabolism , Drug Synergism , Electrophoresis, Polyacrylamide Gel , Glycosaminoglycans/metabolism , Heparin/pharmacology , Humans , Kinetics , Mast Cells/enzymology , Protein Binding , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/pharmacology , Serpins/genetics , Serpins/pharmacology , Spectrometry, Fluorescence
6.
J Biol Chem ; 280(23): 22356-64, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15760906

ABSTRACT

Maspin is a serpin that acts as a tumor suppressor in a range of human cancers, including tumors of the breast and lung. Maspin is crucial for development, because homozygous loss of the gene is lethal; however, the precise physiological role of the molecule is unclear. To gain insight into the function of human maspin, we have determined its crystal structure in two similar, but non-isomorphous crystal forms, to 2.1- and 2.8-A resolution, respectively. The structure reveals that maspin adopts the native serpin fold in which the reactive center loop is expelled fully from the A beta-sheet, makes minimal contacts with the core of the molecule, and exhibits a high degree of flexibility. A buried salt bridge unique to maspin orthologues causes an unusual bulge in the region around the D and E alpha-helices, an area of the molecule demonstrated in other serpins to be important for cofactor recognition. Strikingly, the structural data reveal that maspin is able to undergo conformational change in and around the G alpha-helix, switching between an open and a closed form. This change dictates the electrostatic character of a putative cofactor binding surface and highlights this region as a likely determinant of maspin function. The high resolution crystal structure of maspin provides a detailed molecular framework to elucidate the mechanism of function of this important tumor suppressor.


Subject(s)
Crystallography, X-Ray/methods , Serpins/chemistry , Animals , Binding Sites , Cathepsin L , Cathepsins/chemistry , Chickens , Circular Dichroism , Cysteine Endopeptidases/chemistry , Extracellular Matrix , Genes, Tumor Suppressor , Homozygote , Humans , Mice , Models, Molecular , Plasmids/metabolism , Protein Binding , Protein Conformation , Protein Structure, Secondary , Rats , Recombinant Proteins/chemistry , Spectrophotometry , Static Electricity , Temperature , Xenopus
7.
Bioinformatics ; 20(17): 3108-27, 2004 Nov 22.
Article in English | MEDLINE | ID: mdl-15217816

ABSTRACT

MOTIVATION: Microarrays are becoming an increasingly common tool for observing changes in gene expression over a large cross section of the genome. This experimental tool is particularly valuable for understanding the genome-wide changes in gene transcription in response to thiazolidinedione (TZD) treatment. The TZD class of drugs is known to improve insulin-sensitivity in diabetic patients, and is clinically used in treatment regimens. In cells, TZDs bind to and activate the transcriptional activity of peroxisome proliferator-activated receptor gamma (PPAR-gamma). Large-scale array analyses will provide some insight into the mechanisms of TZD-mediated insulin sensitization. Unfortunately, a theoretical basis for analyzing array data has not kept pace with the rapid adoption of this tool. The methods that are commonly used, particularly the fold-change approach and the standard t-test, either lack statistical rigor or resort to generalized statistical models that do not accurately estimate variability at low replicate numbers. RESULTS: We introduce a statistical framework that models the dependence of measurement variance on the level of gene expression in the context of a Bayesian hierarchical model. We compare several methods of parameter estimation and subsequently apply these to determine a set of genes in 3T3-L1 adipocytes that are differentially regulated in response to TZD treatment. When the number of experimental replicates is low (n = 2-3), this approach appears to qualitatively preserve an equivalent degree of specificity, while vastly improving sensitivity over other comparable methods. In addition, the statistical framework developed here can be readily applied to understand the implicit assumptions made in traditional fold-change approaches to array analysis.


Subject(s)
3T3-L1 Cells/metabolism , Algorithms , Gene Expression Profiling/methods , Gene Expression Regulation/physiology , Models, Biological , Oligonucleotide Array Sequence Analysis/methods , Transcription Factors/metabolism , 3T3-L1 Cells/drug effects , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Dimethyl Sulfoxide/pharmacology , Gene Expression Regulation/drug effects , Genetic Variation , Mice , Models, Statistical , Thiazolidinediones/pharmacology
10.
Methods ; 32(2): 169-76, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14698629

ABSTRACT

Expression systems based on Escherichia coli offer fast, cheap, and convenient means for the production of recombinant serpins. Over 30 active serpins from prokaryotic and eukaryotic organisms have been produced in this way, using a variety of vectors, promoters, fusion partners, and host strains. Serpins forming insoluble inclusion bodies in E. coli can generally be solubilized and refolded. Here, we outline the general approaches and procedures to be considered when contemplating the use of E. coli for recombinant serpin production.


