401
|
|
402
|
Syed S, Kelly KE, Sheffield WP. Inhibition of thrombin by hirudin genetically fused to wild-type or mutant antithrombin. Thromb Res 1996; 84:419-29. [PMID: 8987163 DOI: 10.1016/s0049-3848(96)00210-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Recombinant fusion proteins consisting of hirudin variant 3 (HV3) fused at its C-terminus to either of two forms of mature rabbit antithrombin (AT) were generated in COS-I cells. HV3 fused to wild-type AT was designated HAT, while a similar chimeric protein in which the P12 residue of AT was mutated from Ala to Thr, was designated HAT(H) for the Hamilton (A382T) mutation. Addition of the HV3 domain resulted in a decreased mobility of both HAT and HAT(H) relative to COS-derived AT (68 kDa versus 60 kDa). Both proteins had a greatly increased ability to inhibit thrombin in amidolytic activity assays, relative to recombinant AT. Addition of heparin to these reactions was without effect. Incubation of conditioned media containing recombinant AT with 125I-labelled thrombin resulted in the formation of SDS-stable AT-IIa complexes; no such complexes were detected in identical reactions containing either HV3-AT fusion protein. The two proteins did not differ significantly in their ability to compete for the binding of 125I-labelled thrombin to immobilized HV1 (CGP 39393). Both proteins were found to bind to heparin-Sepharose, but less tightly than unfused AT. This property was demonstrated by the peak elution of the fusion proteins at 0.65 M NaCl, as compared to that of COS-derived AT at 1.05 M NaCl. We conclude that the fusion proteins inhibit thrombin with similar affinity to unfused hirudin via their hirudin and not their antithrombin domains. The heparin-binding capability of these proteins may indicate the acquisition of vessel wall binding capacity by these novel forms of recombinant hirudin.
Collapse
Affiliation(s)
- S Syed
- Department of Pathology, McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
403
|
Urano T, Ihara H, Takada Y, Nagai N, Takada A. The inhibition of human factor Xa by plasminogen activator inhibitor type 1 in the presence of calcium ion, and its enhancement by heparin and vitronectin. BIOCHIMICA ET BIOPHYSICA ACTA 1996; 1298:199-208. [PMID: 8980646 DOI: 10.1016/s0167-4838(96)00131-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Plasminogen activator inhibitor type 1 (PAI-1), a member of serine proteinase inhibitor superfamily, is known to inhibit thrombin in the presence of either heparin or vitronectin. We analyzed possible inhibitory activity of PAI-1 on human factor Xa. PAI-1 inhibited factor Xa in the presence of calcium ion (Ca2+), whereas no inhibition was observed in the absence of Ca2+. Half maximal enhancement by Ca2+ was obtained at 0.8 mM. An equimolar complex formation between factor Xa and PAI-1 in the presence of Ca2+ was observed by SDS polyacrylamide gel electrophoresis. Both unfractionated heparin and vitronectin enhanced the inhibition only in the presence of Ca2+. Apparent second-order rate constant (ki) for the inhibition of factor Xa by PAI-1 at 5 mM Ca2+ was 1.6 x 10(4) M-1 s-1, and was enhanced 3-fold by 2 u/ml of heparin (4.6 x 10(4) M-1 s-1) and 10-fold by 100 nM vitronectin (1.6 x 10(5) M-1 s-1), respectively. The interaction between Ca(2+)-bound factor Xa and PAI-1 could be important from the view of PAI-1 neutralization and enhancement of fibrinolysis.
Collapse
Affiliation(s)
- T Urano
- Department of Physiology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | | | | | | | | |
Collapse
|
404
|
Abstract
The serpin family of protease inhibitors, to which alpha 1-antitrypsin belongs, has the unique feature of a mobile reactive center. Mutations within the critical regions of the molecule that control this mobility can allow premature changes in conformation with consequent abnormalities in folding and accompanying polymer formation. These abnormalities explain the plasma deficiency and liver inclusions associated with the common Z variant, as well as other variants of alpha 1-antitrypsin. The understanding of the molecular mechanisms provides a satisfying explanation for the clinical findings associated with these deficiency variants.
Collapse
Affiliation(s)
- R W Carrell
- Department of Haematology, University of Cambridge, England, UK
| | | | | | | |
Collapse
|
405
|
Abstract
The early history of alpha 1-antitrypsin deficiency, in the years between 1962 and 1965, and its impact on the understanding of emphysema abnormalities were reviewed by me in 1989. This report discusses the disease spectrum (including emphysema, liver, and vasculitic diseases) and focuses on the variable clinical expression of the deficiency state. Some new findings that may be relevant to the function of alpha 1-antitrypsin are also considered.
Collapse
Affiliation(s)
- S Eriksson
- Department of Medicine, University of Lund, University Hospital of Malmö, Sweden
| |
Collapse
|
406
|
Abstract
Among the major products secreted by the uteri of cattle, sheep, and pigs during pregnancy are glycoproteins with amino acid sequences that place them in the serpin (serine proteinase inhibitor) superfamily of proteins. The inferred amino acid sequences for bovine uterine serpin (boUS-1) and ovine uterine serpin (ovUS-1) exhibit about 72% sequence identity to each other but only about 50% and 56% identity, respectively, to two distinct porcine uterine serpins (poUS-1 and poUS-2). Despite these differences in primary structure, the uterine serpins possess well-conserved reactive center loop regions that contain several motifs present in the propeptide regions of pepsinogens. One such motif, VVVK, aligns with the first 4 amino acids of the aspartic proteinase inhibitor pepstatin. Although no inhibitory activity toward any serine proteinase has been found, at least one of the uterine serpins, ovUS-1, can bind specifically to immobilized pepsin A and can weakly inhibit the proteolytic activities of pepsin A and C (but not cathepsins D and E). OvUS-1 is the first specific inhibitor of aspartic proteinases to be identified in vertebrates and provides another example of a serpin with "crossover" activity. The pregnancy-associated glycoproteins (PAGs), which are secreted by the trophoblast layer of the placentas of ungulate species and are inactive members of the aspartic proteinase family, can also bind ovUS-1 and may be the natural target partners for the uterine serpins.
Collapse
Affiliation(s)
- N Mathialagan
- Department of Animal Sciences, University of Missouri, Columbia 65211, USA
| | | |
Collapse
|
407
|
Whisstock J, Lesk AM, Carrell R. Modeling of serpin-protease complexes: antithrombin-thrombin, alpha 1-antitrypsin (358Met-->Arg)-thrombin, alpha 1-antitrypsin (358Met-->Arg)-trypsin, and antitrypsin-elastase. Proteins 1996; 26:288-303. [PMID: 8953650 DOI: 10.1002/(sici)1097-0134(199611)26:3<288::aid-prot5>3.0.co;2-a] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Based on the most recent available crystal structures and biochemical studies of protease complexes of normal and mutant serine protease inhibitors (serpins), we have built models of the complexes: alpha 1-antitrypsin + human neutrophil elastase; alpha 1-antitrypsin Pittsburgh (358Met-->Arg) (Scott et al., J. Clin. Invest. 77:631-634, 1986) + tyrpsin; alpha 1-antitrypsin Pittsburgh (358Met-->Arg) + thrombin; and antithrombin + thrombin. All serpin sequences correspond to human molecules. The models show correct stereochemistry and no steric clashes between protease and inhibitor. The main structural differences in the serpins from the parent structures are: (1) the reactive center loop is inserted into the A-sheet as far as P12; (2) strand s1C is removed from the C-sheet; and (3) the C-terminus has changed conformation and interacts with the protease. In the absence of an X-ray structure determination of a serpin-protease complex, the demonstration that insertion of the reactive center loop into the A-sheet as far as P12 is stereochemically feasible provides structures of a protease-bound conformation of intact serpins with which to rationalize the properties of mutants, guide the design of experiments, and form a basis for further modeling studies, such as the investigation of the interaction of heparin with serpin-protease complexes.
Collapse
Affiliation(s)
- J Whisstock
- Department of Haematology, Medical Research Council Centre, Cambridge, United Kingdom
| | | | | |
Collapse
|
408
|
Jensen PH, Jensen TG, Laug WE, Hager H, Gliemann J, Pepinsky B. The exon 3 encoded sequence of the intracellular serine proteinase inhibitor plasminogen activator inhibitor 2 is a protein binding domain. J Biol Chem 1996; 271:26892-9. [PMID: 8900173 DOI: 10.1074/jbc.271.43.26892] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We have used a combination of biochemical and immunological methods to probe for proteins that interact with the cytoplasmic form of plasminogen activator inhibitor 2 (PAI-2) and to identify the structure in PAI-2 that mediates the binding. By affinity chromatography on immobilized PAI-2, we purified a collection of PAI-2-binding proteins. These proteins bound 125I-labeled PAI-2 in vitro (IC50, approximately 10-100 nM) in a calcium-independent reaction that did not abrogate the proteinase inhibitory function of PAI-2. Annexin I was identified among the eluted proteins, and purified annexins I, II, IV, and V, but not III and VI, possessed 125I-labeled PAI-2 binding activity. Immune precipitation by anti-PAI-2 monoclonal and polyclonal antibodies of metabolically labeled melanoma cells treated with a cleavable cross-linker prior to analysis revealed three prominent proteins with apparent masses of 100, 70, and 50 kDa. We localized the protein binding domain in PAI-2 between amino acid residues 66 and 98, as determined by using a PAI-2 mutant lacking this domain and a synthetic peptide spanning this region. This region of PAI-2 corresponds to exon 3 of the gene sequence thought to be critical for PAI-2 functions.
