1
|
Conway EM. Thrombomodulin and its role in inflammation. Semin Immunopathol 2012; 34:107-25. [PMID: 21805323 DOI: 10.1007/s00281-011-0282-8] [Citation(s) in RCA: 219] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 07/20/2011] [Indexed: 12/30/2022]
Abstract
The goal is to provide an extensive review of the physiologic role of thrombomodulin (TM) in maintaining vascular homeostasis, with a focus on its anti-inflammatory properties. Data were collected from published research. TM is a transmembrane glycoprotein expressed on the surface of all vascular endothelial cells. Expression of TM is tightly regulated to maintain homeostasis and to ensure a rapid and localized hemostatic and inflammatory response to injury. By virtue of its strategic location, its multidomain structure and complex interactions with thrombin, protein C (PC), thrombin activatable fibrinolysis inhibitor (TAFI), complement components, the Lewis Y antigen, and the cytokine HMGB1, TM exhibits a range of physiologically important anti-inflammatory, anti-coagulant, and anti-fibrinolytic properties. TM is an essential cofactor that impacts on multiple biologic processes. Alterations in expression of TM and its partner proteins may be manifest by inflammatory and thrombotic disorders. Administration of soluble forms of TM holds promise as effective therapies for inflammatory diseases, and infections and malignancies that are complicated by disseminated intravascular coagulation.
Collapse
Affiliation(s)
- Edward M Conway
- Division of Hematology-Oncology, Department of Medicine, Centre for Blood Research (CBR), University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
2
|
White MC, McHowat J. Protease activation of calcium-independent phospholipase A2 leads to neutrophil recruitment to coronary artery endothelial cells. Thromb Res 2006; 120:597-605. [PMID: 17188740 PMCID: PMC2170458 DOI: 10.1016/j.thromres.2006.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Revised: 10/18/2006] [Accepted: 11/14/2006] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Thrombin or tryptase cleavage of protease-activated receptors (PAR) on human coronary artery endothelial cells (HCAEC) results in activation of a membrane-associated, calcium-independent phospholipase A2 (iPLA2) that selectively hydrolyzes plasmalogen phospholipids. Atherosclerotic plaque rupture results in a coronary ischemic event in which HCAEC in the ischemic area would be exposed to increased thrombin concentrations in addition to tryptase released by activated mast cells present in the plaque. MATERIALS AND METHODS HCAEC were stimulated with thrombin or tryptase in the absence or presence of bromoenol lactone (BEL), a selective iPLA2 inhibitor, and iPLA2 activation, accumulation of biologically active membrane phospholipid-derived metabolites, upregulation of cell surface P-selectin expression and neutrophil adherence were measured. RESULTS HCAEC exposed to thrombin or tryptase stimulation demonstrated an increase in iPLA2 activity and arachidonic acid release. Additionally, stimulated HCAEC demonstrated increased platelet-activating factor (PAF) production and cell surface P-selectin expression, resulting in increased adhesion of neutrophils to HCAEC monolayers. Pretreatment with bromoenol lactone to inhibit iPLA2, blocked membrane phospholipid-derived metabolite production, increased cell surface P-selectin expression and neutrophil adherence. CONCLUSIONS The similar biochemical and cellular responses in HCAEC exposed to thrombin or tryptase stimulation suggest that the cleavage of two separate PAR serve to extend the range of proteases to which the cells respond rather than resulting in separate intracellular events. This suggests that in conditions such as thrombosis and atherosclerosis that multiple mechanisms can activate the inflammatory response.
Collapse
Affiliation(s)
- Maureen C White
- Saint Louis University School of Medicine, Department of Pathology, 1402 S. Grand, St. Louis, MO 63104, United States.
| | | |
Collapse
|
3
|
Abstract
The coagulation cascade and protease-activated receptors (PARs) together provide an elegant mechanism that links mechanical information in the form of tissue injury to cellular responses. These receptors appear to largely account for the cellular effects of thrombin and can mediate signaling to other trypsin-like proteases. An important role for PARs in hemostasis and thrombosis is established in animal models, and studies in knockout mice and nonhuman primates raise the question of whether PAR inhibition might offer an appealing new approach to the prevention and treatment of thrombosis. PARs may also trigger inflammatory responses to tissue injury. For example, PAR activation on endothelial cells and perhaps sensory afferents can trigger local accumulation of leukocytes and platelets and transudation of plasma. However, panoply of signaling systems and cell types orchestrates inflammatory responses, and efforts to define the relative importance and roles of PARs in various inflammatory processes are just beginning. Lastly, roles for PARs in blood vessel formation and other processes during embryonic development are emerging, and whether these reflect new roles for the coagulation cascade and/or PAR signaling to other proteases remains to be explored.
Collapse
Affiliation(s)
- S R Coughlin
- Cardiovascular Research Institute, Department of Medicine, University of California, San Francisco, CA 94143-0130, USA.
| |
Collapse
|
4
|
Szaba FM, Smiley ST. Roles for thrombin and fibrin(ogen) in cytokine/chemokine production and macrophage adhesion in vivo. Blood 2002; 99:1053-9. [PMID: 11807012 PMCID: PMC3150214 DOI: 10.1182/blood.v99.3.1053] [Citation(s) in RCA: 220] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Extravascular coagulation leading to fibrin deposition accompanies many immune and inflammatory responses. Although recognized by pathologists for decades, and probably pathologic under certain conditions, the physiologic functions of extravascular coagulation remain to be fully defined. This study demonstrates that thrombin can activate macrophage adhesion and prompt interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) production in vivo. Peritoneal macrophages were elicited with thioglycollate (TG) and then activated in situ, either by intraperitoneal injection of lipopolysaccharide (LPS) or by injection of antigen into mice bearing antigen-primed T cells. Others previously established that such treatments stimulate macrophage adhesion to the mesothelial lining of the peritoneal cavity. The present study demonstrates that thrombin functions in this process, as macrophage adhesion was suppressed by Refludan, a highly specific thrombin antagonist, and induced by direct peritoneal administration of purified thrombin. Although recent studies established that protease activated receptor 1 (PAR-1) mediates some of thrombin's proinflammatory activities macrophage adhesion occurred normally in PAR-1-deficient mice. However, adhesion was suppressed in fibrin(ogen)-deficient mice, suggesting that fibrin formation stimulates macrophage adhesion in vivo. This study also suggests that fibrin regulates chemokine/cytokine production in vivo, as direct injection of thrombin stimulated peritoneal accumulation of IL-6 and MCP-1 in a fibrin(ogen)-dependent manner. Given that prior studies have clearly established inflammatory roles for PAR-1, thrombin probably has pleiotropic functions during inflammation, stimulating vasodilation and mast cell degranulation via PAR-1, and activating cytokine/chemokine production and macrophage adhesion via fibrin(ogen).