Subject(s)
Escherichia coli/genetics , Recombinant Proteins/biosynthesis , Serpins/biosynthesis , Bacteriolysis , Endopeptidases/metabolism , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/genetics , Protein Denaturation , Protein Folding , RNA Stability , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Serpins/chemistry , Serpins/genetics , Solubility
11.
Biochem J ; 365(Pt 1): 13-8, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-11994049

ABSTRACT

The final two enzymes in the CoA biosynthetic pathway, phosphopantetheine adenylyltransferase (PPAT; EC 2.7.7.3) and dephospho-CoA kinase (DPCK; EC 2.7.1.24), are separate proteins in prokaryotes, but exist as a bifunctional enzyme in pig liver. In the present study we have obtained sequence information from purified pig-liver enzyme, and identified the corresponding cDNA in a number of species. The human gene localizes to chromosome 17q12-21 and contains regions with sequence similarity to the monofunctional Escherichia coli DPCK and PPAT. The recombinant 564-amino-acid human protein confirmed the associated transferase and kinase activities, and gave similar kinetic properties to the wild-type pig enzyme.


Subject(s)
Nucleotidyltransferases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 17/genetics , Cloning, Molecular , DNA, Complementary/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression , Humans , Kinetics , Liver/enzymology , Molecular Sequence Data , Nucleotidyltransferases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Swine
12.
Biochemistry ; 41(15): 4998-5004, 2002 Apr 16.
Article in English | MEDLINE | ID: mdl-11939796

ABSTRACT

The closely related serpins squamous cell carcinoma antigen-1 and -2 (SCCA-1 and -2, respectively) are capable of inhibiting cysteine proteases of the papain superfamily. To ascertain whether the ability to inhibit cysteine proteases is an intrinsic property of serpins in general, the reactive center loop (RCL) of the archetypal serine protease inhibitor alpha(1)-antitrypsin was replaced with that of SCCA-1. It was found that this simple substitution could convert alpha(1)-antitrypsin into a cysteine protease inhibitor, albeit an inefficient one. The RCL of SCCA-1 is three residues longer than that of alpha(1)-antitrypsin, and therefore, the effect of loop length on the cysteine protease inhibitory activity was investigated. Mutants in which the RCL was shortened by one, two, or three residues were effective inhibitors with second-order rate constants of 10(5)-10(7) M(-)(1) s(-)(1). In addition to loop length, the identity of the cysteine protease was of considerable importance, since the chimeric molecules inhibited cathepsins L, V, and K efficiently, but not papain or cathepsin B. By testing complexes between an RCL-mimicking peptide and the mutants, it was found that the formation of a stable serpin-cysteine protease complex and the inhibition of a cysteine protease were both critically dependent on RCL insertion. The results strongly indicate that the serpin body is intrinsically capable of supporting cysteine protease inhibition, and that the complex with a papain-like cysteine protease would be expected to be analogous to that seen with serine proteases.


Subject(s)
Cathepsins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Papain/antagonists & inhibitors , Serpins/chemistry , Serpins/pharmacology , alpha 1-Antitrypsin/chemistry , Amino Acid Sequence , Antigens, Neoplasm/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Structure-Activity Relationship
13.
J Biol Chem ; 277(13): 10783-8, 2002 Mar 29.
Article in English | MEDLINE | ID: mdl-11788595

ABSTRACT

Maspin (mammary serine protease inhibitor) was originally identified as a tumor suppressor protein in human breast epithelial cells and is a member of the serine proteases inhibitor (serpin) superfamily. It inhibits tumor cell motility and angiogenesis, and although predominantly cytoplasmic, it is also localized to the cell surface. In this study we have investigated the use of the yeast two-hybrid interaction trap to identify novel maspin targets. A target human fibroblast cDNA library was screened, and the alpha-2 chain of type I collagen was identified as a potential interactant. Binding studies with isolated proteins showed interaction between recombinant maspin and types I and III collagen but not other collagen subtypes, a profile strikingly similar to mouse pigment epithelium-derived factor (caspin), which is similarly down-regulated in murine adenocarcinoma tumors and is a potent inhibitor of angiogenesis. Kinetic analysis using an IAsys resonant mirror biosensor determined the dissociation constant of maspin for collagen type I to be 0.63 microm. Further two-hybrid interactions with maspin truncation constructs suggest that collagen binding is localized to amino acids 84-112 of maspin, which aligns with the collagen-binding region of colligin. A direct interaction between exogenous or cell surface maspin and extracellular matrix collagen may contribute to a cell adhesion role in the prevention of tumor cell migration and angiogenesis.