Collapse
Affiliation(s)
- P H Jensen
- Department of Medical Biochemistry, University of Aarhus, DK-8000 Aarhus C, Denmark
| | | | | | | | | | | |
Collapse
|
409
|
Ryu SE, Choi HJ, Kwon KS, Lee KN, Yu MH. The native strains in the hydrophobic core and flexible reactive loop of a serine protease inhibitor: crystal structure of an uncleaved alpha1-antitrypsin at 2.7 A. Structure 1996; 4:1181-92. [PMID: 8939743 DOI: 10.1016/s0969-2126(96)00126-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND The protein alpha1-antitrypsin is a prototype member of the serpin (serine protease inhibitor) family and is known to inhibit the activity of neutrophil elastase in the lower respiratory tract. Members of this family undergo a large structural rearrangement upon binding to a target protease, involving cleavage of the reactive-site loop. This loop is then inserted into the main body of the enzyme following the opening of a central beta sheet, leading to stabilization of the structure. Random mutageneses of alpha1-antitrypsin identified various mutations that stabilize the native structure and retard the insertion of the reactive-site loop. Structural studies of these mutations may reveal the mechanism of the conformational change. RESULTS We have determined the three-dimensional structure of an uncleaved alpha1-antitrypsin with seven such stabilizing mutations (hepta alpha1-antitrypsin) at 2.7 A resolution. From the comparison of the structure with other serpin structures, we found that hepta alpha1-antitrypsin is stabilized due to the release of various strains that exist in native wild type alpha1-antitrypsin, including unfavorable hydrophobic interactions in the central hydrophobic core. The reactive-site loop of hepta alpha1-antitrypsin is an extended strand, different from that of the previously determined structure of another uncleaved alpha1-antitrypsin, and indicates the inherent flexibility of the loop. CONCLUSIONS The present structural study suggests that the uncleaved alpha1-antitrypsin has many folding defects which can be improved by mutations. These folding defects seem to be utilized in a coordinated fashion in the regulation of the conformational switch of alpha1-antitrypsin. Some of the defects, represented by the Phe51 region and possibly the Met374 and the Thr59 regions, are part of the sheet-opening mechanism.
Collapse
Affiliation(s)
- S E Ryu
- Protein Engineering Research Division, Korea Research Institute of Bioscience and Biotechnology, KIST, Yusong, Taejon, South Korea.
| | | | | | | | | |
Collapse
|
410
|
Abstract
Alpha-1-antitrypsin (alpha 1-AT) deficiency is a well known cause of emphysema in adults. A subgroup of deficient individuals develops liver injury during infancy and childhood. In fact, it is the most common genetic cause of liver disease in children. Although lung injury is due to the decrease in alpha 1-AT function in the lung, allowing uninhibited elastolytic destruction of its connective tissue integrity, liver injury is probably due to retention of the mutant alpha 1-AT molecule in the endoplasmic reticulum (ER) of liver cells. Recent studies have shown that the mutant alpha 1-AT molecule polymerizes in the ER by a novel loop-sheet insertion mechanism. Other recent studies show that the subgroup of deficient individuals is susceptible to liver injury by virtue of unlinked genetic traits and/or environmental factors which interfere with degradation of the mutant alpha 1-AT molecules within the ER.
Collapse
Affiliation(s)
- D H Perlmutter
- Department of Pediatrics, Cell Biology and Physiology, Washington University, School of Medicine, St. Louis Children's Hospital, MO 63110, USA
| |
Collapse
|
411
|
Kjøller L, Martensen PM, Sottrup-Jensen L, Justesen J, Rodenburg KW, Andreasen PA. Conformational changes of the reactive-centre loop and beta-strand 5A accompany temperature-dependent inhibitor-substrate transition of plasminogen-activator inhibitor 1. EUROPEAN JOURNAL OF BIOCHEMISTRY 1996; 241:38-46. [PMID: 8898886 DOI: 10.1111/j.1432-1033.1996.0038t.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We have studied conformational changes of type-1 plasminogen-activator inhibitor (PAI-1) during a temperature-dependent inhibitor-substrate transition by measuring susceptibility of the molecule to non-target proteinases. When incubated at 0 degree C instead of the normally used 37 degrees C, a tenfold decrease in the specific inhibitory activity of active PAI-1 was observed. Accordingly, PAI-1 was recovered in a reactive-centre-cleaved form from incubations with urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA) at 0 degree C, but not at 37 degrees C. It thus behaved as a substrate for the target proteinases at the lower temperature. Active PAI-1 was exposed to a variety of non-target proteinases, including elastase, papain, thermolysin, trypsin, and V8 proteinase. It was found that specific peptide bonds in the reactive centre loop (RCL) and strand 5 in beta-sheet A (s5A) had a temperature-dependent proteolytic susceptibility, while the P17-P16 (E332-S333) bond, forming the hinge between s5A and the RCL, showed indistinguishable susceptibility to proteolysis by V8 proteinase at 0 degree and 37 degrees C. In latent and reactive-centre-cleaved PAI-1, all the bonds were resistant to proteolysis at the higher as well as the lower temperature. An anti-PAI-1 monoclonal antibody maintained the inhibitory activity of PAI-1 and prevented reactive centre cleavage at 0 degree C, and thus prevented substrate behaviour. Concomitantly, it caused specific changes in proteolytic susceptibility of s5A and the RCL, but it did not affect cleavage of the P17-P16 bond by V8 proteinase. Our observations suggest that temperature-dependent conformational changes of beta-sheet A and the RCL determine whether the serpin act as an inhibitor or a substrate. Furthermore they suggest that the RCL of PAI-1 is fully extracted from beta-sheet A in the inhibitory as well as in the substrate form, favoring a so-called induced conformational state model to explain why inhibitory activity requires partial insertion of the RCL into beta-sheet A.
Collapse
Affiliation(s)
- L Kjøller
- Department of Molecular and Structural Biology, University of Aarhus, Denmark
| | | | | | | | | | | |
Collapse
|
412
|
Schulze AJ, Quarzago D, Andreasen PA. A Spectroscopic Study of the Structures of Latent, Active and Reactive-Center-Cleaved Type-1 Plasminogen-Activator Inhibitor. ACTA ACUST UNITED AC 1996; 240:550-5. [PMID: 8856053 DOI: 10.1111/j.1432-1033.1996.0550h.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Type-1 plasminogen-activator inhibitor (PAI-1) was studied by Fourier-transform infrared spectroscopy, far-ultraviolet CD spectroscopy, and fluorescence-emission spectroscopy, with the aim to obtain structural information about its active form. The spectra of latent, active and reactive-center-cleaved forms of PAI-1 produced by HT-1080 cells were different. While the cleaved and the latent forms were similar with regard to their beta-structure content, comparison of the spectra of these forms with the spectra of active PAI-1 suggested a much higher degree of unordered structure for the active form compared with the latent and reactive-center-cleaved forms than previously assumed. We discuss our results with reference to the known three-dimensional X-ray structures of latent PAI-1, of reactive-center-cleaved serpins, including reactive-center-cleaved PAI-1, and of intact serpins, and with reference to previous results on the differences in the affinity of mAbs for the different PAI-1 forms. We interpret our results in favor of a global rearrangement of secondary structure during latency transition and reactive-center cleavage in PAI-1, not only involving the reactive-center loop and parts of beta-sheets A and C, but also the "rear' side of the molecule, such as helices H and G. Thus, we suggest flexibility in serpin structural elements that were previously regarded as rigid.
Collapse
Affiliation(s)
- A J Schulze
- Max-Planck-Institut für Biochemie, Abteilung Strukturforschung, Planegg-Martinsried, Germany.
| | | | | |
Collapse
|
413
|
Shirk RA, Church FC, Wagner WD. Arterial smooth muscle cell heparan sulfate proteoglycans accelerate thrombin inhibition by heparin cofactor II. Arterioscler Thromb Vasc Biol 1996; 16:1138-46. [PMID: 8792767 DOI: 10.1161/01.atv.16.9.1138] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Heparin cofactor II (HCII) is a potent thrombin inhibitor in the presence of heparin and dermatan sulfate, glycosaminoglycans that accelerate the inhibition reaction. HCII is postulated to be an extravascular thrombin inhibitor that is stimulated physiologically by dermatan sulfate proteoglycans. To understand how thrombin activity may be downregulated within the artery wall, cultured monkey aorta smooth muscle cell (SMC) proteoglycans were tested for their ability to accelerate thrombin inhibition by HCII. Early confluent SMC monolayers increased thrombin-HCII inhibition rates 2-fold to 4-fold compared with reactions in cell-free control wells (7.3 +/- 0.5 versus 2.7 +/- 0.2 x 10(4) mol.L-1.min-1, with and without SMCs, respectively; n = 7 experiments). Extracellular matrix obtained by cell monolayer removal also accelerated the thrombin-HCII inhibition reaction 3-fold to 5-fold. Rate increases were abolished by Polybrene or protamine sulfate. Pretreatment of monolayers with heparitinase I (and of extracellular matrix with HNO2) to degrade heparan sulfate blocked the thrombin-HCII inhibition rate increase. In contrast, pretreatment with chondroitinase ABC in the presence of proteinase inhibitors had no effect. "Pericellular" (cell surface- and extracellular matrix-derived) SMC heparan sulfate proteoglycans (HSPGs) were purified and fractionated by charge on DEAE-Sephacel. At a concentration of 1 microgram/mL hexuronic acid, high-charge HSPG stimulated a 7-fold thrombin-HCII inhibition rate increase relative to reactions without proteoglycan, whereas low-charge HSPG induced a 2-fold rate increase. In comparison, an 18-fold rate increase was observed with 1 microgram/mL dermatan sulfate proteoglycan purified from SMC culture media. These results indicate that SMC HSPG could contribute significantly to thrombin inhibition by HCII in the artery wall.
