Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 248
Filter
1.
Oncogene ; 26(6): 851-8, 2007 Feb 08.
Article in English | MEDLINE | ID: mdl-16878148

ABSTRACT

Tumor markers can facilitate understanding molecular cell biology of neoplasia and provide potential targets for the diagnosis and insight for intervention. We here identify a novel murine gene, hepcarcin (hcn), encoding a 7-kb mRNA-like transcript. The gene appears to be the murine ortholog of the human alpha gene, that is, MALAT-1. The gene and homologs lack credible open reading frames, consistent with a highly conserved large noncoding RNA (ncRNA). In all nodules of procarcinogen-induced murine hepatocellular carcinomas (HCCs) and human HCCs, expression was markedly elevated compared to the uninvolved liver. Quantitative analyses indicated a 6-7-fold increased RNA level in HCCs versus uninvolved liver, advancing this as a molecule of interest. This ncRNA was overexpressed in all five non-hepatic human carcinomas analysed, consistent with a potential marker for neoplastic cells and potential participant in the molecular cell biology of neoplasia.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Neoplasms/genetics , RNA, Untranslated/genetics , Amino Acid Sequence , Animals , Base Sequence , Biomarkers , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Conserved Sequence , Humans , Mice , Molecular Sequence Data , Neoplasms/metabolism , Neoplasms/pathology , Phylogeny , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , RNA, Untranslated/metabolism , Sequence Alignment , Transcription, Genetic/genetics
2.
J Control Release ; 114(2): 230-41, 2006 Aug 28.
Article in English | MEDLINE | ID: mdl-16876899

ABSTRACT

Subcutaneously injected therapeutics must pass through the interstitial matrix of the skin in order to reach their intended targets. This complex, three-dimensional structure limits the type and quantity of drugs that can be administered by local injection. Here we found that depolymerization of the viscoelastic component of the interstitial matrix in animal models with a highly purified recombinant human hyaluronidase enzyme (rHuPH20) increased the dispersion of locally injected drugs, across a broad range of molecular weights without tissue distortion. rHuPH20 increased infusion rates and the pattern and extent of appearance of locally injected drugs in systemic blood. In particular, rHuPH20 changed the pharmacokinetic profiles and significantly augmented the absolute bioavailability of locally injected large protein therapeutics. Importantly, within 24 h of injection, the interstitial viscoelastic barriers were restored without histologic alterations or signs of inflammation. rHuPH20 may function as an interstitial delivery enhancing agent capable of increasing the dispersion and bioavailability of coinjected drugs that may enable subcutaneous administration of therapeutics and replace intravenous delivery.


Subject(s)
Hyaluronoglucosaminidase/pharmacology , Pharmaceutical Preparations/metabolism , Adenoviridae/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibody Formation/drug effects , Biological Availability , Biological Transport, Active/drug effects , Capillaries/cytology , Capillaries/metabolism , Capillary Permeability/drug effects , Cytokines/administration & dosage , Cytokines/pharmacokinetics , Drug Delivery Systems , Drug Therapy , Endothelial Cells/metabolism , Female , Genetic Therapy , Humans , Hyaluronoglucosaminidase/administration & dosage , Injections, Subcutaneous , Interferon Type I/administration & dosage , Interferon Type I/pharmacokinetics , Interferon Type I/therapeutic use , Macaca mulatta , Male , Mice , Mice, Nude , Molecular Weight , Particle Size , Polyethylene Glycols , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology
3.
Clin Lab Haematol ; 25(5): 321-5, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12974724