Collapse
Affiliation(s)
- Frank M Szaba
- Trudeau Institute, 100 Algonquin Ave, Saranac Lake, NY 12983, USA
| | | |
Collapse
|
5
|
Tsunoda S, Yamabe H, Osawa H, Kaizuka M, Shirato K, Okumura K. Cultured rat glomerular epithelial cells show gene expression and production of transforming growth factor-beta: expression is enhanced by thrombin. Nephrol Dial Transplant 2001; 16:1776-82. [PMID: 11522858 DOI: 10.1093/ndt/16.9.1776] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Glomerular crescents play an important role in progressive glomerular injury. The lesions consist of epithelial cells, macrophages, and deposits of fibrin and extracellular matrix. Transforming growth factor beta (TGF-beta) contributes to the modulation of cell growth and extracellular matrix synthesis. Thrombin is involved in fibrin formation in crescents. The purpose of this study was to examine whether glomerular epithelial cells (GEC) could produce TGF-beta, and if so, to clarify the role of TGF-beta in GEC proliferation. We also investigated whether thrombin could modulate the production of TGF-beta and extracellular matrix by GEC. METHODS Bioassay using the TGF-beta-dependent mink pulmonary epithelial cell line (CCL-64), immunoblot analysis, and reverse transcriptase polymerase chain reaction (RT-PCR) were used to demonstrate TGF-beta production by rat GEC. TGF-beta gene expression was examined by RT-PCR in GEC incubated with thrombin, and type IV collagen and fibronectin were quantified by enzyme immunoassay in culture supernatants of GEC incubated with thrombin or TGF-beta. RESULTS TGF-beta activity was demonstrated in GEC supernatants by bioassay. Immunoblot analysis of concentrated culture supernatants using anti-TGF-beta antibody revealed a 12.5-kDa protein, which was compatible with TGF-beta. Concentrated GEC supernatants inhibited GEC proliferation as well as porcine TGF-beta. RT-PCR demonstrated TGF-beta gene expression in GEC. Thrombin (0.5-5.0 U/ml) enhanced TGF-beta mRNA expression in a dose-dependent manner. Thrombin (5.0 U/ml) and porcine TGF-beta (5.0 ng/ml) stimulated the production of type IV collagen and fibronectin by GEC. CONCLUSIONS Rat GEC produce TGF-beta in vitro. Thrombin may participate in the progression of glomerulosclerosis in crescentic glomerulonephritis through the stimulation of TGF-beta production by GEC.
Collapse
Affiliation(s)
- S Tsunoda
- Second Department of Internal Medicine, Hirosaki University School of Medicine, Zaifu-cho 5, Hirosaki 036-8216, Japan
| | | | | | | | | | | |
Collapse
|
6
|
Robey RB, Raval BJ, Ma J, Santos AV. Thrombin is a novel regulator of hexokinase activity in mesangial cells. Kidney Int 2000; 57:2308-18. [PMID: 10844601 DOI: 10.1046/j.1523-1755.2000.00091.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Hexokinase (HK) activity is fundamentally important to cellular glucose uptake and metabolism. Phorbol esters increase both HK activity and glucose utilization in cultured mesangial cells via a protein kinase C (PKC)- and extracellular signal-regulated kinases 1 and 2 (ERK1/2)-dependent mechanism. In adult kidneys, increased HK activity has been reported in both glomerular injury and in diabetes, but the mechanisms responsible for these changes are unknown. Thrombin, a known activator of both PKC and ERK1/2, is increased in the settings of renal injury and diabetes. Thus, thrombin may contribute to the observed changes in HK activity in vivo. METHODS Thrombin and thrombin receptor agonists were tested for the ability to increase HK activity and glucose metabolism in murine mesangial (SV40 MES 13) cells. ERK1/2 activation was also evaluated in parallel. Thrombin inhibition (hirudins), PKC depletion, Ser-Thr kinase inhibition (H-7), MEK1/2 inhibition (PD98059), pertussis toxin (PTX), and general inhibitors of transcription or translation were then tested for the ability to attenuate these effects. RESULTS Thrombin (>/=0.01 U/mL) mimicked the effect of phorbol esters, increasing HK activity> 50% within 12 to 24 hours (P < 0.05). This effect was inhibited by hirudins, mimicked by thrombin receptor agonists, and accompanied by increased Glc utilization. H-7, PD98059, and general inhibitors of transcription or translation-but not PTX-prevented thrombin-induced HK activity at 24 hours. PKC depletion and PD98059 also blocked the associated phosphorylation and activation of ERK1/2. CONCLUSIONS Thrombin increases mesangial cell HK activity via a PTX-insensitive mechanism involving thrombin receptor activation, PKC-dependent activation of ERK1/2, and both ongoing gene transcription and de novo protein synthesis. As such, thrombin is a novel regulator of HK activity in mesangial cells and may play a role in coupling renal injury to metabolism.
Collapse
Affiliation(s)
- R B Robey
- Department of Medicine, Section of Nephrology, University of Illinois at Chicago College of Medicine and VA Chicago Health Care System, West Side Division, Chicago, Illinois 60612-7315, USA.
| | | | | | | |
Collapse
|
7
|
Gentry PA, Plante L, Schroeder MO, LaMarre J, Young JE, Dodds WG. Human ovarian follicular fluid has functional systems for the generation and modulation of thrombin. Fertil Steril 2000; 73:848-54. [PMID: 10731552 DOI: 10.1016/s0015-0282(99)00635-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To determine whether prothrombin is present in follicular fluid and whether the enzymatic pathways for prothrombin activation are similar to those in plasma. DESIGN Follicular fluid samples collected at the time of oocyte harvest for an assisted reproductive technology procedure (ART) were analyzed for a panel of hemostatic proteins with use of a combination of functional, chromogenic, and Western ligand blot analysis. SETTING An ART clinic and an academic research laboratory. PATIENT(S) Women undergoing ART. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Determination of components of thrombin generation and thrombin modulatory systems using functional and antigenic assay procedures. RESULT(S) Both prothrombin and components of the prothrombinase enzyme complex, which includes factors V, VII, and X, are present in follicular fluid. Other hemostatic proteins, including factors VIII and IX and vonWillebrand factor, are absent. The direct activation of prothrombin to thrombin is similar in follicular fluid and plasma. Like plasma, inhibitors of both thrombin and thrombin generation, including antithrombin, protein C, and alpha2-macroglobulin, are present in follicular fluid. CONCLUSION(S) Only a select group of hemostatic plasma proteins are present in follicular fluid. There is no direct correlation between molecular size and concentration of individual proteins in follicular fluid. These results indicate that the proteins involved in the thrombin-generating and thrombin modulatory pathways may be derived from ovarian cells, suggesting that thrombin may have a role in folliculogenesis.