Subject(s)
Collagen Type III/metabolism , Collagen Type I/metabolism , Proteins/metabolism , Serpins/metabolism , Base Sequence , Binding Sites , Cloning, Molecular , DNA Primers , Genes, Tumor Suppressor , HeLa Cells , Humans , Kinetics , Polymerase Chain Reaction , Serpins/genetics , Two-Hybrid System Techniques
14.
FEBS Lett ; 508(3): 313-7, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11728442

ABSTRACT

In Drosophila, the Toll family of proteins mediates the innate immune response. Toll is activated by Spaetzle, which is generated in response to pathogens via a serine protease cascade. We wished to investigate if lipopolysaccharides (LPS) might activate Toll-like receptor (TLR) 4 via a serine protease in humans. The serpin antithrombin III (ATIII) and the thrombin inhibitor hirudin both inhibited nuclear factor (NF)-kappaB activation by LPS and Lipid A. ATIII and hirudin were also able to inhibit LPS-induced NF-kappaB activation in cells stably transfected with TLR4. These results suggest that LPS may activate a mammalian serine protease, which generates a product required for TLR4 signalling.


Subject(s)
Antithrombin III/pharmacology , Drosophila Proteins , Membrane Glycoproteins/metabolism , NF-kappa B/metabolism , Receptors, Cell Surface/metabolism , Cell Line , Enzyme Activation , Hirudins/pharmacology , Humans , Lipid A/pharmacology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/genetics , Monocytes , Receptors, Cell Surface/genetics , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Signal Transduction , Toll-Like Receptor 4 , Toll-Like Receptors
15.
Proc Natl Acad Sci U S A ; 98(24): 13514-8, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11707596

ABSTRACT

We have developed a methodology of prodrug delivery by using a modified insulin species whose biological activity potentially can be regulated in vivo. Native insulin was derivatized with aldol-terminated chemical modifications that can be selectively removed by the catalytic aldolase antibody 38C2 under physiologic conditions. The derivatized organoinsulin (insulin(D)) was defective with respect to receptor binding and stimulation of glucose transport. The affinity of insulin(D) for the insulin receptor was reduced by 90% in binding studies using intact cells. The ability of insulin(D) to stimulate glucose transport was reduced by 96% in 3T3-L1 adipocytes and by 55% in conscious rats. Incubation of insulin(D) with the catalytic aldolase antibody 38C2 cleaved all of the aldol-terminated modifications, restoring native insulin. Treatment of insulin(D) with 38C2 also restored insulin(D)'s receptor binding and glucose transport-stimulating activities in vitro, as well as its ability to lower glucose levels in animals in vivo. We propose that these results are the foundation for an in vivo regulated system of insulin activation using the prohormone insulin(D) and catalytic antibody 38C2 with potential therapeutic application.


Subject(s)
Antibodies, Catalytic/metabolism , Fructose-Bisphosphate Aldolase/metabolism , Immunoglobulin Fab Fragments/metabolism , Insulin/metabolism , Protein Precursors/metabolism , 3T3 Cells , Actins/metabolism , Animals , Catalysis , Cell Line , Glucose/metabolism , Humans , Insulin/biosynthesis , Male , Mice , Protein Precursors/biosynthesis , Rats , Rats, Wistar , Receptor, Insulin/metabolism
16.
Eur J Biochem ; 268(22): 5868-75, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11722574

ABSTRACT

The squamous cell carcinoma antigens, SCCA1 and SCCA2, are members of the serine protease inhibitors (serpin) superfamily and are transcribed by two tandomly arrayed genes. A number of serpins are known to inhibit apoptosis in mammalian cells. In this study we demonstrate the ability of SCCA2 to inhibit tumor necrosis factor-alpha (TNF alpha)-induced apoptosis. HeLa cells stably transfected with SCCA2 cDNA had increased percentage cell survival and reduced DNA fragmentation. We investigated if the reactive centre loop (RCL) was necessary to allow SCCA2 to inhibit TNF alpha-mediated apoptosis. The RCL amino acids (E353Q, L354G, S355A), flanking the predicted cleavage site, were mutated and the resulting SCCA2 lost both the ability to inhibit cathepsin G and to protect stably transfected cells from TNF alpha-induced apoptosis. The presence of SCCA2 caused a decrease in the activation of caspase-3 upon induction with TNF alpha but no direct inhibition of caspases by SCCA2 has been found. Expression of cathepsin G was found to be induced in HeLa cells following treatment with TNF alpha. This protease has recently been shown to have a role in apoptosis through cleavage of substrates, so maybe the relevant target for SCCA2 in this system.