ABSTRACT

Monocytes express tissue factor (TF) as a result of cytokine stimulation or endothelial adherence. We evaluated monocyte-platelet interaction in vitro as another trigger for monocyte TF enhancement in human mononuclear cells isolated by density gradient centrifugation from peripheral blood. Cell TF procoagulant activity (TF-PCA) was quantitated by a one-stage recalcification clotting time assay. Platelets were counted and identified by whole blood flow cytometry as CD61 positive particles, activated platelets were characterized by P-Selectin (CD62) expression, and monocytes by surface CD14 expression. A significant correlation between normalized TF-PCA of isolated mononuclear cells and platelet count was shown (r = 0.43, P < 0.001). Percentage of activated platelets in baseline samples was 4.2 +/- 3.5 while adenosine diphosphate (ADP) increased platelet positivity to 34 +/- 17% (P < 0.001). After isolation, 52 +/- 12% of platelets within suspensions were activated (P < 0.0001). Percentage of CD62-positive monocytes (CD14+ particles) increased from baseline 5% to 13 +/- 6% in ADP-stimulated samples to 53 +/- 17% after isolation (P < 0.001). These findings suggest that density gradient centrifugation activates platelets and that an adhesive interaction between monocytes and platelets may promote TF-PCA expression in isolated mononuclear suspensions. Enhanced monocyte TF expression as a result of an activated platelet-monocyte interaction seems to be an important laboratory effect requiring consideration when utilizing this technique in an experimental setup.


Subject(s)
Blood Platelets/physiology , Monocytes/metabolism , Thromboplastin/metabolism , Cell Communication , Centrifugation, Density Gradient , Humans , Lipopolysaccharide Receptors/analysis , P-Selectin/analysis , Platelet Aggregation
5.
Biochemistry ; 39(37): 11467-76, 2000 Sep 19.
Article in English | MEDLINE | ID: mdl-10985793

ABSTRACT

Tissue factor (TF) is a transmembrane receptor that initiates the thrombogenic cascade by assembly with the serine protease factor VII or VIIa (VII/VIIa) resulting in formation of the bimolecular active complex TF.VIIa. Chemical cross-linking studies identified that a minor population of TF forms dimers on the surface of cells, possibly influencing TF.VIIa proteolytic function as a result of dimerization. We here investigate the effects of dimerization of the extracellular domain of TF on the proteolytic function of the TF. VIIa complex. The leucine zipper dimerization domain of the yeast transcriptional factor GCN4 (LZ) was genetically fused at the C-terminus of the extracellular domain of TF separated by a short linker (TF(L)LZ). TF(L)LZ homodimerized with a K(d) similar to that of the LZ peptide. Tryptophan fluorescence indicated that the two TF moieties were in close proximity and parallel orientation in TF(L)LZ. TF(L)LZ dimers bound two molecules of VIIa, and VIIa binding did not influence the TF dimer equilibrium. Dimerization influenced neither amidolytic nor the factor X activation activities of the TF. VIIa complexes. Notably, dimer TF(L)LZ efficiently promoted the autoactivation of VII to VIIa in solution in contrast to monomeric TF(L)LZ or TF(1)(-)(218). Thus, TF dimerization on cells may serve to "prime" the initiation of the coagulation pathway by generating active TF.VIIa complexes for the subsequent activation of downstream macromolecular substrates.


Subject(s)
DNA-Binding Proteins , Factor VII/metabolism , Factor X/metabolism , Saccharomyces cerevisiae Proteins , Thrombin/metabolism , Thromboplastin/chemistry , Amides/metabolism , Cross-Linking Reagents , Dimerization , Factor VII/chemistry , Factor VIIa/chemistry , Factor VIIa/metabolism , Factor X/chemistry , Factor Xa/chemistry , Factor Xa/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Hydrolysis , Leucine Zippers/genetics , Macromolecular Substances , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding/genetics , Protein Conformation , Protein Kinases/genetics , Protein Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Solubility , Solutions , Thromboplastin/genetics , Thromboplastin/metabolism
7.
Br J Haematol ; 107(4): 747-55, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10606879