Collapse
Affiliation(s)
- P A Gentry
- Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
8
|
Kaizuka M, Yamabe H, Osawa H, Okumura K, Fujimoto N. Thrombin stimulates synthesis of type IV collagen and tissue inhibitor of metalloproteinases-1 by cultured human mesangial cells. J Am Soc Nephrol 1999; 10:1516-23. [PMID: 10405207 DOI: 10.1681/asn.v1071516] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Glomerular accumulation of extracellular matrix (ECM) is the common pathologic feature following glomerular injury, and the alteration in the synthesis and degradation of ECM may be involved in the glomerular accumulation of ECM. Glomerular fibrin formation occurs in various forms of human and experimental glomerulonephritis, and it may play an important role in progressive glomerular injury. Thrombin, a multifunctional serine proteinase that is generated at the site of vascular injury, has central functions in hemostasis and it also shows various biologic effects. In this study, it is hypothesized that thrombin may alter the production and the degradation of type IV collagen, which is an important component of ECM in the glomeruli. Human mesangial cells (HMC) were cultured, and the levels of type IV collagen, tissue inhibitor of metalloproteinase-1 (TIMP-1), and matrix metalloproteinase-2 (MMP-2) in the culture supernatants were measured by enzyme immunoassay using specific antibodies. MMP-2 activity was also evaluated by zymography using polyacrylamide/ sodium dodecyl sulfate gel-containing gelatin. Thrombin increased the production of type IV collagen and TIMP-1 in a dose-and time-dependent manner, but it did not increase MMP-2. Thrombin also stimulated the gene expressions of the type IV collagen and TIMP-1 in HMC in a dose- and time-dependent manner. Thrombin treated with diisopropylfluorophosphate, a serine proteinase inhibitor, did not show any of these effects. Hirudin, a natural thrombin inhibitor, and anti-transforming growth factor-beta-neutralizing antibody inhibited the stimulating effect of thrombin. These findings suggest that thrombin may contribute to the excessive accumulation of ECM and progression of glomerulosclerosis through an increase of type IV collagen production and a decreased matrix degradation presumably via a transforming growth factor-beta-dependent mechanism.
Collapse
Affiliation(s)
- M Kaizuka
- Second Department of Internal Medicine, Hirosaki University School of Medicine, Japan
| | | | | | | | | |
Collapse
|
9
|
Sawada M, Yanamoto H, Nagata I, Hashimoto N, Nakahara I, Akiyama Y, Kikuchi H, Macdonald RL. Prevention of neointimal formation by a serine protease inhibitor, FUT-175, after carotid balloon injury in rats. Stroke 1999; 30:644-50. [PMID: 10066865 DOI: 10.1161/01.str.30.3.644] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE In vivo and vitro studies revealed the activation of thrombin and the complement system in vascular lesion formation during the process of atherosclerosis, along with pathological proliferation of smooth muscle cells. We examined the effect of the synthetic serine protease inhibitor FUT-175 (developed as a potent inhibitor of thrombin and the complement system) on vascular lesions using balloon dilatation-induced neointimal formation in the carotid artery of rats. METHODS Sprague-Dawley (SD) rats underwent balloon dilatation injury of the left carotid artery to induce neointimal formation. Three groups of these rats (n=8, each) were treated with daily intraperitoneal injections of 1 of the following doses of FUT-175: 0.5, 1.0, or 2.0 mg/d in 1 mL of saline for 7 consecutive days. The control group (n=8) was similarly treated with 1 mL of saline for 7 days. The injections were started immediately after balloon injury. Two weeks after the injury, the left carotid arteries were perfusion-fixed, and the areas of the neointimal and medial layer were analyzed under a microscope. RESULTS A morphometric analysis revealed that there were significant differences in the intima-media ratio between the 4 groups treated with vehicle (saline) or a low, medium, or high dose of FUT-175 (1.45+/-0.11, 1.08+/-0.06, 0.71+/-0.04, or 0.32+/-0.04, respectively). This suppression was achieved in a dose-dependent manner by the administration of FUT-175 after balloon injury. In the histological study, it was demonstrated that FUT-175 suppresses the production of platelet-derived growth factor (PDGF)-BB in the neointima and the medial smooth muscle cell layer. CONCLUSIONS After balloon injury activated proteases that were inhibited by FUT-175 were demonstrated to have an essential role in the development of the pathological thickening of the arterial wall.
Collapse
Affiliation(s)
- M Sawada
- Department of Cerebrovascular Surgery, Laboratory for Cerebrovascular Disorders of Neurosurgery, Kyoto University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Vaingankar SM, Martins-Green M. Thrombin aivation of the 9E3/CEF4 chemokine involves tyrosine kinases including c-src and the epidermal growth factor receptor. J Biol Chem 1998; 273:5226-34. [PMID: 9478978 DOI: 10.1074/jbc.273.9.5226] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The 9E3/CEF4 gene codes for a chemokine that is highly homologous to human interleukin-8 and melanoma growth-stimulating activity/groalpha. These chemokines belong to a family of molecular mediators that are importantly involved in inflammation, wound healing, tumor development, and viral entry into cells. On the chorioallantoic membrane the 9E3 protein is chemotactic for monocyte/macrophages and lymphocytes and is angiogenic. In cultured chicken embryo fibroblasts, which have many of the properties of wound fibroblasts, the gene is stimulated by a variety of agents including oncogenes, growth factors, phorbol esters, and thrombin. The strong stimulation of 9E3 by thrombin in culture correlates well with the observation that in young chicks this gene is stimulated to very high levels in fibroblasts upon wounding and remains high throughout wound repair. Activation of 9E3 by thrombin: (i) occurs very rapidly, one minute exposure to thrombin is sufficient to initiate the signals necessary for gene activation; (ii) is independent of mitogenesis; (iii) operates through the proteolytically activated receptor for thrombin; (iv) is mediated by tyrosine kinases, including c-src and the epidermal growth factor (EGF) receptor, rather than Ser/Thr kinases such as protein kinase C and protein kinase A. Inhibition of either c-src or the EGF receptor tyrosine kinase inhibits the stimulation of 9E3 by thrombin. We show here for the first time that activation of the EGF receptor through a cell-surface receptor that does not have tyrosine kinase activity can lead to expression of an immediate early response gene which encodes for a secreted protein, a chemokine. This rapidly activated tyrosine kinase pathway may be a general stress response by which in vivo a localized cell population reacts to emergency situations such as viral infection, wounding, or tumor growth.