Collapse
Affiliation(s)
- R A Shirk
- Department of Comparative Medicine, Bowman Gray School of Medicine of Wake Forest University, Winston-Salem, NC 27157-1040, USA
| | | | | |
Collapse
|
414
|
Voorberg J, van Stempvoort G, Bos JM, Mertens K, van Mourik JA, Donath MJ. Enhanced thrombin sensitivity of a factor VIII-heparin cofactor II hybrid. J Biol Chem 1996; 271:20985-8. [PMID: 8702860 DOI: 10.1074/jbc.271.35.20985] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Generation of thrombin at a site of vascular injury is a key event in the arrest of bleeding. In addition to the conversion of fibrinogen into the insoluble fibrin, thrombin can initiate a number of positive and negative feedback mechanisms that either sustain or down-regulate clot formation. We have modulated the thrombin sensitivity of human blood coagulation factor VIII, an essential cofactor in the intrinsic pathway of blood coagulation. We have substituted an acidic region of factor VIII corresponding to amino acid sequence Asp712-Ala736 by amino acid sequence Ile51-Leu80 of the thrombin inhibitor heparin cofactor II. Functional analysis of the resulting factor VIII-heparin cofactor II hybrid, termed des-(868-1562)-factor VIII-HCII, revealed an increase in procoagulant activity as measured in a one-stage clotting assay. Incubation of purified des-(868-1562)-factor VIII-HCII with different amounts of thrombin showed that this protein was more readily activated by thrombin when compared with des-(868-1562)-factor VIII, a control protein lacking amino acid sequence Ile51-Leu80 of heparin cofactor II. This was manifested by an increase in the second order rate constant of activation by thrombin for des-(868-1562)-factor VIII-HCII (12.0 +/- 0.48 x 10(6) M-1 s-1) compared with des-(868-1562)-factor VIII (1.77 +/- 0.21 x 10(6) M-1 s-1). Our data suggest that amino acid sequence Ile51-Leu80 of heparin cofactor II endows factor VIII with increased sensitivity towards thrombin which results in accelerated clot formation.
Collapse
Affiliation(s)
- J Voorberg
- Department of Blood Coagulation, Central Laboratory of the Netherlands Red Cross Blood Transfusion Service, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
415
|
Kridel SJ, Chan WW, Knauer DJ. Requirement of lysine residues outside of the proposed pentasaccharide binding region for high affinity heparin binding and activation of human antithrombin III. J Biol Chem 1996; 271:20935-41. [PMID: 8702852 DOI: 10.1074/jbc.271.34.20935] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Variant forms of human antithrombin III with glutamine or threonine substitutions at Lys114, Lys125, Lys133, Lys136, and Lys139 were expressed in insect cells to evaluate their roles in heparin binding and activation. Recombinant native ATIII and all of the variants had very similar second order rate constants for thrombin inhibition in the absence of heparin, ranging from 1.13 x 10(5) M-1min-1 to 1.66 x 10(5) M-1min-1. Direct binding studies using 125I-flouresceinamine-heparin yielded a Kd of 6 nM for the recombinant native ATIII and K136T, whereas K114Q and K139Q bound heparin so poorly that a Kd could not be determined. K125Q had a moderately reduced affinity. Heparin binding affinity correlated directly with heparin cofactor activity. Recombinant native ATIII was nearly identical to plasma-purified ATIII, whereas K114Q and K139Q were severely impaired in heparin cofactor activity. K125Q and K136T were only slightly impaired. Based on these data, Lys114 and Lys139, which are outside of the putative pentasaccharide binding site, play pivotal roles in the high affinity binding of heparin to ATIII and the activation of thrombin inhibitory activity.
Collapse
Affiliation(s)
- S J Kridel
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, California 92717, USA
| | | | | |
Collapse
|
416
|
Niessen RW, Rezaee F, Reitsma PH, Peters M, de Vijlder JJ, Sturk A. Ligand-dependent enhancement of human antithrombin gene expression by retinoid X receptor alpha and thyroid hormone receptor beta. Biochem J 1996; 318 ( Pt 1):263-70. [PMID: 8761481 PMCID: PMC1217617 DOI: 10.1042/bj3180263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We studied potential modulators of antithrombin gene expression. A putative hormone response element (HRE) was identified by sequence similarity analysis of the antithrombin promoter, situated between nucleotides -92 and -54 relative to the transcription start site. This HRE contains three hexa-nucleotide motifs with an AGGTCA consensus, which are potential targets of members of the steroid/thyroid superfamily of nuclear receptors. Stimulation of the hepatoma cell line HepG2 with the receptor ligands L-3,5,3'-tri-iodothyronine, all-trans retinoic acid, or their combination, increased production of antithrombin into the culture medium by 1.3-, 1.6-, and 2.0-fold, respectively. In contrast, the receptor ligand 1,25-dihydroxy-cholecalciferol[1,25-(OH)2VitD3] did not influence antithrombin production. Analysis of promoter chloramphenicol acetyl-transferase (CAT) constructs, showed that the first 86 bp of the antithrombin promoter region are sufficient for basal transcription. The DNA length polymorphism of 32 bp or 108 bp, located upstream of position -276, did not influence anti-thrombin promoter activity. The antithrombin promoter activity dropped to background values when deleting the region -97/-49 of promoter fragment -453/+57. Transactivation of the antithrombin promoter by retinoid X receptor alpha (RXR alpha) (5-7-fold) or thyroid hormone receptor beta (TR beta) (4-5-fold) was only observed when at least -167/+57 bp of the promoter region is present in CAT constructs, and when the appropriate ligand of the nuclear receptor was added. This transactivation was not observed upon deletion of the antithrombin promoter region -97/-49. With three copies of the antithrombin promoter fragment -109/-42 in front of the thymidine kinase minimal promoter, transactivation was only obtained with RXR alpha, and not with TR beta. In conclusion, these results indicate that the ligand-dependent enhancement of antithrombin gene expression is regulated by RXR alpha as well as by TR beta. Transactivation of antithrombin gene expression by RXR alpha and TR beta appears to be dependent upon the presence of promoter region up to nucleotide -167. The HRE segment (-109/-42) only confers RXR alpha responsiveness to a heterologous promoter. Further study is needed to unravel the exact nature of this HRE and its 5'-flanking sequences.
Collapse
Affiliation(s)
- R W Niessen
- Center for Haemostasis, Thrombosis, Atherosclerosis and Inflammation Research, Academic Medical Centre, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
417
|
Maekawa H, Tollefsen DM. Role of the proposed serpin-enzyme complex receptor recognition site in binding and internalization of thrombin-heparin cofactor II complexes by hepatocytes. J Biol Chem 1996; 271:18604-9. [PMID: 8702511 DOI: 10.1074/jbc.271.31.18604] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Several serpin-enzyme complexes bind to a receptor on hepatocytes that mediates their endocytosis and lysosomal degradation. Joslin et al. (Joslin, G., Fallon, R. J., Bullock, J., Adams, S. P., and Perlmutter, D. H.(1991) J. Biol. Chem. 266, 11282-11288) proposed that a sequence near the C-terminal end of the serpin (e.g. FVFLM in alpha1-antitrypsin) binds to the serpin-enzyme complex receptor (SEC receptor). In experiments with synthetic peptides, they found that substitution of alanine at the fourth or fifth position in this sequence reduced the affinity of peptide binding to Hep G2 cells. To test the hypothesis that the corresponding sequence in heparin cofactor II (HCII), FLFLI (residues 456-460), mediates binding and uptake of the thrombin-HCII complex by Hep G2 cells, we constructed five recombinant HCII variants, F456A, L457A, F458A, L459A, and I460A. At 4 degrees C, the 125I-thrombin-HCII(native) complex bound reversibly to 0.6-2.6 x 10(5) sites per Hep G2 cell with a Kd of 19-32 nM. Binding was inhibited by excess unlabeled thrombin-HCII(native), thrombin-antithrombin, or elastase-alpha1-antitrypsin, but not by free HCII or thrombin, which is consistent with the reported specificity of the SEC receptor. However, complexes of thrombin with each of the HCII variants inhibited binding as effectively as the complex with native HCII. Competitive binding experiments with various concentrations of unlabeled thrombin-HCII(native) or thrombin-HCII(I460A) indicated that these complexes bind to Hep G2 cells with equal affinity. At 37 degrees C, complexes of 125I-thrombin with each of the five HCII variants were internalized and degraded at the same rate as the complex with native HCII. Our data suggest that the pentapeptide FLFLI in HCII is not involved in binding, internalization, and degradation of thrombin-HCII complexes by Hep G2 cells.
Collapse
Affiliation(s)
- H Maekawa
- Departments of Internal Medicine, Washington University, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
418
|
Elliott PR, Lomas DA, Carrell RW, Abrahams JP. Inhibitory conformation of the reactive loop of alpha 1-antitrypsin. NATURE STRUCTURAL BIOLOGY 1996; 3:676-81. [PMID: 8756325 DOI: 10.1038/nsb0896-676] [Citation(s) in RCA: 185] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The reactive site loop of the serpin family of serine proteinase inhibitors is flexible and can adopt a number of diverse conformations. A 2.9 A resolution structure of alpha 1-antitrypsin-the principal proteinase inhibitor in human plasma-shows the loop in a stable canonical conformation matching that found in all other families of serine proteinase inhibitors. This unexpected finding in the absence of loop insertion into the body of the molecule favours a two-stage mechanism of inhibition and provides a model for the heparin activation of antithrombin. The beta-pleated strand conformation of the loop also accounts for the polymerization of the serpins in disease and for their association with other beta-sheet structures, most notably the beta-amyloid of Alzheimer's disease.