ABSTRACT

Leukaemia inhibitory factor (LIF) and interleukin (IL)-6 are members of a cytokine group that share a common signal transducer gp130 and induce pleiotropic biological effects in cells of diverse lineage. In monocytes, LIF facilitates differentiation, which may stimulate the biosynthesis of tissue factor (TF) that initiates the coagulation cascade. We tested the hypothesis that LIF would enhance TF expression in human monocyte-derived macrophages (MDMs). Human peripheral blood mononuclear cells separated from whole blood by density centrifugation were allowed to differentiate into MDMs in primary culture, and were then exposed to LIF, IL-6 and oncostatin M (OSM) for 24 h. LIF and IL-6 receptors, and gp130 were demonstrated in MDMs by immunocytochemistry and RT-PCR. TF procoagulant activity (TF-PCA) was measured by recalcification clotting time and TF protein by Western blotting. The results show that both TF procoagulant activity and TF protein increased significantly in response to LIF over the concentration range of 1-100 nM (P < 0.03). Although OSM and IL-6 tended to enhance TF expression by MDMs, the increase did not reach statistical significance. Anti-LIF receptor and anti-gp130 antibodies attenuated the effect of LIF on TF expression as assayed by both bioassay and flow-cytometry. In conclusion, LIF increases TF-PCA and TF protein in MDMs, and specific anti-LIF receptor antibodies attenuate this effect. Thus, LIF may regulate by a gp130-dependent pathway macrophage-mediated procoagulant function in diverse pathological states involving inflammation and thrombosis and seems to serve as an important mediator at the interface between these processes.


Subject(s)
Antigens, CD/physiology , Growth Inhibitors/pharmacology , Lymphokines/pharmacology , Macrophages/metabolism , Membrane Glycoproteins/physiology , Thromboplastin/metabolism , Blotting, Western , Cell Differentiation , Cytokine Receptor gp130 , Humans , Immunohistochemistry , Interleukin-6/pharmacology , Leukemia Inhibitory Factor , Leukocytes, Mononuclear/cytology , Reverse Transcriptase Polymerase Chain Reaction/methods
8.
J Biol Chem ; 274(34): 24171-5, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10446191

ABSTRACT

The upstream coagulation enzymes are homologous trypsin-like serine proteases that typically function in enzyme-cofactor complexes, exemplified by coagulation factor VIIa (VIIa), which is allosterically activated upon binding to its cell surface receptor tissue factor (TF). TF cooperates with VIIa to create a bimolecular recognition surface that serves as an exosite for factor X binding. This study analyzes to what extent scissile bond docking to the catalytic cleft contributes to macromolecular substrate affinity. Mutation of the P1 Arg residue in factor X to Gln prevented activation by the TF.VIIa complex but did not reduce macromolecular substrate affinity for TF.VIIa. Similarly, mutations of the S and S' subsites in the catalytic cleft of the enzyme VIIa failed to reduce affinity for factor X, although the affinity for small chromogenic substrates and the efficiency of factor X scissile bond cleavage were reduced. Thus, docking of the activation peptide bond to the catalytic cleft of this enzyme-cofactor complex does not significantly contribute to affinity for macromolecular substrate. Rather, it appears that the creation of an extended macromolecular substrate recognition surface involving enzyme and cofactor is utilized to generate substrate specificity between the highly homologous, regulatory proteases of the coagulation cascade.


Subject(s)
Factor VIIa/metabolism , Thromboplastin/metabolism , Binding, Competitive , Catalytic Domain , Factor VIIa/chemistry , Humans , Mutagenesis, Site-Directed , Substrate Specificity , Thromboplastin/chemistry
9.
Biochemistry ; 38(7): 1957-66, 1999 Feb 16.
Article in English | MEDLINE | ID: mdl-10026279

ABSTRACT

Macromolecular substrate docking with coagulation enzyme-cofactor complexes involves multiple contacts distant from the enzyme's catalytic cleft. Here we characterize the binding of the Gla-domain of macromolecular substrate coagulation factor X to the complex of tissue factor (TF) and VIIa. Site-directed mutagenesis of charged residue side chains in the VIIa Gla-domain identified Arg-36 as being important for macromolecular substrate docking. Ala substitution for Arg-36 resulted in an increased KM and a decreased rate of X activation. X with a truncated Gla-domain was activated by mutant and wild-type VIIa at indistinguishable rates, demonstrating that Arg-36 interactions require a properly folded Gla-domain of the macromolecular substrate. VIIa Arg-36 was also required for effective docking of the X Gla-domain in the absence of phospholipid, demonstrating that the Gla-domain of VIIa participates in protein-protein interactions with X. In the absence of TF, the mutant VIIa had essentially normal function, indicating that the cofactor positions VIIa's Gla-domain for optimal macromolecular substrate docking. Computational docking suggests multiple charge complementary contacts of the X Gla-domain with TF.VIIa. A prominent interaction is made by the functionally important X residue Gla-14 with the center of the extended docking site created by residues in the carboxyl module of TF and the contiguous VIIa Gla-domain. These data demonstrate the functional importance of interactions of the Gla-domains of enzyme and substrate, and begin to elucidate the molecular details of the ternary TF.VIIa.X complex.