Collapse
Affiliation(s)
- S M Vaingankar
- Department of Biology, University of California, Riverside, California 92521, USA
| | | |
Collapse
|
11
|
Smiley ST, Stitt TN, Grusby MJ. Cross-linking of protein S bound to lymphocytes promotes aggregation and inhibits proliferation. Cell Immunol 1997; 181:120-6. [PMID: 9398399 DOI: 10.1006/cimm.1997.1210] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recently, we reported that expression of the anticoagulant protein S is IL-4-inducible in primary T cells, and that protein S inhibits lymphoid cell procoagulant activity. Here, using a flow cytometric assay, we demonstrate that protein S binds to the surface of B and T lymphocytes. In addition, we show that cross-linking of protein S bound to lymphocytes induces aggregation and inhibits growth in cultures of primary B and T lymphocytes. Thus, our studies suggest that protein S is an IL-4-inducible T cell product that can affect B and T cell growth and aggregation via a lymphocyte protein S receptor. Interestingly, protein S joins thrombin and factor Xa as coagulation factors that modulate lymphocyte activation, suggesting that the clotting pathway may regulate wound-related inflammatory responses.
Collapse
Affiliation(s)
- S T Smiley
- Department of Cancer Biology, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
12
|
Smiley ST, Boyer SN, Heeb MJ, Griffin JH, Grusby MJ. Protein S is inducible by interleukin 4 in T cells and inhibits lymphoid cell procoagulant activity. Proc Natl Acad Sci U S A 1997; 94:11484-9. [PMID: 9326636 PMCID: PMC23513 DOI: 10.1073/pnas.94.21.11484] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Extravascular procoagulant activity often accompanies cell-mediated immune responses and systemic administration of pharmacologic anticoagulants prevents cell-mediated delayed-type hypersensitivity reactions. These observations suggest a direct association between coagulation and cell-mediated immunity. The cytokine interleukin (IL)-4 potently suppresses cell-mediated immune responses, but its mechanism of action remains to be determined. Herein we demonstrate that the physiologic anticoagulant protein S is IL-4-inducible in primary T cells. Although protein S was known to inhibit the classic factor Va-dependent prothrombinase assembled by endothelial cells and platelets, we found that protein S also inhibits the factor Va-independent prothrombinase assembled by lymphoid cells. Thus, protein S-mediated down-regulation of lymphoid cell procoagulant activity may be one mechanism by which IL-4 antagonizes cell-mediated immunity.
Collapse
Affiliation(s)
- S T Smiley
- Department of Cancer Biology, Harvard School of Public Health, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
13
|
Guitteny AF, Herbert JM. Failure of heparin to inhibit the expression of the thrombin receptor following endothelial injury of the rabbit carotid artery. Eur J Pharmacol 1997; 327:157-62. [PMID: 9200554 DOI: 10.1016/s0014-2999(97)89655-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The effect of heparin on thrombin receptor expression was evaluated in an experimental model of myointimal smooth muscle cell proliferation in rabbits. Myointimal hyperplasia was induced by an air-drying injury of the carotid artery and thrombin receptor expression following endothelial injury was measured by in situ hybridisation and immunohistochemistry. In healthy arteries, thrombin receptor mRNA and protein were detected in the endothelial cells only. In contrast, 14 days after endothelial injury, thrombin receptor mRNA expression increased in the smooth muscle cells present in the neointima, predominantly in areas of active cell proliferation. A 2-week subcutaneous treatment with heparin (10 mg/kg per day, s.c.) inhibited smooth muscle cell hyperplasia occurring in the intima following deendothelialization (80 +/- 7.8% inhibition, P < 0.001). The 14-day heparin treatment strongly reduced thrombin receptor gene and protein expression observed in the endothelial cells in healthy arteries but did not affect thrombin receptor expression which occurred in smooth muscle cells which have proliferated in the neointima as a consequence of endothelial injury. These results therefore establish that thrombin receptor expression during intimal hyperplasia is an heparin-insensitive event.
Collapse
MESH Headings
- Animals
- Anticoagulants/pharmacology
- Carotid Arteries/drug effects
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carotid Artery Injuries
- Cell Division
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/injuries
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Heparin/pharmacology
- Hyperplasia
- In Situ Hybridization, Fluorescence
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- RNA, Messenger/metabolism
- Rabbits
- Receptors, Thrombin/genetics
- Receptors, Thrombin/metabolism
Collapse
Affiliation(s)
- A F Guitteny
- Haemobiology Research Department, Sanofi Recherche, Toulouse, France
| | | |
Collapse
|
14
|
Hajjar DP, Nicholson AC. Viral activation of coagulation: implications for thrombosis and atherosclerosis. Ann N Y Acad Sci 1997; 811:155-65; discussion 165-7. [PMID: 9186594 DOI: 10.1111/j.1749-6632.1997.tb51998.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- D P Hajjar
- Department of Pathology, Cornell University Medical College, New York, New York 10021, USA
| | | |
Collapse
|
15
|
The Amino Terminal Lectin-Like Domain of Thrombomodulin Is Required for Constitutive Endocytosis. Blood 1997. [DOI: 10.1182/blood.v89.2.652] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThrombomodulin (TM) is a multidomain protein that serves as a cofactor in a major natural anticoagulant system. To further characterize the structure-function of TM, we have transfected COS cells with different truncated forms of TM. In the first form, COS cells expressing TM that lacks the putative signal peptide (17 residues); the lectin-like, hydrophobic N-terminal domain (226 residues); and 12 residues of the first epidermal growth factor (EGF )-like repeat (COSdel.238 cells) were found to function normally with respect to TM transport to the cell surface and thrombin-dependent protein C activation. However, in contrast to wild-type TM, as visually studied by immunofluorescence and immunogold electron microscopy, the COSdel.238 cells did not constitutively internalize anti-TM–TM or thrombin-TM complexes. To identify the region responsible for mediating the endocytic process, deletant forms of TM lacking either the lectin-like region (residues 2-155) or the hydrophobic region of the N-terminal domain (residues 161-202) were expressed in COS cells (COSdel.2-155 and COSdel.161-202, respectively). Protein C cofactor activity was maintained in both cells. Although the COSdel.161-202 cells behaved similarly to wild-type TM-transfected cells, visual studies showed a lack of constitutive internalization of thrombin-TM or anti-TM–TM complexes in the COSdel.2-155 cells. We conclude that the lectin-like domain of human TM serves to regulate cell surface expression of TM via the endocytic route and therefore may also play a major physiologic role in controlling intracellular and extracellular accumulation of thrombin in a variety of biologic systems.