Collapse
Affiliation(s)
- P R Elliott
- Department of Haematology, University of Cambridge, UK
| | | | | | | |
Collapse
|
419
|
Riewald M, Schleef RR. Human cytoplasmic antiproteinase neutralizes rapidly and efficiently chymotrypsin and trypsin-like proteases utilizing distinct reactive site residues. J Biol Chem 1996; 271:14526-32. [PMID: 8662739 DOI: 10.1074/jbc.271.24.14526] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Human cytoplasmic antiproteinase (CAP) is an intracellular serpin that has been reported to utilize Arg341 as the reactive site P1 residue to neutralize a broad variety of extracellular serine proteases with trypsin-like specificity. Both native CAP and recombinant CAP purified from Escherichia coli were observed to form SDS-stable complexes not only with 125I-thrombin and 125I-urokinase, but also with 125I-chymotrypsin. Kinetic studies indicated that the amidolytic activity of chymotrypsin is inhibited efficiently and rapidly by CAP in a two-step process with a dissociation constant Ki of an initial loose complex of 3.3 nM, a forward isomerization rate constant k2 to the tight complex of 0.014 s-1, and an overall second order association rate constant of 6 x 10(6) M-1 s-1, similar to the kinetic constants obtained for the formation of the trypsin-CAP complex. N-terminal amino acid sequencing and mass spectrometry indicated that chymotrypsin interacts with CAP at Met340, in contrast to thrombin, which interacts as expected at Arg341. Thus, CAP is the first serpin that has been shown to be capable to inhibit efficiently and with similar association rate constants different proteases at distinct reactive site residues, strongly supporting the notion of a highly mobile and flexible serpin reactive site loop and suggesting that this inhibitor may have evolved separate reactive sites for the specific regulation of different proteolytic activities.
Collapse
Affiliation(s)
- M Riewald
- Department of Vascular Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
420
|
Strömqvist M, Karlsson KE, Björquist P, Andersson JO, Byström M, Hansson L, Johansson T, Deinum J. Characterisation of the complex of plasminogen activator inhibitor type 1 with tissue-type plasminogen activator by mass spectrometry and size-exclusion chromatography. BIOCHIMICA ET BIOPHYSICA ACTA 1996; 1295:103-9. [PMID: 8679667 DOI: 10.1016/0167-4838(96)00035-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Glycosylated human plasminogen activator inhibitor type 1 (PAI-1), produced in Chinese hamster ovary (CHO) cells, showed a variety of compounds with different molecular weights when subjected to electrospray mass spectrometry (ES-MS), owing to the heterogeneity of the carbohydrate chains. However, non-glycosylated human PAI-1, produced in E. coli, gave rise to a prominent species with a molecular weight of 42,774, consistent with the amino-acid sequence. A non-glycosylated mutant of the proteinase domain (B-chain) of tissue-type plasminogen activator (tPA) produced in C 127 cells, had a molecular weight of 28,168. Full-length, glycosylated, tPA showed a large heterogeneity in molecular mass. For a mass study, a tPA-PAI-1 complex was formed, composed of non-glycosylated PAI-1 and non-glycosylated B-chain. This complex was remarkably stable at room temperature in buffer with a neutral pH. The mass spectrum of the complex provided two main species, a peptide with a mass of 3803 and a dominating species of 67,133. These masses are consistent with a complex where PAI-1 is cleaved at the P1-P1' position. A trace of a species with a molecular mass of 70,942 was also found, corresponding to the complete, non-dissociated complex with PAI-1. Separation of the cleaved peptide, corresponding to the hydrophobic C-terminal 33 amino-acid residues of PAI-1, from the complex, was achieved by size-exclusion chromatography in the presence of 30% acetonitrile. Thus, in the complex between tPA and PAI-1, the proteins are held together by a tight covalent bond, but the C-terminal cleaved peptide of PAI-1 is only bound to the complex by hydrophobic forces. To assess whether this is specific to the tPA B-chain alone, experiments with the complex of full-length, glycosylated tPA and glycosylated PAI-1 were also performed, and it was possible to demonstrate the release of the C-terminal PAI-1 peptide by chromatography, mass spectrometry, as well as by SDS-PAGE.
Collapse
|
421
|
Menoud PA, Sappino N, Boudal-Khoshbeen M, Vassalli JD, Sappino AP. The kidney is a major site of alpha(2)-antiplasmin production. J Clin Invest 1996; 97:2478-84. [PMID: 8647939 PMCID: PMC507332 DOI: 10.1172/jci118694] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The serpin alpha2-antiplasmin (alpha2-AP) is the major circulating inhibitor of plasmin; it plays a determining role in the regulation of intravascular fibrinolysis, In addition to blood plasma, plasmin formation occurs in various organs where it is thought to fulfill a spectrum of functions not restricted to clot lysis. Alpha2-AP is synthesized by hepatocytes, but other possible sites of production have not been investigated. To explore the potential extravascular contribution of alpha2-AP in the regulation of proteolysis, we have isolated the murine alpha2-AP cDNA and determined its mRNA distribution in adult tissues. In addition to liver, kidneys are major sites of alpha2-AP mRNA accumulation in the mouse. The transcript is present in epithelial cells lining the convoluted portion of proximal tubules, and its accumulation is under androgen control. Human kidneys also contain high levels of alpha2-AP mRNA. Moderate amounts Of alpha2-AP mRNA are detected in other murine tissues such as muscle, intestine, central nervous system, and placenta. Our observations indicate that alpha2-AP can be synthesized in a number of tissues, where it could function as a distal regulator of plasmin-mediated extracellular proteolysis.
Collapse
Affiliation(s)
- P A Menoud
- Department of Medicine, Division of Oncology, University of Geneva Medical School, 1211 Geneva 4, Switzerland
| | | | | | | | | |
Collapse
|
422
|
Abstract
Serine proteinase cleavage of proteins is essential to a wide variety of biological processes and is primarily regulated by protein inhibitors. Many inhibitors are conformationally rigid simulations of optimal serine proteinase substrates, which makes them highly efficient competitive inhibitors of target proteinases. In contrast, members of the serpin family of serine proteinase inhibitors display extensive flexibility and polymorphism, particularly in their reactive site segments and in beta-sheet secondary structure, which can take up and expel strands. Reactive site and beta-sheet polymorphism appear to be coupled in the serpins and may account for the extreme stability of serpin-proteinase complexes through the insertion of the reactive site strand into a beta-sheet. These unusual properties may have opened an adaptive pathway of proteinase regulation that was unavailable to the conformationally rigid proteinase inhibitors.
Collapse
Affiliation(s)
- H T Wright
- Department of Biochemistry and Molecular Biophysics, Virginia Commonwealth University, Richmond 23298, USA
| |
Collapse
|
423
|
Lee KN, Park SD, Yu MH. Probing the native strain iin alpha1-antitrypsin. NATURE STRUCTURAL BIOLOGY 1996; 3:497-500. [PMID: 8646533 DOI: 10.1038/nsb0696-497] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
424
|
Abstract
Antithrombin is the major proteinase inhibitor of thrombin and other blood coagulation proteinases. Antithrombin has two functional domains, a heparin binding site and a reactive centre (that complexes and inactivates the proteinase). Its deficiency results in an increased risk of venous thromboembolism. Appreciable progress has been made in recent years in understanding the structure and function of this protein, the genetic cause of inherited deficiency and its clinical consequence. The structure of antithrombin is now considered in terms of the models derived from X-ray crystallography, which have provided explanations for the function of its heparin interaction site and of its reactive loop. The structural organization of the antithrombin gene has been defined and numerous mutations have been identified that are responsible for antithrombin deficiency: these may reduce the level of the protein (Type I deficiency), alter the function of the protein (Type II deficiency, altering heparin binding or reactive sites), or even have multiple or 'pleiotropic effects' (Type II deficiency, altering both functional domains and the level of protein).
Collapse
Affiliation(s)
- D A Lane
- Department of Haematology, Charing Cross and Westminister Medical School, Hammersmith, London, UK
| | | | | | | |
Collapse
|
425
|
Deng G, Royle G, Wang S, Crain K, Loskutoff DJ. Structural and functional analysis of the plasminogen activator inhibitor-1 binding motif in the somatomedin B domain of vitronectin. J Biol Chem 1996; 271:12716-23. [PMID: 8662688 DOI: 10.1074/jbc.271.22.12716] [Citation(s) in RCA: 85] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Plasminogen activator inhibitor 1 (PAI-1) binds to the somatomedin B (SMB) domain of vitronectin (VN), a domain present in at least seven other proteins. In this study, we investigate the PAI-1 binding activity of these SMB homologs and attempt to more specifically localize the PAI-1 binding site within this domain. SMBVN and several of its homologs were expressed in Escherichia coli, purified, and tested for PAI-1 binding activity in a competitive ligand binding assay. Although recombinant SMBVN was fully active in this assay, none of the homologs bound to PAI-1 or competed with VN for PAI-1 binding. These inactive homologs are structurally related to SMBVN, having 33-45% sequence identity and containing all 8 cysteines at conserved positions. Thus, homolog-scanning experiments were conducted by exchanging progressively larger portions of the NH2- or COOH-terminal regions of active SMBVN with the corresponding regions of the inactive homologs. These experiments revealed that the minimum PAI-1-binding sequence was present in the central region (residues 12-30) of SMBVN. Alanine scanning mutagenesis further demonstrated that each of the 8 cysteines as well as Gly12, Asp22, Leu24, Try27, Tyr28, and Asp34 were critical for PAI-1 binding and were required to stabilize PAI-1 activity. These results indicate that the PAI-1 binding motif is localized to residues 12-30 of SMBVN and suggest that this motif is anchored in the active conformation by disulfide bonds.