Subject(s)
1-Carboxyglutamic Acid/metabolism , Arginine/chemistry , Factor VIIa/chemistry , Factor X/metabolism , 1-Carboxyglutamic Acid/chemistry , 1-Carboxyglutamic Acid/genetics , Arginine/genetics , Arginine/metabolism , Endopeptidases , Factor VIIa/genetics , Factor VIIa/metabolism , Factor X/chemistry , Humans , Hydrolysis , Macromolecular Substances , Models, Molecular , Mutagenesis, Site-Directed , Peptide Fragments/metabolism , Phospholipids/metabolism , Protein Binding , Protein Structure, Tertiary , Software , Static Electricity , Substrate Specificity/genetics , Thromboplastin/metabolism
10.
J Mol Biol ; 275(5): 873-94, 1998 Feb 06.
Article in English | MEDLINE | ID: mdl-9480775

ABSTRACT

The tissue factor (TF)-initiated blood coagulation protease cascade can be greatly inhibited in vivo by a potent anti-human-TF monoclonal antibody, 5G9. This antibody binds the carboxyl module of the extracellular domain of TF with a nanomolar binding constant and inhibits the formation of the TF.VIIa.X ternary initiation complex. We have determined the crystal structures of the extra-cellular modules of human TF, Fab 5G9, and their complex (TF.5G9) to 2.4 A, 2. 5 A, and 3.0 A, respectively, and measured the apparent inhibition constants of 5G9 on a panel of TF mutants. In our unliganded TF structure, a 7 degrees change in the relative orientation between the D1 and D2 modules was observed when compared with other published TF structures. Comparison of the free and bound Fab 5G9 indicates that small segmental and side chain variation of the antibody complementarity determining regions occurred on complexation with TF. The antibody-antigen recognition involves 18 TF antigen residues and 19 Fab residues from six CDR with one of the largest buried surface areas seen to date. A combination of structural and mutagenesis data indicate that Tyr156, Lys169, Arg200, and Lys201 play the major role in the antibody recognition. The TF. 5G9 structure provides insights into the mechanism by which the antibody 5G9 inhibits formation of the TF.VIIa.X ternary complex.


Subject(s)
Antigen-Antibody Complex/chemistry , Immunoglobulin Fab Fragments/chemistry , Protein Conformation , Thromboplastin/chemistry , Animals , Blood Coagulation/immunology , Cricetinae , Cricetulus , Crystallography, X-Ray , Humans , Models, Molecular , Mutagenesis
11.
Am J Pathol ; 152(2): 399-411, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9466566

ABSTRACT

Thrombin-catalyzed, cross-linked fibrin (XLF) formation is a characteristic histopathological finding in many human and experimental tumors and is thought to be of importance in the local host defense response. Although the pathogenesis of tumor-associated fibrin deposition is not entirely clear, several tumor procoagulants have been described as likely primary stimuli for the generation of thrombin (and XLF) in the tumor microenvironment (TME). In a previous study of a variety of human tumors we have shown that tissue factor (TF) is the major procoagulant. However, the relative contribution to fibrin deposition in the TME of tumor cell TF and host cell TF (eg, macrophage-derived) was not established. In addition, recent evidence has implicated TF in the regulation of the synthesis of the pro-angiogenic factor vascular endothelial growth factor (VEGF) by tumor cells. In the current study we used in situ techniques to determine the cellular localization of XLF, TF, VEGF, and an alternative tumor procoagulant, so-called cancer procoagulant (CP), a cysteine protease that activates clotting factor X. In lung cancer we have found XLF localized predominantly to the surface of tumor-associated macrophages, as well as to some endothelial cells and perivascular fibroblasts in the stromal area of the tumors co-distributed with TF at the interface of the tumor and host cells. Cancer pro-coagulant was localized to tumor cells in several cases but not in conjunction with the deposition of XLF. TF and VEGF were co-localized in both lung cancer and breast cancer cells by in situ hybridization and immunohistochemical staining. Furthermore, a strong relationship was found between the synthesis of TF and VEGF levels in human breast cancer cell lines (r2 = 0.84; P < 0.0001). Taken together, these data are consistent with a highly complex interaction between tumor cells, macrophages, and endothelial cells in the TME leading to fibrin formation and tumor angiogenesis.