Subject(s)
Antigens, Neoplasm/physiology , Apoptosis/physiology , Serpins , Tumor Necrosis Factor-alpha/physiology , Antigens, Neoplasm/genetics , Caspase 3 , Caspases/metabolism , Cathepsin G , Cathepsins/biosynthesis , Enzyme Induction , Escherichia coli/genetics , HeLa Cells , Humans , Mutagenesis, Site-Directed , Recombinant Proteins/metabolism , Serine Endopeptidases , Transfection
18.
J Biol Chem ; 275(35): 26870-6, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10842168

ABSTRACT

Osmotic shock treatment of 3T3-L1 adipocytes causes an increase in glucose transport activity and translocation of GLUT4 protein similar to that elicited by insulin treatment. Insulin stimulation of GLUT4 translocation and glucose transport activity was completely inhibited by wortmannin, however, activation by osmotic shock was only partially blocked. Additionally, we have found that the newly identified insulin receptor substrate Gab-1 (Grb2-associated binder-1) is tyrosine-phosphorylated following sorbitol stimulation. Treatment of cells with the tyrosine kinase inhibitor genistein inhibited osmotic shock-stimulated Gab-1 phosphorylation as well as shock-induced glucose transport. Furthermore, pretreatment with the selective Src family kinase inhibitor PP2 completely inhibited the ability of sorbitol treatment to cause tyrosine phosphorylation of Gab-1. We have also shown that microinjection of anti-Gab-1 antibody inhibits osmotic shock-induced GLUT4 translocation. Furthermore, phosphorylated Gab-1 binds and activates phosphatidylinositol 3-kinase (PI3K) in response to osmotic shock. The PI3K activity associated with Gab-1 was 82% of that associated with anti-phosphotyrosine antibodies, indicating that Gab-1 is the major site for PI3K recruitment following osmotic shock stimulation. Although wortmannin only causes a partial block of osmotic shock-stimulated glucose uptake, wortmannin completely abolishes Gab-1 associated PI3K activity. This suggests that other tyrosine kinase-dependent pathways, in addition to the Gab-1-PI3K pathway, contribute to osmotic shock-mediated glucose transport. To date, Gab-1 is the first protein identified as a member of the osmotic shock signal transduction pathway.


Subject(s)
Adipocytes/drug effects , Glucose/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/physiology , 3T3 Cells , Adaptor Proteins, Signal Transducing , Adipocytes/metabolism , Androstadienes/pharmacology , Animals , Biological Transport , Enzyme Inhibitors/pharmacology , Mice , Osmotic Pressure , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Sorbitol/pharmacology , Tyrosine/metabolism , Wortmannin
19.
Gynecol Oncol ; 78(1): 62-6, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10873412

ABSTRACT

OBJECTIVES: Squamous cell carcinoma antigen (SCCA) is a member of the serpin superfamily, and has been used as a serological tumor marker for cervical squamous cell carcinomas. We have identified a closely related serpin gene, leupin (SCCA-2), which may be the fraction previously thought to be the acidic isoform of SCCA. The purpose of this study is to isolate the individual recombinant proteins, to examine their reactivity with current immunological detection methods, and to use a gene-specific method to examine their expression in the uterine cervix. METHODS: We have expressed and purified recombinant forms of SCCA and leupin individually. The proteins were characterized with respect to their isoelecric points and their reactivity with the monoclonal antibody from the current tumor marker diagnostic immunoassay (IMx SCC). Reverse transcription polymerase chain reaction (RT-PCR) with gene-specific primers was used to examine expression of both genes. RESULTS: Isoelectric focusing shows that leupin is the more acidic antigen with a determined pI for recombinant leupin (rLeupin) of 6.01, with rSCCA having a pI of 6.17. The IMx SCC monoclonal antibody recognized both rSCCA and rLeupin in immunoassays and immunoblots and both genes are expressed in normal cervix and in cervical carcinoma tissue. CONCLUSIONS: The findings from this study suggest that all previous clinical studies examining SCCA expression have used methodology that detects two gene products. The confirmation that leupin or SCCA-2 is the more acidic protein and that its expression is significantly elevated in cervical cancer suggests that this gene product may be the more important tumor marker.


Subject(s)
Antigens, Neoplasm/immunology , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/immunology , Serpins/immunology , Uterine Cervical Neoplasms/immunology , Antibodies, Monoclonal , Antigens, Neoplasm/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , DNA Primers , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoassay , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Serpins/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
20.
Cryobiology ; 40(3): 218-27, 2000 May.
Article in English | MEDLINE | ID: mdl-10860621

ABSTRACT

Extracts from a range of evolutionarily diverse plant and lichen species from the UK and maritime Antarctic have been assayed for inhibition of ice recrystallization. Approximately 25% of overwintering UK species and all Antarctic species exhibited antifreeze activity when exposed to low temperature. Preliminary characterization of the active extracts has demonstrated that the molecules co-opted to antifreeze activity by different species are biochemically diverse.


Subject(s)
Cryoprotective Agents/isolation & purification , Fungal Proteins/isolation & purification , Plant Proteins/isolation & purification , Acclimatization , Antarctic Regions , Cold Climate , Crystallization , Drug Stability , Freezing , Lichens/chemistry , Lichens/growth & development , Plant Development , Plants/chemistry , Sucrose , United Kingdom
SELECTION OF CITATIONS
SEARCH DETAIL
...