Collapse
Affiliation(s)
- G Deng
- Department of Vascular Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
426
|
Villoutreix BO, Lilja H, Pettersson K, Lövgren T, Teleman O. Structural investigation of the alpha-1-antichymotrypsin: prostate-specific antigen complex by comparative model building. Protein Sci 1996; 5:836-51. [PMID: 8732755 PMCID: PMC2143410 DOI: 10.1002/pro.5560050505] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Prostate-specific antigen (PSA), produced by prostate cells, provides an excellent serum marker for prostate cancer. It belongs to the human kallikrein family of enzymes, a second prostate-derived member of which is human glandular kallikrein-1 (hK2). Active PSA and hK2 are both 237-residue kallikrein-like proteases, based on sequence homology. An hK2 model structure based on the serine protease fold is presented and compared to PSA and six other serine proteases in order to analyze in depth the role of the surface-accessible loops surrounding the active site. The results show that PSA and hK2 share extensive structural similarity and that most amino acid replacements are centered on the loops surrounding the active site. Furthermore, the electrostatic potential surfaces are very similar for PSA and hK2. PSA interacts with at least two serine protease inhibitors (serpins): alpha-1-antichymotrypsin (ACT) and protein C inhibitor (PCI). Three-dimensional model structures of the uncleaved ACT molecule were developed based upon the recent X-ray structure of uncleaved antithrombin. The serpin was docked both to PSA and hK2. Amino acid replacements and electrostatic complementarities indicate that the overall orientation of the proteins in these complexes is reasonable. In order to investigate PSA's heparin interaction sites, electrostatic computations were carried out on PSA, hK2, protein C, ACT, and PCI. Two heparin binding sites are suggested on the PSA surface and could explain the enhanced complex formation between PSA and PCI, while inhibiting the formation of the ACT-PSA complex, PSA, hK2, and their preliminary complexes with ACT should facilitate the understanding and prediction of structural and functional properties for these important proteins also with respect to prostate diseases.
Collapse
|
427
|
Gimenez-Roqueplo AP, Leconte I, Cohen P, Simon D, Guyene TT, Célerier J, Pau B, Corvol P, Clauser E, Jeunemaitre X. The natural mutation Y248C of human angiotensinogen leads to abnormal glycosylation and altered immunological recognition of the protein. J Biol Chem 1996; 271:9838-44. [PMID: 8621667 DOI: 10.1074/jbc.271.16.9838] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Common molecular variants of the angiotensinogen gene have been associated with human hypertension. The rare Tyr to Cys change at residue 248 of mature angiotensinogen was identified in one pedigree. Heterozygous individuals (Y248C) had a 40% decrease in plasma angiotensinogen concentration and a 35% reduction of the angiotensin I production rate. Recombinant wild-type (Tyr-248) and mutant (Cys-248) proteins were stably expressed in Chinese hamster ovary cells. Angiotensinogen monoclonal antibodies revealed marked differences in the epitope recognition of the mutant protein and allowed the demonstration of its presence in plasma of Y248C individuals. Similar kinetic constants of angiotensin I production with human renin were observed for both proteins. Western blot analysis showed similar heterogeneities; however, a 3-kDa increase in molecular mass for the Cys-248 protein was observed after immunopurification. Metabolic labeling of the intracellular Cys-248 protein showed a 61-kDa band in addition to the 55.5- and 58-kDa bands observed for the Tyr-248 protein, with all bands being sensitive to endoglycosidase H. In addition, pulse-chase studies revealed a slower intracellular processing for the Cys-248 protein. In conclusion, the Cys-248 mutation alters the structure, glycosylation, and secretion of angiotensinogen in Chinese hamster ovary cells and is accompanied by a decrease in plasma angiotensinogen concentration in Y248C individuals.
Collapse
|
428
|
Tucker HM, Gerard RD. Sequence requirements in the reactive-center loop of plasminogen-activator inhibitor-1 for recognition of plasminogen activators. EUROPEAN JOURNAL OF BIOCHEMISTRY 1996; 237:180-7. [PMID: 8620872 DOI: 10.1111/j.1432-1033.1996.0180n.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is a member of the serpin superfamily of proteins and is the fast acting inhibitor of both urinary plasminogen activator and tissue-type plasminogen activator. We have assessed the functional significance of reactive center residues on the carboxy-terminal side of the cleavage site of recombinant human PAI-1. Using site-directed mutagenesis, the P1'-P5' residues (P1' is the first residue on the carboxy-terminal side of the protease cleavage site) of the wild-type PAI-1 reactive center sequence were replaced with the corresponding sequences of plasminogen activator inhibitor-2, antithrombin, alpha 2-antiplasmin and protease nexin I. Rate constants of inhibition of the serine proteases urinary plasminogen activator, tissue-type plasminogen activator, plasmin and thrombin by the variants were determined. The results suggest a crucial role for both reactive center length and sequence in the inhibition of plasminogen activators by PAI-1. Analysis of substitutions at positions P4' and P5' both confirms and extends our previous work demonstrating a favorable electrostatic interaction between these residues and tissue-type plasminogen activator. None of the variants show dramatic increases in the rate constants of inhibition of other serine proteases, suggesting that these residues alone are not sufficient to confer protease specificity on PAI-1. Apparently, the determinants of the rapid inhibitory specificity of PAI-1 are localized to the P1'-P5' region of the reactive center and these residues act synergistically to produce the exquisite specificity of PAI-1 for plasminogen activators.
Collapse
Affiliation(s)
- H M Tucker
- Department of Biochemistry, University of Texas, Southwestern Medical Center, Dallas, USA
| | | |
Collapse
|
429
|
Patston PA, Gettins PG. Significance of secondary structure predictions on the reactive center loop region of serpins: a model for the folding of serpins into a metastable state. FEBS Lett 1996; 383:87-92. [PMID: 8612799 DOI: 10.1016/0014-5793(96)00231-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
To address how serpins might fold so as to adopt the mechanistically required metastable conformation we have compared the predicted secondary structures of the reactive center loops (RCLs) of a large number of serpins with those of the equivalent regions of other non-serpin protein proteinase inhibitors. Whereas the RCLs of non-serpin inhibitors are predicted to be loop or beta-strand, those of inhibitory serpins are strongly predicted to be alpha-helical. However, non-inhibitory serpins, which also adopt the metastable conformation, show no consistent preference for alpha-helix. We propose that the RCL primary structure plays little role in promoting the metastable serpin conformation. Instead we hypothesize that preference for the metastable state results from the incorporation of part of the RCL into beta-sheet C, which as a consequence precludes incorporation of the RCL into beta-sheet A to give the most stable conformation. Consequently the RCL must be exposed and by default will adopt the most stable conformation in this particular context, which is likely to be an alpha-helix irrespective of the primary structure. Thus the observed correlation between inhibitory properties in serpins and prediction of alpha-helix in the RCL may instead reflect a need for alanine residues between positions P12 and P9 for functioning as an inhibitor rather than a structural or mechanistic requirement for alpha-helix.
Collapse
Affiliation(s)
- P A Patston
- Department of Oral Medicine and Diagnostic Sciences, University of Illinois at Chicago, 60612 USA
| | | |
Collapse
|
430
|
Riewald M, Morgenstern KA, Schleef RR. Identification and characterization of the cytoplasmic antiproteinase (CAP) in human platelets. Evidence for the interaction of CAP with endogenous platelet proteins. J Biol Chem 1996; 271:7160-7. [PMID: 8636153 DOI: 10.1074/jbc.271.12.7160] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
To define the presence and potential role of platelet-associated protease inhibitors, we initiated a study designed to characterize the platelet components that are responsible for the formation of two SDS-stable complexes of approximately 58 and 70 kDa initially observed following the incubation of 125I-thrombin and human platelets. We demonstrate that thermal-mediated unfolding of the 58-kDa complex between 125I-thrombin and a nonsecreted platelet protein leads to an apparent molecular mass of 70 kDa. This platelet component is functionally and immunologically indistinguishable from the cytoplasmic antiproteinase (CAP), also known as placental thrombin inhibitor, a recently cloned member of the ovalbumin family of intracellular serpins (serine proteinase inhibitors). CAP-specific mRNA and antigen were detected in human platelets, suggesting that CAP synthesis occurs concurrent with platelet development. Utilizing quantitative immunoblotting, CAP antigen was estimated at 1.014 +/- 0.181 microg/10(9) nonstimulated platelets. After platelet activation with the calcium ionophore A23187, CAP antigen was detected in released microparticles at approximately 0. 195 +/- 0.031 microg/10(9) platelets and a fraction of platelet CAP was proteolytically modified. We provide evidence that these lower molecular mass species arise by cleavage of CAP at or near the reactive site loop. Most importantly, molecular sieving chromatography indicates the presence of an approximately 68-kDa SDS-labile complex between cleaved CAP and a cellular component in A23187-stimulated platelets, suggesting a physiological target of this intracellular serpin and a potential role for this inhibitor in regulating proteolytic activity that may be formed during platelet activation.
Collapse
Affiliation(s)
- M Riewald
- Department of Vascular Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | |
Collapse
|
431
|
Koch C, Jensen SS, Oster A, Houen G. A comparison of the immunogenicity of the native and denatured forms of a protein. APMIS 1996; 104:115-25. [PMID: 8619913 DOI: 10.1111/j.1699-0463.1996.tb00696.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The effect of heat denaturation on the physicochemical and immunological properties of a model protein, ovalbumin, and its formaldehyde/lysine-treated form was investigated. Polyacrylamide gel electrophoresis and gel filtration showed that heat denaturation converted ovalbumin to high Mr polymers, whereas formaldehyde/lysine-treated ovalbumin remained monomeric with only a small proportion forming oligomers. NMR analysis demonstrated that non-denatured structures could easily be differentiated from the denatured structures. Intraperitoneal immunization of rabbits and mice showed that both native and denatured forms of ovalbumin induced an immune response, but denatured forms of ovalbumin were found to be less immunogenic and to have a lower epitope density than native ovalbumin. Analysis of the antisera in crossed immunoelectrophoresis showed that they were specific for either native or denatured forms of ovalbumin. These findings were further investigated by ELISA and immunoaffinity chromatography, and the high specificity and low cross-reactivity was confirmed. We conclude that the immunogenic epitopes on denatured ovalbumin are different from those on ovalbumin, and that these epitopes reflect a continuum of denatured conformations.