Subject(s)
Blood Coagulation Factors/metabolism , Blood Coagulation/physiology , Breast Neoplasms/physiopathology , Endothelial Growth Factors/metabolism , Lung Neoplasms/physiopathology , Lymphokines/metabolism , Neovascularization, Pathologic/physiopathology , Breast Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
J Biol Chem ; 272(28): 17795-801, 1997 Jul 11.
Article in English | MEDLINE | ID: mdl-9211933

ABSTRACT

Biosynthesis of tumor necrosis factor-alpha (TNF-alpha) is predominantly by cells of the monocytic lineage. This study examined the role of various cis-acting regulatory elements in the lipopolysaccharide (LPS) induction of the human TNF-alpha promoter in cells of monocytic lineage. Functional analysis of monocytic THP-1 cells transfected with plasmids containing various lengths of TNF-alpha promoter localized enhancer elements in a region (-182 to -37 base pairs (bp)) that were required for optimal transcription of the TNF-alpha gene in response to LPS. Two regions were identified: region I (-182 to -162 bp) contained an overlapping Sp1/Egr-1 site, and region II (-119 to -88) contained CRE and NF-kappaB (designated kappaB3) sites. In unstimulated THP-1, CRE-binding protein and, to a lesser extent, c-Jun complexes were found to bind to the CRE site. LPS stimulation increased the binding of c-Jun-containing complexes. In addition, LPS stimulation induced the binding of cognate nuclear factors to the Egr-1 and kappaB3 sites, which were identified as Egr-1 and p50/p65, respectively. The CRE and kappaB3 sites in region II together conferred strong LPS responsiveness to a heterologous promoter, whereas individually they failed to provide transcriptional activation. Furthermore, increasing the spacing between the CRE and the kappaB3 sites completely abolished LPS induction, suggesting a cooperative interaction between c-Jun complexes and p50/p65. These studies indicate that maximal LPS induction of the TNF-alpha promoter is mediated by concerted participation of at least two separate cis-acting regulatory elements.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Immediate-Early Proteins , Lipopolysaccharides/pharmacology , Monocytes/metabolism , NF-kappa B/physiology , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-jun/physiology , Transcription Factors/physiology , Tumor Necrosis Factor-alpha/genetics , Zinc Fingers , Activating Transcription Factor 2 , Binding Sites , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , DNA/metabolism , Early Growth Response Protein 1 , Electrophoresis, Polyacrylamide Gel , Humans , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
13.
Thromb Haemost ; 78(1): 401-5, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9198187

ABSTRACT

Cell surface tissue factor (TF), the major in vivo initiator of coagulation, activates coagulation by binding and allosteric activation of the serine protease factor. VIIa (VIIa). A graphic scheme to account for function of this initial bimolecular activation complex has emerged from the integration of structural with functional analyses. The VIIa light chain, specifically the Gla and EGF-1 domains, form extended hydrophobic contacts with TF which account for most of the free energy of binding. These contacts tether VIIa and facilitate interactions of the protease domain with TF necessary for induction of protease function. Several contact residues in the VIIa protease domain-TF interface are involved in the activation of VIIa by complex allosteric effects. Macromolecular substrate zymogens interact with both the VIIa protease domain and the carboxyl-terminal module of TF. Docking of the VIIa Gla-domain to the latter region of TF appears to contribute to substrate assembly. The current data suggest an extended embrace between TF and VIIa to form the bimolecular enzyme TF.VIIa.