Collapse
|
16
|
Nicholson AC, Nachman RL, Altieri DC, Summers BD, Ruf W, Edgington TS, Hajjar DP. Effector cell protease receptor-1 is a vascular receptor for coagulation factor Xa. J Biol Chem 1996; 271:28407-13. [PMID: 8910465 DOI: 10.1074/jbc.271.45.28407] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
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.
Collapse
Affiliation(s)
- A C Nicholson
- Department of Pathology, Cornell University Medical College, New York, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The control of cell proliferation by thrombin was studied in vitro in cultured epithelial and stromal cells of the endometrium. The effect of thrombin was studied after chronic treatment (72 hr) in medium containing 10% fetal bovine serum (FBS) combined or not with sex steroids. Thrombin inhibited slightly the proliferation (based on DNA measurements) only in epithelial cells (P < 0.05). 17 beta-estradiol (E) and progesterone (P4) had no mitogenic effects. The presence of functional thrombin receptors was estimated by stimulation of second messenger generation in response to increasing doses of thrombin (0-1,500 ng/ml). In confluent cultures of epithelial cells, the addition of thrombin for 10 min stimulated cAMP production by 50% with a maximal response at 500 ng/ml (P < 0.05). Similarly, in stromal cells, thrombin stimulated cAMP production in a dose-dependent manner (P < 0.01). Generation of inositol-phosphates was also stimulated by 50% in epithelial cells (P < 0.03), with a maximal response at 500 ng/ml, and by 45% in stromal cells (P < 0.01), with a maximal response at 50 ng/ml. The effect of thrombin on cell proliferation was investigated by 3H-thymidine incorporation in serum-free medium for 24 hr. Thrombin inhibited incorporation in epithelial cells (P < 0.0001) in a dose-dependent manner. Conversely, thrombin stimulated significantly incorporation of stromal cells (P < 0.05) at 50 ng/ml. The effect of sex steroids was also evaluated and it was found that E had no effect on cell proliferation, while P4 inhibited the incorporation in both epithelial (P < 0.001) and stromal cells (P < 0.001). The effect of a combined treatment with thrombin and E inhibited both epithelial (P < 0.001) and stromal cell (P < 0.001) growth, but a combination of thrombin and P4 had no additional effect on growth compared to P4 alone. Further investigation of the role of thrombin has been carried out by measuring prostaglandin (PG) responses. Addition of thrombin for 24 hr inhibited PGF2 alpha production by epithelial cells (P < 0.0001) but had no effect on PGE2 production by stromal cells. Therefore, functional receptors for thrombin appear to be present in epithelial and stromal cells of the bovine endometrium. The minimal effect of thrombin alone or in combination with sex steroids on endometrial cell proliferation in vitro combined with the evidence of functional thrombin receptor in these cells, suggest that: (1) the effect of sex steroids in cultured endometrial cells is not modulated by the presence of thrombin, and (2) other factors are necessary for the full expression of mitogenic responses to sex steroids in vitro.
Collapse
Affiliation(s)
- E Asselin
- Département d'Ontogénie et Reproduction, Centre de Recherches du Centre Hospitalier de l'Université Laval, Sainte-Foy, Québec, Canada
| | | |
Collapse
|
18
|
Abstract
Trypsin, thrombin, and peptide analogues of the new amino terminus of the proteolyzed thrombin receptor, SFLLRN and SFLLRNPNDKYEPF, stimulated embryonic fibroblasts cultured as 3-dimensional tissue-like aggregates to elaborate a fibronectin-rich extracellular matrix. Enzymatically inactive thrombin and the control peptide FLLRN failed to stimulate matrix production. The induction of cell proliferation correlated with production of the fibronectin matrix. The regions of active cell proliferation in the fibroblast aggregates co-localized with the matrix and peptide analogues of the RGD cell-adhesion site of fibronectin reversibly inhibited the accumulation of the fibronectin matrix and the stimulation of cell proliferation by SFLLRN. Two different preparations of the fibronectin matrix stimulated cell proliferation in aggregates cultured in growth factor-free medium. We suggest that the stimulation of matrix production is a necessary event for mitogenic signaling in mesenchymal tissue. The tight coupling between the matrigenic and mitogenic activities of growth factors was absent in monolayer cultures of chick embryonic fibroblasts since thrombin and trypsin induced proliferation of monolayer-cultured cells without inducing the production of a fibronectin matrix.
Collapse
Affiliation(s)
- M T Armstrong
- Department of Molecular and Cellular Biology, University of California, Davis 95616-8755, USA
| | | | | | | |
Collapse
|
19
|
Affiliation(s)
- G Fager
- Wallenberg Laboratory for Cardiovascular Research, Sahlgren's Hospital, Göteborg, Sweden
| |
Collapse
|
20
|
Murakami K, Ueno A, Yamanouchi K, Kondo T. Thrombin induces GRO alpha/MGSA production in human umbilical vein endothelial cells. Thromb Res 1995; 79:387-94. [PMID: 7482442 DOI: 10.1016/0049-3848(95)00127-d] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Thrombin, besides being a potent coagulation factor, exerts influence on endothelial and leukocyte functions and may thus be involved in the regulation of inflammatory reactions. The present study investigated whether thrombin stimulates the production of growth-related cytokine/melanoma growth-stimulatory activity (GRO alpha/MGSA) in endothelial cells. Human umbilical vein endothelial cells (HUVEC) stimulated with thrombin were found to product GRO alpha/MGSA in a dose- and time-dependent manner. This action of thrombin was completely suppressed by preincubation with either hirudin or antithrombin-III (AT-III)-heparin. Interestingly, the thrombin receptor-activating peptide SFLLRN mimicked the action of thrombin. In addition, staurosporine, a protein kinase C (PKC) inhibitor, attenuated the production of GRO alpha/MGSA by thrombin, SFLLRN and phorbol 12-myristate 13-acetate (PMA), but left the action of interleukin-1 beta (IL-1 beta) unchanged. These results suggest that catalytic activation of thrombin receptor by thrombin results in GRO alpha/MGSA production, at least in part, via a pathway involving PKC in HUVEC.