Collapse
Affiliation(s)
- C Koch
- Centre for Biomolecular Recognition, Department of Immunology, Statens Seruminstitut, Copenhagen, Denmark
| | | | | | | |
Collapse
|
432
|
Komiyama T, Quan LT, Salvesen GS. Inhibition of cysteine and serine proteinases by the cowpox virus serpin CRMA. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1996; 389:173-6. [PMID: 8861008 DOI: 10.1007/978-1-4613-0335-0_21] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- T Komiyama
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
433
|
Carrell RW, Stein PE. The biostructural pathology of the serpins: critical function of sheet opening mechanism. BIOLOGICAL CHEMISTRY HOPPE-SEYLER 1996; 377:1-17. [PMID: 8929809 DOI: 10.1515/bchm3.1996.377.1.1] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The serpins illustrate the way in which the study of a protein family as a whole can clarify the functions of its individual members. Although the individual serpins have become remarkably diversified by evolution they all share a common structural pathology. We have previously shown how plotting of the dysfunctional natural mutations of the serpins on a template structure defines the domains controlling the mobility of the reactive centre loop of the molecule. Here we compare these natural mutations with reciprocal mutations in recombinants that restore the inhibitory stability of a labile member of the family, plasminogen activator inhibitor-1 (PAI-1). The combined results emphasise the critical part played by residues involved in the sliding movement that opens the A-sheet to allow reactive loop insertion. It is concluded that changes in these residues provide the prime explanation for the ready conversion of PAI-1 to the inactive latent state. The consistency of the overall results gives confidence in predicting the likely consequences of mutations in individual serpins. In particular the two common polymorphic mutations present in human angiotensinogen are likely to affect molecular stability and hence may be contributory factors to the observed association with vascular disease.
Collapse
Affiliation(s)
- R W Carrell
- Department of Haematology, University of Cambridge, UK
| | | |
Collapse
|
434
|
Abstract
Maspin, a serpin found in mammary epithelial cells, has been shown to have tumor suppressor activity. The gene is expressed in normal human mammary epithelial cells but down-regulated in invasive breast carcinomas. Similar patterns of expression at the RNA and protein levels are seen by Northern analysis with cells grown in culture and by immunostaining of tissues. Biological assays of invasion by tumor cells through matrigel membranes and of motility have shown that recombinant maspin inhibits both processes, and that its inhibitory action is totally lost by a single cleavage at the reaction center. Tumor transfectants expressing maspin are inhibited in growth and invasion in nude mice. Maspin is located in the cell membrane and extracellular matrix, and does not behave as a classical inhibitory serpin against any known target protease. Its mode of action is presently unknown.
Collapse
Affiliation(s)
- R Sager
- Department of Cancer Genetics, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
435
|
Wilczynska M, Fa M, Ohlsson PI, Ny T. The inhibition mechanism of serpins. Evidence that the mobile reactive center loop is cleaved in the native protease-inhibitor complex. J Biol Chem 1995; 270:29652-5. [PMID: 8530349 DOI: 10.1074/jbc.270.50.29652] [Citation(s) in RCA: 148] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Inhibitors that belong to the serine protease inhibitor or serpin family have reactive centers that constitute a mobile loop with P1-P1' residues acting as a bait for cognate protease. Current hypotheses are conflicting as to whether the native serpin-protease complex is a tetrahedral intermediate with an intact inhibitor or an acyl-enzyme complex with a cleaved inhibitor P1-P1' peptide bond. Here we show that the P1' residue of the plasminogen activator inhibitor type 1 mutant (P1' Cys) became more accessible to radiolabeling in complex with urokinase-type plasminogen activator (uPA) compared with its complex with catalytically inactive anhydro-uPA, indicating that complex formation with cognate protease leads to a conformational change whereby the P1' residue becomes more accessible. Analysis of chemically blocked NH2 termini of serpin-protease complexes revealed that the P1-P1' peptide bonds of three different serpins are cleaved in the native complex with their cognate protease. Complex formation and reactive center cleavage were found to be rapid and coordinated events suggesting that cleavage of the reactive center loop and the subsequent loop insertion induce the conformational changes required to lock the serpin-protease complex.
Collapse
Affiliation(s)
- M Wilczynska
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | | | | | | |
Collapse
|
436
|
Sprecher CA, Morgenstern KA, Mathewes S, Dahlen JR, Schrader SK, Foster DC, Kisiel W. Molecular cloning, expression, and partial characterization of two novel members of the ovalbumin family of serine proteinase inhibitors. J Biol Chem 1995; 270:29854-61. [PMID: 8530382 DOI: 10.1074/jbc.270.50.29854] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
A human placental lambda gt11 cDNA library was screened for sequences encoding proteins related to human proteinase inhibitor 6 (PI6), and two plaques were identified that displayed weak hybridization at high stringency. Isolation and characterization of the DNA inserts revealed two novel sequences encoding proteins composed of 376 and 374 amino acids with predicted molecular masses of approximately 42 kDa. The novel proteins displayed all of the structural features unique to the ovalbumin family of intracellular serpins including the apparent absence of a cleavable N-terminal signal sequence. The degree of amino acid sequence identity between the novel serpins and PI6 (63-68%) significantly exceeds that of any other combination of known intracellular serpins. The two novel serpins encoded by the two novel cDNA sequences have been designated as proteinase inhibitor 8 (PI8) and proteinase inhibitor 9 (PI9). The putative reactive center P1-P1' residues for PI8 and PI9 were identified as Arg339-Cys340 and Glu340-Cys341, respectively. PI9 appears to be unique in that it is the first human serpin identified with an acidic residue in the reactive center P1 position. In addition, the reactive center loop of PI9 exhibits 54% identity with residues found in the reactive center loop of the cowpox virus CrmA serpin. Two PI8 transcripts of 1.4 kilobases (kb) and 3.8 kb were detected by Northern analysis in equal and greatest abundance in liver and lung, while the 1.4-kb mRNA was in excess over the 3.8-kb mRNA in skeletal muscle and heart. Two PI9 transcripts of 3.4 and 4.4 kb were detected in equal and greatest abundance in lung and placenta and were weakly detected in all other tissues. PI8 and PI9 were expressed in baby hamster kidney and yeast cells, respectively. Immunoblot analyses using rabbit anti-PI6 IgG indicated the presence of PI8 in the cytosolic fraction of stably transfected cells that formed an SDS-stable 67-kDa complex with human thrombin. PI9 was purified to homogeneity from the yeast cell lysate by a combination of heparin-agarose chromatography and Mono Q fast protein liquid chromatography and migrated as a single band in SDS-polyacrylamide gel electrophoresis with an apparent molecular mass of 42 kDa. Purified recombinant PI9 failed to inhibit the amidolytic activities of trypsin, papain, thrombin, or Staphylococcus aureus endoproteinase Glu-C and did not form an SDS-stable complex when incubated with thrombin. The cognate intracellular proteinases that interact with PI8 and PI9 are unknown.
Collapse
Affiliation(s)
- C A Sprecher
- ZymoGenetics, Inc., Seattle, Washington 98102, USA
| | | | | | | | | | | | | |
Collapse
|
437
|
Debrock S, Declerck PJ. Characterization of common neoantigenic epitopes generated in plasminogen activator inhibitor-1 after cleavage of the reactive center loop or after complex formation with various serine proteinases. FEBS Lett 1995; 376:243-6. [PMID: 7498551 DOI: 10.1016/0014-5793(95)01289-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Plasminogen activator inhibitor-1 (PAI-1), an important risk factor for thrombotic diseases, is a member of the superfamily of serine proteinase inhibitors. To define structural rearrangements occurring during interaction between PAI-1 and its target proteinases we have raised monoclonal antibodies against the PAI-1/t-PA complex. Thirteen out of 401 monoclonal antibodies reacted preferentially with the PAI-1/t-PA complex as compared to free PAI-1 or free t-PA. Detailed characterization revealed the presence of two non-overlapping neoantigenic epitopes in the PAI-1/t-PA complex. Both neoantigenic epitopes were also exposed after complex formation between PAI-1 and either urokinase-type plasminogen activator, plasmin or thrombin as well as after cleavage of the reactive site loop of non-inhibitory substrate type PAI-1 variants. Thus, we have identified two neoantigenic epitopes, localized entirely in PAI-1, and commonly exposed after complex formation of active PAI-1 with various proteinases or after cleavage of substrate PAI-1. These monoclonal antibodies should facilitate further studies on the mechanism of interaction between various PAI-1 forms and its target proteinases.