Subject(s)
Blood Coagulation/physiology , Cell Physiological Phenomena , Factor VIIa/physiology , Thromboplastin/physiology , Allosteric Regulation , Factor IX/physiology , Factor VIIa/chemistry , Factor X/physiology , Humans , Protein Binding , Thromboplastin/chemistry
14.
Trends Cell Biol ; 7(5): 180, 1997 May.
Article in English | MEDLINE | ID: mdl-17708939
15.
Biochem J ; 321 ( Pt 3): 787-93, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9032467

ABSTRACT

The C-terminal fibronectin-type-III-like module of the tissue factor (TF) extracellular domain plays a requisite role in the activation of macromolecular substrates by factor VIIa (VIIa) in complex with TF. Unlike the mutations Lys165-->Ala, Lys166-->Ala in TF, which prevent efficient proteolysis of factor X, we found that the coagulant defect of a site-specific Trp158-->Arg, Ser160-->Gly replacement mutant of TF is largely attributable to the inability of TF to efficiently support the activation of the bound zymogen VII to the active protease VIIa. Binding studies demonstrated comparable affinity of binding of VIIa or VII by wild-type TF and TF(R158G160). In comparison with wild-type TF, the catalytic efficiency of factor X activation was reduced 56-fold with TF(A165A166) as the cofactor, but only 3.5-fold with TF(R165G160). The activation of VII bound to TF by factor Xa or VIIa was reduced 2-fold in the presence of TF(R158G160) and 7-8-fold with TF(A165A166). This suggests that the molecular recognition of VII in complex with TF by the enzymes TF-VIIa and factor Xa are similar. Generation of factor IXa by TF(R158G160)-VIIa was unaltered, but reduced 2-fold with TF(A165A166). In addition, the mutations affected the cleavage of the two scissile bonds of factor IX differently, providing further support for the idea that the cofactor, TF, influences the fine specificity of activation of macromolecular substrates by the TF-VIIa complex.


Subject(s)
Mutation/genetics , Thromboplastin/genetics , Blood Coagulation/drug effects , Blood Coagulation/physiology , Electrophoresis, Polyacrylamide Gel , Factor IX/metabolism , Factor VIIa/metabolism , Factor X/metabolism , Humans , Kinetics , Phospholipids/pharmacology , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thromboplastin/chemistry , Thromboplastin/metabolism
16.
Science ; 275(5299): 547-50, 1997 Jan 24.
Article in English | MEDLINE | ID: mdl-8999802

ABSTRACT

Selective occlusion of tumor vasculature was tested as a therapy for solid tumors in a mouse model. The formation of blood clots (thrombosis) within the tumor vessels was initiated by targeting the cell surface domain of human tissue factor, by means of a bispecific antibody, to an experimentally induced marker on tumor vascular endothelial cells. This truncated form of tissue factor (tTF) had limited ability to initiate thrombosis when free in the circulation, but became an effective and selective thrombogen when targeted to tumor endothelial cells. Intravenous administration of the antibody-tTF complex to mice with large neuroblastomas resulted in complete tumor regressions in 38 percent of the mice.


Subject(s)
Blood Coagulation , Immunoconjugates/therapeutic use , Neuroblastoma/blood supply , Neuroblastoma/drug therapy , Thromboplastin/therapeutic use , Animals , Antibodies/immunology , Endothelium, Vascular/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/metabolism , Injections, Intravenous , Mice , Mice, Inbred BALB C , Mice, Nude , Neuroblastoma/pathology , Thromboplastin/administration & dosage , Thromboplastin/metabolism , Tumor Cells, Cultured
17.
J Biol Chem ; 271(45): 28407-13, 1996 Nov 08.
Article in English | MEDLINE | ID: mdl-8910465