Collapse
Affiliation(s)
- K Murakami
- Department of Biochemistry, Green Cross Corporation, Osaka, Japan
| | | | | | | |
Collapse
|
21
|
Turnell AS, Brant DP, Brown GR, Finney M, Gallimore PH, Kirk CJ, Pagliuca TR, Campbell CJ, Michell RH, Grand RJ. Regulation of neurite outgrowth from differentiated human neuroepithelial cells: a comparison of the activities of prothrombin and thrombin. Biochem J 1995; 308 ( Pt 3):965-73. [PMID: 8948457 PMCID: PMC1136817 DOI: 10.1042/bj3080965] [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: 02/03/2023]
Abstract
The mechanism by which thrombin and prothrombin control neurite retraction was studied in Ad12E1HER10 human neuroepithelial cells. Morphological changes in differentiated cells were apparent within minutes of the addition of very low concentrations of thrombin (3 pM). Higher concentrations (2 nM) of prothrombin were required to elicit a similar response. Doses of thrombin and prothrombin sufficient to cause neurite retraction stimulated protein tyrosine kinase activity. Protein tyrosine kinase activation also correlated positively with thrombin- and prothrombin-induced phosphoinositide 3-kinase activation and InsP6 dephosphorylation. However, thrombin-stimulated Ins(1,4,5)P3 generation and intracellular Ca2+ mobilization only occurred at concentrations in excess of those needed to induce retraction. No fluctuations in Ins(1,4,5)P3 were detected after stimulation with prothrombin, and no rapid synchronized release of Ca2+ was observed, even at very high concentrations. Prothrombin did, however, cause small oscillations in the intracellular Ca2+ concentration, similar to those produced by low concentrations of thrombin, after approximately 30 min. We conclude that prothrombin- and thrombin-induced neurite retractions are not dependent on PtdIns(4,5)P2 and Ca2+ mobilization, but are more probably mediated through an effector mechanism involving protein tyrosine kinase activation. No intracellular Ca2+ mobilization, protein tyrosine kinase activity or neurite retraction was observed after treatment of cells with proteolytically inactive mutant thrombin (S205-->A). Prothrombin-mediated intracellular Ca2+ mobilization and neurite retraction were inhibited by hirudin, which was shown to interact with thrombin but not prothrombin. It is concluded that cleavage of prothrombin to thrombin is a necessary prerequisite for biological activity on differentiated Ad12E1HER10 cells and that differences in agonist concentration are capable of coupling the thrombin receptor to different pathways within the cell.
Collapse
Affiliation(s)
- A S Turnell
- Centre for Clinical Research in Immunology and Signalling, Medical School, University of Birmingham, U.K
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Herbert JM, Dupuy E, Laplace MC, Zini JM, Bar Shavit R, Tobelem G. Thrombin induces endothelial cell growth via both a proteolytic and a non-proteolytic pathway. Biochem J 1994; 303 ( Pt 1):227-31. [PMID: 7945245 PMCID: PMC1137580 DOI: 10.1042/bj3030227] [Citation(s) in RCA: 82] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Binding of 125I-thrombin to human umbilical vein endothelial cells (HUVECs) was specifically displaced by the synthetic tetradecapeptide SFLLRNPNDKYEPF, named thrombin receptor agonist peptide (TRAP), which has recently been described as a peptide mimicking the new N-terminus created by cleavage of the thrombin receptor, and F-14, a tetradecapeptide representing residues 365-378 of the human alpha-thrombin B chain. Binding of 125I-TRAP to HUVECs was time-dependent, reversible and saturable, showing high affinity (KD = 1.5 +/- 0.4 microM) and high binding capacity (Bmax. = 7.1 +/- 0.6 x 10(6) sites/cell) (n = 3). Unlabelled thrombin and TRAP competitively and selectively inhibited the specific binding of 125I-TRAP with IC50 values of 5.8 +/- 0.7 nM and 2.8 +/- 0.4 microM respectively, whereas F-14 remained ineffective at displacing 125I-TRAP from its binding sites, suggesting the presence of at least two different types of thrombin-binding sites on HUVECs. TRAP was a potent mitogen for HUVECs in culture. Both TRAP and alpha-thrombin stimulated the proliferation of HUVECs with half-maximum mitogenic responses between 1 and 10 nM. F-14 also promoted HUVEC growth. The mitogenic effects of F-14 and TRAP were additive. N alpha-(2-Naphthylsulphonylglycyl)-DL-p-amidinophenylalanylpiper idine (NAPAP) and hirudin (two specific inhibitors of the enzyme activity of thrombin) specifically inhibited thrombin-induced HUVEC growth (IC50 values 400 +/- 60 and 52 +/- 8 nM respectively) but remained without effect on the mitogenic effect of TRAP or F-14. This demonstrated that the mitogenic effect of alpha-thrombin for HUVECs was intimately linked to its esterolytic activity but also showed that thrombin can stimulate HUVEC growth via another non-enzymic pathway. This hypothesis was further reinforced by the fact that F-14-induced proliferation of HUVECs remained unaltered by two antibodies directed against TRAP or the cleavage site on the extracellular portion of the thrombin receptor, which both strongly reduced thrombin-induced proliferation of HUVECs. Thrombin-, TRAP- or F-14-induced HUVEC proliferation was strongly inhibited by a neutralizing monoclonal antibody directed against basic fibroblast growth factor (bFGF), suggesting that thrombin regulates the autocrine release of bFGF in HUVECs.