Collapse
Affiliation(s)
- S Debrock
- Laboratory for Pharmaceutical Biology and Phytopharmacology, Faculty of Pharmaceutical Sciences, Katholieke Universiteit Leuven, Belgium
| | | |
Collapse
|
438
|
Conese M, Nykjaer A, Petersen CM, Cremona O, Pardi R, Andreasen PA, Gliemann J, Christensen EI, Blasi F. alpha-2 Macroglobulin receptor/Ldl receptor-related protein(Lrp)-dependent internalization of the urokinase receptor. J Cell Biol 1995; 131:1609-22. [PMID: 8522616 PMCID: PMC2120674 DOI: 10.1083/jcb.131.6.1609] [Citation(s) in RCA: 146] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The GPI-anchored urokinase plasminogen activator receptor (uPAR) does not internalize free urokinase (uPA). On the contrary, uPAR-bound complexes of uPA with its serpin inhibitors PAI-1 (plasminogen activator inhibitor type-1) or PN-1 (protease nexin-1) are readily internalized in several cell types. Here we address the question whether uPAR is internalized as well upon binding of uPA-serpin complexes. Both LB6 clone 19 cells, a mouse cell line transfected with the human uPAR cDNA, and the human U937 monocytic cell line, express in addition to uPAR also the endocytic alpha 2-macroglobulin receptor/low density lipoprotein receptor-related protein (LRP/alpha 2-MR) which is required to internalize uPAR-bound uPA-PAI-1 and uPA-PN-1 complexes. Downregulation of cell surface uPAR molecules in U937 cells was detected by cytofluorimetric analysis after uPA-PAI-1 and uPA-PN-1 incubation for 30 min at 37 degrees C; this effect was blocked by preincubation with the ligand of LRP/alpha 2-MR, RAP (LRP/alpha 2-MR-associated protein), known to block the binding of the uPA complexes to LRP/alpha 2-. MR. Downregulation correlated in time with the intracellular appearance of uPAR as assessed by confocal microscopy and immuno-electron microscopy. After 30 min incubation with uPA-PAI-1 or uPA-PN-1 (but not with free uPA), confocal microscopy showed that uPAR staining in permeabilized LB6 clone 19 cells moved from a mostly surface associated to a largely perinuclear position. This effect was inhibited by the LRP/alpha 2-MR RAP. Perinuclear uPAR did not represent newly synthesized nor a preexisting intracellular pool of uPAR, since this fluorescence pattern was not modified by treatment with the protein synthesis inhibitor cycloheximide, and since in LB6 clone 19 cells all of uPAR was expressed on the cell surface. Immuno-electron microscopy confirmed the plasma membrane to intracellular translocation of uPAR, and its dependence on LRP/alpha 2-MR in LB6 clone 19 cells only after binding to the uPA-PAI-1 complex. After 30 min incubation at 37 degrees C with uPA-PAI-1, 93% of the specific immunogold particles were present in cytoplasmic vacuoles vs 17.6% in the case of DFP-uPA. We conclude therefore that in the process of uPA-serpin internalization, uPAR itself is internalized, and that internalization requires the LRP/alpha 2-MR.
Collapse
Affiliation(s)
- M Conese
- Dipartimento di Ricerca Biologica e Tecnologica, H.S. Raffaele Scientific Institute, Università di Milano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
439
|
Nishibori M, Chikai T, Kawabata M, Ohta J, Ubuka T, Saeki K. Purification of a novel serpin-like protein from bovine brain. Neurosci Res 1995; 24:47-52. [PMID: 8848289 DOI: 10.1016/0168-0102(95)00973-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We purified a novel serine proteinase inhibitor (serpin)-like protein from the bovine brain and named it B-43 from its molecular mass, 43 kDa. A cleaved peptide from B-43 was copurified with the native B-43. Partial amino acid sequencing of the purified B-43 showed that this protein was homologous to glia-derived nexin/protease nexin-1 (GDN/PN-1), plasminogen activator inhibitor 2, leukocyte elastase inhibitor (LEI) and placental thrombin inhibitor (PTI) among the serpins. Although B-43 had a similar amino acid composition to these serpins, the biochemical features of B-43 were different from them. B-43 did not form sodium dodecyl sulfate (SDS)-resistant serpin-proteinase complexes with thrombin, urokinase, pancreatic elastase and plasmin, suggesting that these proteinases were not the targets of B-43. In contrast to GDN/PN-1, B-43 did not have an affinity for heparin. B-43, having different biochemical properties from GDN/PN-1, appears to be an additional serpin expressed in the brain.
Collapse
Affiliation(s)
- M Nishibori
- Department of Pharmacology, Okayama University Medical School, Japan
| | | | | | | | | | | |
Collapse
|
440
|
Janssen OE, Chen B, Büttner C, Refetoff S, Scriba PC. Molecular and structural characterization of the heat-resistant thyroxine-binding globulin-Chicago. J Biol Chem 1995; 270:28234-8. [PMID: 7499319 DOI: 10.1074/jbc.270.47.28234] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Thyroxine-binding globulin (TBG) is the main transport protein for thyroxine (T4) in blood. It shares considerable sequence homology with alpha 1-antitrypsin (AT) and other members of the serine proteinase inhibitor (serpin) superfamily of proteins. The crystallographic structure of AT has been determined and was found to represent the archetype of the serpins. This model has been used for structure-function correlations of TBG. Sequence analysis of the heat-resistant variant TBG-Chicago (TBG-CH) revealed a substitution of the normal tyrosine 309 with phenylalanine. For further analysis, vectors containing the coding regions of normal TBG (TBG-N) and TBG-CH were constructed, transcribed in vitro, and expressed in Xenopus oocytes. Both TBGs were secreted into the culture medium and could not be distinguished by gel electrophoresis. Scatchard analysis of T4 binding to TBG-N and -CH revealed no significant differences in binding affinity. The rate of heat denaturation of TBGs was determined by measurement of residual T4 binding capacity after incubation at 60 degrees C for various periods of time. The half-life values of denaturation of TBG-N and -CH were 7 and 132 min, respectively. The tyrosine 309 to phenylalanine substitution of TBG-CH involves a highly conserved phenylalanine residue of the serpins. The respective phenylalanine 312 of AT ties the alpha-helix hI1 to the molecule, thus stabilizing the tertiary structure. A substitution with tyrosine would disrupt this interaction. Accordingly, stabilization of the TBG molecule by replacement of tyrosine with phenylalanine in position 309 causes the increased heat stability of TBG-CH.
Collapse
Affiliation(s)
- O E Janssen
- Department of Medicine, Klinikum Innenstadt, Ludwig-Maximilians-University, Munich, Federal Republic of Germany
| | | | | | | | | |
Collapse
|
441
|
Riewald M, Schleef RR. Molecular cloning of bomapin (protease inhibitor 10), a novel human serpin that is expressed specifically in the bone marrow. J Biol Chem 1995; 270:26754-7. [PMID: 7592909 DOI: 10.1074/jbc.270.45.26754] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Serine proteinase inhibitors or serpins are a super-family of homologous proteins that are for the most part involved in the regulation of proteolytic processes in a variety of biological systems. Utilizing a polymerase chain reaction-based strategy we have cloned a novel member of the ovalbumin family of serpins from a human bone marrow cDNA library. The new gene encodes a 397-amino acid protein, designated bomapin, with a calculated molecular mass of 45 kDa and 48% amino acid identity with plasminogen activator inhibitor-2, human leukocyte elastase inhibitor, and cytoplasmic antiproteinase. A single 2.3-kilobase bomapin transcript is highly expressed in human bone marrow cells but was undetectable in all other analyzed human tissues. In vitro transcription and translation of the bomapin cDNA revealed the synthesis of an appropriately sized protein that was able to form SDS-stable complexes with thrombin and trypsin. The restricted expression of bomapin to the bone marrow raises the possibility that this serpin may play a role in the regulation of protease activities during hematopoiesis.
Collapse
Affiliation(s)
- M Riewald
- Department of Vascular Biology, Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
442
|
Owen CA, Campbell MA, Sannes PL, Boukedes SS, Campbell EJ. Cell surface-bound elastase and cathepsin G on human neutrophils: a novel, non-oxidative mechanism by which neutrophils focus and preserve catalytic activity of serine proteinases. J Cell Biol 1995; 131:775-89. [PMID: 7593196 PMCID: PMC2120617 DOI: 10.1083/jcb.131.3.775] [Citation(s) in RCA: 289] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Serine proteinases of human polymorphonuclear neutrophils play an important role in neutrophil-mediated proteolytic events; however, the non-oxidative mechanisms by which the cells can degrade extracellular matrix in the presence of proteinase inhibitors have not been elucidated. Herein, we provide the first report that human neutrophils express persistently active cell surface-bound human leukocyte elastase and cathepsin G on their cell surface. Unstimulated neutrophils have minimal cell surface expression of these enzymes; however, phorbol ester induces a 30-fold increase. While exposure of neutrophils to chemoattractants (fMLP and C5a) stimulates modest (two- to threefold) increases in cell surface expression of serine proteinases, priming with concentrations of lipopolysaccharide as low as 100 fg/ml leads to striking (up to 10-fold) increase in chemoattractant-induced cell surface expression, even in the presence of serum proteins. LPS-primed and fMLP-stimulated neutrophils have approximately 100 ng of cell surface human leukocyte elastase activity per 10(6) cells. Cell surface-bound human leukocyte elastase is catalytically active, yet is remarkably resistant to inhibition by naturally occurring proteinase inhibitors. These data indicate that binding of serine proteinases to the cell surface focuses and preserves their catalytic activity, even in the presence of proteinase inhibitors. Upregulated expression of persistently active cell surface-bound serine proteinases on activated neutrophils provides a novel mechanism to facilitate their egress from the vasculature, penetration of tissue barriers, and recruitment into sites of inflammation. Dysregulation of the cell surface expression of these enzymes has the potential to cause tissue destruction during inflammation.