ABSTRACT

The binding and assembly of the coagulation proteases on the endothelial cell surface are important steps not only in the generation of thrombin and thrombogenesis, but also in vascular cell signaling. Effector cell protease receptor (EPR-1) was identified as a novel leukocyte cell surface receptor recognizing the coagulation serine protease Factor Xa but not the precursor Factor X. We now demonstrate that EPR-1 is expressed on vascular endothelial cells and smooth muscle cells. Northern blots of endothelial and smooth muscle cells demonstrated three abundant mRNA bands of 3.0, 1.8, and 1.3 kDa. 125I-Labeled Factor Xa bound to endothelial cells in a dose-dependent saturable manner, and the binding was inhibited by antibody to EPR-1. No specific binding was observed with a recombinant mutant Factor X in which the activation site was substituted by Arg196 --> Gln to prevent the proteolytic conversion to Xa. EPR-1 was identified immunohistochemically on microvascular endothelial and smooth muscle cells. Functionally, exposure of smooth muscle cells or endothelial cells to Factor Xa induced a 3-fold and a 2-fold increase in [3H]thymidine uptake, respectively. However, receptor occupancy alone is insufficient for mitogenic signaling because the active site of the enzyme is required for mitogenesis. Thus, EPR-1 represents a site of specific protease-receptor complex assembly, which during local initiation of the coagulation cascade could mediate cellular signaling and responses of the vessel wall.


Subject(s)
Factor Xa/metabolism , Receptors, Cell Surface/metabolism , Serine Endopeptidases/metabolism , Arthropod Proteins , Blotting, Western , Endothelium, Vascular/metabolism , Factor Xa Inhibitors , Flow Cytometry , Humans , Inhibitor of Apoptosis Proteins , Intercellular Signaling Peptides and Proteins , Mitosis/drug effects , Muscle, Smooth/metabolism , Peptides/pharmacology , RNA, Messenger/metabolism , Serine Proteinase Inhibitors/pharmacology , Survivin
18.
Biochem J ; 315 ( Pt 1): 145-51, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8670099

ABSTRACT

The coagulation pathways are initiated by the cell-surface receptor Tissue Factor (TF), which binds the serine proteinase coagulation Factor VIIa (VIIa), resulting in enhanced catalytic function, both amidolytic, towards small pseudo-substrates, and proteolytic, towards macromolecular substrates. Here we implicate Asp44 in TF as a ligand-interactive residue that, in contrast with previously characterized binding residues, is involved in the enhancement of VIIa catalytic function. Whereas charge neutralization by replacement of Asp44 with Asn did not reduce function of human TF, the exchange by Ala resulted in mutants with 8-fold reduced affinity for binding of VIIa. Enhancement of VIIa amidolytic function by TF Ala44 was reduced by 20-25% relative to wild-type and support of proteolytic function was diminished 6-fold indicating that this cofactor residue is significantly enhancing proteolysis of the macromolecular substrate by VIIa. Replacement of Asp44 by Glu, Thr, and Arg exhibited a less severe phenotype with an approx. 4-fold reduced affinity for VIIa and a 2-3 fold diminished activation of Factor X. The improved activity of these mutants as compared with the Ala replacement is consistent with functional importance of an extended side chain at this position. The specific influence of the Asp44 exchange on catalytic function of the TF x VIIa complex indicates fine specificity of the TF ligand interface in mediating receptor and cofactor function.


Subject(s)
Aspartic Acid/metabolism , Factor VIIa/metabolism , Thromboplastin/physiology , Alanine/metabolism , Animals , Binding, Competitive , CHO Cells/metabolism , CHO Cells/physiology , Catalysis , Cricetinae , DNA/genetics , Humans , Kinetics , Mutation , Peptide Hydrolases/metabolism , Solubility , Structure-Activity Relationship , Thromboplastin/genetics , Thromboplastin/metabolism
19.
Coron Artery Dis ; 7(1): 57-62, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8773434