Collapse
|
23
|
Albrightson CR, Zabko-Potapovich B, Dytko G, Bryan WM, Hoyle K, Moore ML, Stadel JM. Analogues of the thrombin receptor tethered-ligand enhance mesangial cell proliferation. Cell Signal 1994; 6:743-50. [PMID: 7888301 DOI: 10.1016/0898-6568(94)00043-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Thrombin stimulates cytosolic calcium mobilization and tritiated thymidine incorporation in rat glomerular mesangial cells. This effect may be mediated by a thrombin receptor similar to the receptor found in human platelets. In order to test this possibility, a series of analogues of the thrombin receptor peptide, SFLL-RNPNDKYEPF, was evaluated for their effects on mesangial cells. Analogues of the thrombin receptor peptide containing five, six, seven and 14 amino acids were as efficacious as thrombin with respect to calcium mobilization and thymidine incorporation, although they were significantly less potent. The dissimilarity in potency between thrombin and the thrombin receptor peptides is consistent with the kinetics of the proposed mechanism of action of the enzyme, since the cleavage by thrombin of its receptor results in a tethered ligand which is at a relatively high concentration compared to the free peptides in solution. Those thrombin receptor peptide analogues which showed decreased activity in platelets were tested in mesangial cells. Removal of serine at position one, N-acetylation, or replacement of the phenylalanine at position two with alanine resulted in analogues which were inactive in stimulating mesangial cell proliferation or calcium mobilization. In addition, those analogues which had no stimulatory effects in mesangial cells were not antagonists of SFLLRN-mediated calcium mobilization and thymidine incorporation in mesangial cells.
Collapse
Affiliation(s)
- C R Albrightson
- Department of Renal Pharmacology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406-0939
| | | | | | | | | | | | | |
Collapse
|
24
|
Shankar R, de la Motte C, Poptic E, DiCorleto P. Thrombin receptor-activating peptides differentially stimulate platelet-derived growth factor production, monocytic cell adhesion, and E-selectin expression in human umbilical vein endothelial cells. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)36738-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
25
|
|
26
|
Mari B, Imbert V, Belhacene N, Far D, Peyron J, Pouysségur J, Van Obberghen-Schilling E, Rossi B, Auberger P. Thrombin and thrombin receptor agonist peptide induce early events of T cell activation and synergize with TCR cross-linking for CD69 expression and interleukin 2 production. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)37225-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
27
|
|
28
|
Morris R, Winyard PG, Blake DR, Morris CJ. Thrombin in inflammation and healing: relevance to rheumatoid arthritis. Ann Rheum Dis 1994; 53:72-9. [PMID: 8311562 PMCID: PMC1005249 DOI: 10.1136/ard.53.1.72] [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: 01/29/2023]
Affiliation(s)
- R Morris
- Inflammation Research Group, London Hospital Medical College, United Kingdom
| | | | | | | |
Collapse
|
29
|
The protein encoded by a growth arrest-specific gene (gas6) is a new member of the vitamin K-dependent proteins related to protein S, a negative coregulator in the blood coagulation cascade. Mol Cell Biol 1993. [PMID: 8336730 DOI: 10.1128/mcb.13.8.4976] [Citation(s) in RCA: 392] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A set of growth arrest-specific genes (gas) whose expression is negatively regulated after serum induction has previously been described (C. Schneider, R. M. King, and L. Philipson, Cell 54:787-793, 1988). The detailed analysis of one of them, gas6, is reported here, gas6 mRNA (2.6 kb) is abundantly expressed in serum-starved (48 h in 0.5% fetal calf serum) NIH 3T3 cells but decreases dramatically after fetal calf serum or basic fibroblast growth factor stimulation. The human homolog of gas6 was also cloned and sequenced, revealing a high degree of homology and a similar pattern of expression in IMR90 human fibroblasts. Computer analysis of the protein encoded by murine and human gas6 cDNAs showed significant homology (43 and 44% amino acid identity, respectively) to human protein S, a negative coregulator in the blood coagulation pathway. By using an anti-human Gas6 monospecific affinity-purified antibody, we show that the biosynthetic level of human Gas6 fully reflects mRNA expression in IMR90 human fibroblasts. This finding thus defines a new member of vitamin K-dependent proteins that is expressed in many human and mouse tissues and may be involved in the regulation of a protease cascade relevant in growth regulation.
Collapse
|
30
|
Manfioletti G, Brancolini C, Avanzi G, Schneider C. The protein encoded by a growth arrest-specific gene (gas6) is a new member of the vitamin K-dependent proteins related to protein S, a negative coregulator in the blood coagulation cascade. Mol Cell Biol 1993; 13:4976-85. [PMID: 8336730 PMCID: PMC360142 DOI: 10.1128/mcb.13.8.4976-4985.1993] [Citation(s) in RCA: 146] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
A set of growth arrest-specific genes (gas) whose expression is negatively regulated after serum induction has previously been described (C. Schneider, R. M. King, and L. Philipson, Cell 54:787-793, 1988). The detailed analysis of one of them, gas6, is reported here, gas6 mRNA (2.6 kb) is abundantly expressed in serum-starved (48 h in 0.5% fetal calf serum) NIH 3T3 cells but decreases dramatically after fetal calf serum or basic fibroblast growth factor stimulation. The human homolog of gas6 was also cloned and sequenced, revealing a high degree of homology and a similar pattern of expression in IMR90 human fibroblasts. Computer analysis of the protein encoded by murine and human gas6 cDNAs showed significant homology (43 and 44% amino acid identity, respectively) to human protein S, a negative coregulator in the blood coagulation pathway. By using an anti-human Gas6 monospecific affinity-purified antibody, we show that the biosynthetic level of human Gas6 fully reflects mRNA expression in IMR90 human fibroblasts. This finding thus defines a new member of vitamin K-dependent proteins that is expressed in many human and mouse tissues and may be involved in the regulation of a protease cascade relevant in growth regulation.
Collapse
Affiliation(s)
- G Manfioletti
- International Center for Genetic Engineering and Biotechnology, Trieste, Italy
| | | | | | | |
Collapse
|
31
|
Howells GL, Macey M, Curtis MA, Stone SR. Peripheral blood lymphocytes express the platelet-type thrombin receptor. Br J Haematol 1993; 84:156-60. [PMID: 8393335 DOI: 10.1111/j.1365-2141.1993.tb03039.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Northern blot analysis of human mononuclear cells indicated that the platelet thrombin receptor may be expressed by lymphocytes. In order to investigate this, we prepared affinity purified rabbit antibodies against the thrombin receptor which bound platelets and blocked thrombin activation. Using flow cytometry on peripheral blood cells, we found that the vast majority of NK cells (CD16/CD56 positive) and a fraction of CD3/CD4 positive T cells expressed the thrombin receptor. B cells, neutrophils and monocytes were negative. These data suggest that potentially thrombin may play a direct role in regulating NK and T cell function.