Collapse
Affiliation(s)
- C A Owen
- Department of Medicine, University of Utah Health Sciences Center, Salt Lake City 84132, USA
| | | | | | | | | |
Collapse
|
443
|
One-phase reverse zymography after denaturing gel electrophoresis: High sensitivity detection of activity of plasminogen activator inhibitor 2 and other protease inhibitors. ACTA ACUST UNITED AC 1995. [DOI: 10.1016/s0268-9499(95)80080-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
444
|
Becerra SP, Sagasti A, Spinella P, Notario V. Pigment epithelium-derived factor behaves like a noninhibitory serpin. Neurotrophic activity does not require the serpin reactive loop. J Biol Chem 1995; 270:25992-9. [PMID: 7592790 DOI: 10.1074/jbc.270.43.25992] [Citation(s) in RCA: 172] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Pigment epithelium-derived factor (PEDF), a neurite-promoting factor, has an amino acid primary structure that is related to members of the serine protease inhibitor (serpin) family. Controlled proteolysis of native PEDF (50 kDa) with either trypsin, chymotrypsin, elastase, or subtilisin yields in each case one major limited product of 46 kDa as analyzed by SDS-polyacrylamide gel electrophoresis. N-terminal sequence analysis of the isolated 46-kDa products indicates a favored cleavage region located toward the C-terminal end of PEDF. A proteolyzed PEDF protein reaction mixture reveals two overlapping sequences: that of the N terminus of intact PEDF and that of an internal region, consistent with cleavage of PEDF about position 382. These data indicate that PEDF protein has a globular conformation with one protease-sensitive exposed loop that contains the homologous serpin-reactive site. Cleavage within the reactive-site loop of PEDF does not cause a conformational change in the molecules (the stressed (S)-->relaxed (R) transition) and results in heat denaturation identical to its native counterpart. This lack of conformational change is also seen upon cleavage within the reactive-site loop of the noninhibitory serpin ovalbumin. Furthermore, the PEDF neurite-promoting function is not lost with cleavage of the exposed loop. Recombinant PEDF polypeptide fragments with larger truncations from the C-terminal end show neurotrophic activity. Our results clearly indicate that integrity of the PEDF homologous serpin reactive center is dispensable for neurotrophic activity. Thus, the PEDF induction of neurites must be mediated by a mechanism other than serine protease inhibition. Altogether our data indicate that PEDF belongs to the subgroup of noninhibitory serpins and that its N-terminal region confers a neurite-promoting activity to the protein. The neurotrophic active site of PEDF is separated from the serpin reactive-site loop, not only in the primary structure, but also in the folded protein structure.
Collapse
Affiliation(s)
- S P Becerra
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
445
|
Lawrence DA, Ginsburg D, Day DE, Berkenpas MB, Verhamme IM, Kvassman JO, Shore JD. Serpin-protease complexes are trapped as stable acyl-enzyme intermediates. J Biol Chem 1995; 270:25309-12. [PMID: 7592687 DOI: 10.1074/jbc.270.43.25309] [Citation(s) in RCA: 208] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The serine protease inhibitors of the serpin family are an unusual group of proteins thought to have metastable native structures. Functionally, they are unique among polypeptide protease inhibitors, although their precise mechanism of action remains controversial. Conflicting results from previous studies have suggested that the stable serpin-protease complex is trapped in either a tight Michaelis-like structure, a tetrahedral intermediate, or an acyl-enzyme. In this report we show that, upon association with a target protease, the serpin reactive-center loop (RCL) is cleaved resulting in formation of an acyl-enzyme intermediate. This cleavage is coupled to rapid movement of the RCL into the body of the protein bringing the inhibitor closer to its lowest free energy state. From these data we suggest a model for serpin action in which the drive toward the lowest free energy state results in trapping of the protease-inhibitor complex as an acyl-enzyme intermediate.
Collapse
Affiliation(s)
- D A Lawrence
- American Red Cross Holland Laboratory, Rockville, Maryland 20855, USA
| | | | | | | | | | | | | |
Collapse
|
446
|
Packard BZ, Lee SS, Remold-O'Donnell E, Komoriya A. A serpin from human tumor cells with direct lymphoid immunomodulatory activity: mitogenic stimulation of human tumor-infiltrating lymphocytes. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1269:41-50. [PMID: 7578269 DOI: 10.1016/0167-4889(95)00113-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A serum-free supernatant from an epidermal carcinoma cell line has previously been shown to contain mitogenic activity for human tumor infiltrating lymphocytes in culture [1]. From this conditioned medium we have now purified to homogeneity, as determined by SDS-PAGE analysis, a ca. 45 kDa protein which stimulates [3H]thymidine incorporation into the DNA of these human T-lymphocytes. Amino acid composition data and immunoreactivity of the purified protein as well as sequence analyses of 7 tryptic fragments obtained therefrom suggest a strong similarity with human monocyte/neutrophil elastase inhibitor, which is a member of the serine protease inhibitor (serpin) superfamily. We have previously identified and purified from the same conditioned medium a 36 kDa protein with myeloid immunomodulatory activity [2]. Taken together, these two reports support the role of tumor-derived soluble factors in tumor immunosurveillance.
Collapse
Affiliation(s)
- B Z Packard
- OncoImmunin, Inc., College Park, MD 20742, USA
| | | | | | | |
Collapse
|
447
|
Kwon KS, Song M, Yu MH. Purification and characterization of alpha 1-antitrypsin secreted by recombinant yeast Saccharomyces diastaticus. J Biotechnol 1995; 42:191-5. [PMID: 7576538 DOI: 10.1016/0168-1656(95)00079-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The secreted human alpha 1-antitrypsin (alpha 1AT) produced by yeast was purified from the culture medium by ultrafiltration, ammonium sulfate fractionation (60-75% saturation), protamine sulfate treatment, and ion-exchange chromatography. Molecular mass of the purified alpha 1AT was 52 kDa, which is similar to that of human plasma alpha 1AT. Yeast-produced alpha 1AT was fully functional as an inhibitor compared with the plasma form. Unlike plasma alpha 1AT, however, treatment of the yeast-produced alpha 1AT with endoglycosidase H decreased the molecular mass to that of recombinant alpha 1AT produced in Escherichia coli, indicating the high-mannose type N-linked glycosylation of the secreted alpha 1AT. Glycosylation in yeast cells enhanced kinetic stability of alpha 1AT towards heat deactivation.
Collapse
Affiliation(s)
- K S Kwon
- Korea Research Institute of Bioscience and Biotechnology, Korea Institute of Science and Technology, Taejon, South Korea
| | | | | |
Collapse
|
448
|
Lomas DA, Stone SR, Llewellyn-Jones C, Keogan MT, Wang ZM, Rubin H, Carrell RW, Stockley RA. The control of neutrophil chemotaxis by inhibitors of cathepsin G and chymotrypsin. J Biol Chem 1995; 270:23437-43. [PMID: 7559504 DOI: 10.1074/jbc.270.40.23437] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Neutrophil chemotaxis plays an important role in the inflammatory response and when excessive or persistent may augment tissue damage. The effects of inhibitors indicated the involvement of one or more serine proteinases in human neutrophil migration and shape change in response to a chemoattractant. Monospecific antibodies, chloromethylketone inhibitors, and reactive-site mutants of alpha 1-antitrypsin and alpha 1-antichymotrypsin were used to probe the specificity of the proteinases involved in chemotaxis. Antibodies specific for cathepsin G inhibited chemotaxis. Moreover, rapid inhibitors of cathepsin G and alpha-chymotrypsin suppressed neutrophil chemotaxis to the chemoattractants N-formyl-L-methionyl-L-leucyl-L-phenylalanine (fMLP) and zymosan-activated serum in multiple blind well assays and to fMLP in migration assays under agarose. The concentrations of antichymotrypsin mutants that reduced chemotaxis by 50% would inactivate free cathepsin G with a half-life of 1.5-3 s, whereas the concentrations of chloromethylketones required to produce a similar inhibition of chemotaxis would inactivate cathepsin G with a half-life of 345 s. These data suggest different modes of action for these two classes of inhibitors. Indeed the chloromethylketone inhibitors of cathepsin G (Z-Gly-Leu-Phe-CMK) and to a lesser extent of chymotrypsin (Cbz-Gly-Gly-Phe-CMK) mediated their effect by preventing a shape change in the purified neutrophils exposed to fMLP. Antichymotrypsin did not affect shape change in response to fMLP even at concentrations that were able to reduce neutrophil chemotaxis by 50%. These results support the involvement of cell surface proteinases in the control of cell migration and show that antichymotrypsin and chloromethylketones have differing modes of action. This opens the possibility for the rational design of anti-inflammatory agents targeted at neutrophil membrane enzymes.
Collapse
Affiliation(s)
- D A Lomas
- Department of Medicine, University of Cambridge, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
449
|
Barnes RC, Worrall DM. Identification of a novel human serpin gene; cloning sequencing and expression of leupin. FEBS Lett 1995; 373:61-5. [PMID: 7589435 DOI: 10.1016/0014-5793(95)01015-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A novel serpin gene has been isolated, cloned and sequenced. A PCR amplified fragment of the gene was originally identified from human genomic DNA, and the full-length cDNA was subsequently isolated from HeLa cells and sequenced. The novel serpin is very high in protein sequence similarity (91.8%) to the squamous cell carcinoma antigen (SCCA), but contains substantial differences in the reactive site loop sequence, including a different amino acid (leucine) in the P1 position. The gene product, named leupin, is expressed in HeLa cells, SKGIIIa cells and human placenta. The protein has a predicted M(r) of 44,857 and an isoelectric point of 6.04 which is consistent with the more acidic form of SCCA associated with squamous cell carcinomas.
Collapse
Affiliation(s)
- R C Barnes
- Department of Biochemistry, University College Dublin, Ireland
| | | |
Collapse
|
450
|
Aertgeerts K, De Bondt HL, De Ranter CJ, Declerck PJ. Mechanisms contributing to the conformational and functional flexibility of plasminogen activator inhibitor-1. NATURE STRUCTURAL BIOLOGY 1995; 2:891-7. [PMID: 7552714 DOI: 10.1038/nsb1095-891] [Citation(s) in RCA: 87] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is unique among the serine proteinase inhibitors (serpins) in that it can adopt at least three different conformations (active, substrate and latent). We report the X-ray structure of a cleaved substrate variant of human PAI-1, which has a new beta-strand s4A formed by insertion of the amino-terminal portion of the reactive-site loop into beta-sheet A subsequent to cleavage. This is in contrast to the previous suggestion that the non-inhibitory function of substrate-type serpins is mainly due to an inability of the reactive-site loop to adopt this conformation. Comparison with the structure of latent PAI-1 provides insights into the molecular determinants responsible for the transition of the stressed active conformation to the thermostable latent conformation.
Collapse
Affiliation(s)
- K Aertgeerts
- Laboratory for Analytical Chemistry and Medicinal Physicochemistry, Faculty of Pharmaceutical Sciences, Katholieke Universiteit Leuven, Belgium
| | | | | | | |
Collapse
|