ABSTRACT

BACKGROUND: Rethrombosis limits the efficacy of coronary thrombolysis and may result from surface-associated thrombin, de-novo prothrombin activation, or both. This study was designed to determine the relative roles of thrombin, factor Xa, and the complex of tissue factor and factor VIIa in the procoagulant activity on injured arteries with evolving thrombi. METHODS: Extensive vascular injury and platelet-rich thrombi were induced in the abdominal aorta of 25 anesthetized rabbits by applying anodal current through a transluminal electrode for 3 h. Injured vessel segments were excised and placed in a chamber permitting perfusion over the luminal surface and associated thrombus. RESULTS: Vessel segments perfused with recalcified, citrated human plasma induced marked increases in the concentration of fibrinopeptide A, a marker of thrombin-induced fibrin formation, in the effluent plasma after 10 min (4636 +/- 1894% of fibrinopeptide A in the nonperfused plasma, n = 5). Perfusion with plasma depleted of vitamin K-dependent coagulation factors prevented the increase in fibrinopeptide A (122 +/- 30%, n = 4), indicating the lack of preformed functional thrombin. Furthermore, appearance of fibrinopeptide A was attenuated by perfusion with plasma containing 0.1 mumol/l recombinant tick anticoagulant peptide, a specific inhibitor of factor Xa (594 +/- 320%, n = 3), and by preincubation of vessel segments with a monoclonal antibody to rabbit tissue factor (438 +/- 220%, n = 3). CONCLUSIONS: Procoagulant activity on injured vessels and associated thrombi is mediated by factor Xa, a product of the functional initiation of coagulation by factor VIIa associated with tissue factor. Accordingly, inhibition of tissue factor-mediated coagulation may be effective for attenuation of active thrombogenesis on injured vessels and during thrombolysis.


Subject(s)
Blood Coagulation/physiology , Factor VIIa/physiology , Factor Xa/physiology , Prothrombin/physiology , Thrombin/physiology , Thromboplastin/physiology , Thrombosis/blood , Animals , Aorta, Abdominal/injuries , Aorta, Abdominal/pathology , Coronary Thrombosis/blood , Disease Models, Animal , Fibrinopeptide A/physiology , Humans , Microscopy, Electron, Scanning , Rabbits , Thrombolytic Therapy , Thrombosis/pathology
20.
Circulation ; 92(11): 3323-30, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7586321

ABSTRACT

BACKGROUND: Activation of coagulation has been implicated in both acute thrombotic occlusion and restenosis after balloon angioplasty. However, concomitant administration of antithrombotic agents has thus far failed to prevent these complications. Importantly, the factors contributing to procoagulant activity of balloon-injured arteries over time have not been defined. This study was designed to determine the duration of procoagulant activity on the luminal surface of balloon-injured arteries and the relative roles of tissue factor and thrombin in this response. METHODS AND RESULTS: Abdominal aortas in rabbits were subjected to repetitive balloon hyperinflations sufficient to disrupt the internal elastic lamina. Aortas were excised at < 1, 2, 4, 8, 16, 24, 48, and 72 hours and 1, 2, and 4 weeks after injury; divided into segments; and perfused with recalcified human pooled plasma (n = 58) or plasma depleted of vitamin K-dependent coagulation factors (n = 27) or first incubated with a monoclonal antibody to rabbit tissue factor (n = 33) followed by perfusion with human plasma. Samples of the effluent and plasma perfusate were collected over 10 minutes and assayed for fibrinopeptide A (FPA) as an index of the rate of thrombin-induced fibrin formation. FPA in the effluent from segments perfused with recalcified plasma, expressed as a percentage of FPA in the perfusate, was elevated for 16 hours after balloon-induced injury and exhibited two distinct increases occurring < 1 hour (1297 +/- 473%, mean +/- SD, n = 5) and 8 hours (1052 +/- 330%, n = 6) after injury (P < or = .000001 versus uninjured vessels). Preincubation of segments at these intervals with an antibody to tissue factor markedly attenuated the increases in FPA, as did perfusion of segments with plasma depleted of vitamin K-dependent coagulation factors, indicating that the observed increases in FPA in whole plasma did not result from performed thrombin bound to the injured vessel wall. CONCLUSIONS: Tissue factor-mediated coagulation appears to be primarily responsible for prolonged procoagulant activity of balloon-injured arteries.


Subject(s)
Angioplasty, Balloon/adverse effects , Aorta, Abdominal/injuries , Blood Coagulation/physiology , Fibrinopeptide A/metabolism , Thromboplastin/physiology , Animals , Enzyme-Linked Immunosorbent Assay , Humans , Plasma , Rabbits , Radioimmunoassay , Thrombin/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...