Collapse
Affiliation(s)
- G L Howells
- Department of Oral Pathology, London Hospital Medical College
| | | | | | | |
Collapse
|
32
|
Parry MA, Stone SR, Hofsteenge J, Jackman MP. Evidence for common structural changes in thrombin induced by active-site or exosite binding. Biochem J 1993; 290 ( Pt 3):665-70. [PMID: 8457193 PMCID: PMC1132332 DOI: 10.1042/bj2900665] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The gamma-loop of thrombin is a flexible, surface-accessible loop in free thrombin that appears to be one of several sites participating in the interaction of the enzyme with macromolecular substrates and inhibitors. Using limited proteolysis and intrinsic fluorescence measurements, we have studied changes in thrombin structure induced by small, site-specific ligands. Binding of a C-terminal peptide of hirudin to the anion-binding exosite of thrombin induced a structural change in the gamma-loop, which caused a 6-fold reduction in the susceptibility of the enzyme to limited proteolysis by elastase and chymotrypsin. Binding of several active site-specific thrombin inhibitors conferred an even greater protection from proteolysis at the gamma-loop. For example, the covalent complex of thrombin with D-Phe-Pro-Arg-CH2Cl was 95-fold less susceptible to cleavage by chymotrypsin than the free enzyme. Furthermore, binding of either exosite or active-site probes induced a common intrinsic fluorescence change in thrombin (a fractional increase of 0.13). These results are surprising because crystallographic studies indicate that direct contact between the bound probes and relevant residues of the gamma-loop is very unlikely. Thus we have identified an allosteric interaction that couples the active site of thrombin to the gamma-loop. An interaction of this nature may be one way in which thrombomodulin modulates the reactivity of thrombin.
Collapse
Affiliation(s)
- M A Parry
- Friedrich Miescher-Institut, Basel, Switzerland
| | | | | | | |
Collapse
|
33
|
Mihara J, Holt SC. Purification and characterization of fibroblast-activating factor isolated from Porphyromonas gingivalis W50. Infect Immun 1993; 61:588-95. [PMID: 8380795 PMCID: PMC302768 DOI: 10.1128/iai.61.2.588-595.1993] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
A 24-kDa polypeptide which activated the incorporation of [3H]thymidine into human fibroblasts was isolated from the outer membrane vesicles of Porphyromonas gingivalis W50. This polypeptide, named fibroblast activating factor (FAF), was isolated by 3-[(3-cholamidopropyl)-dimethyl-ammonio]-1-propane-sulfonate (CHAPS) detergent extraction and purified by DEAE ion-exchange chromatography and preparative isoelectric focusing. Purified FAF (100 ng of protein per ml) caused a 400% increase in [3H]thymidine incorporation into human gingival fibroblasts (HGFs) compared with results for controls. FAF was characterized as (i) a polypeptide with molecular masses of 24 kDa when heated at 100 degrees C for 5 min and 44 kDa when unheated, (ii) heat sensitive but not affected by selected reducing reagents, and (iii) possessing slight phosphatase activity. N'-terminal sequence analysis revealed no homology with P. gingivalis peptides or with any host-derived growth factors. These data suggest that FAF functions as a significant virulence factor which in vivo is capable of modulating homeostasis in local connective tissues.
Collapse
Affiliation(s)
- J Mihara
- Department of Periodontics, University of Texas Health Science Center, San Antonio 78284-7894
| | | |
Collapse
|
34
|
Nelken NA, Soifer SJ, O'Keefe J, Vu TK, Charo IF, Coughlin SR. Thrombin receptor expression in normal and atherosclerotic human arteries. J Clin Invest 1992; 90:1614-21. [PMID: 1328304 PMCID: PMC443210 DOI: 10.1172/jci116031] [Citation(s) in RCA: 262] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Thrombin is a multifunctional serine protease generated at sites of vascular injury. A host of thrombin actions on vascular endothelial cells, smooth muscle cells, and macrophages has been defined in cell culture systems, but the in vivo significance of these activities is unknown. We have defined the expression of the recently identified receptor for thrombin in human arteries by both in situ hybridization and immunohistochemistry. In normal-appearing arteries, thrombin receptor was expressed almost exclusively in the endothelial layer. By contrast, in human atheroma, the receptor was widely expressed, both in regions rich in macrophages and in regions rich in vascular smooth muscle cells and mesenchymal-appearing intimal cells of unknown origin. Thrombin receptor was expressed by human vascular endothelial cells and smooth muscle cells in culture and by macrophages obtained by bronchioalveolar lavage, thus demonstrating that all three cell types are indeed capable of expressing the thrombin receptor. These results establish thrombin receptor activation as a candidate for contributing to sclerotic and inflammatory processes in the human vasculature, such as those that occur in atherosclerosis and restenosis.
Collapse
Affiliation(s)
- N A Nelken
- Cardiovascular Research Institute, University of California, San Francisco 94143
| | | | | | | | | | | |
Collapse
|
35
|
Scarborough R, Naughton M, Teng W, Hung D, Rose J, Vu T, Wheaton V, Turck C, Coughlin S. Tethered ligand agonist peptides. Structural requirements for thrombin receptor activation reveal mechanism of proteolytic unmasking of agonist function. J Biol Chem 1992. [DOI: 10.1016/s0021-9258(18)42184-9] [Citation(s) in RCA: 140] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
36
|
Hung DT, Vu TK, Wheaton VI, Ishii K, Coughlin SR. Cloned platelet thrombin receptor is necessary for thrombin-induced platelet activation. J Clin Invest 1992; 89:1350-3. [PMID: 1313452 PMCID: PMC442997 DOI: 10.1172/jci115721] [Citation(s) in RCA: 146] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Platelet activation by thrombin is critical for hemostasis and thrombosis. Structure-function studies with a recently cloned platelet thrombin receptor suggest that a hirudin-like domain in the receptor's extracellular amino terminal extension is a thrombin-binding determinant important for receptor activation. We now report that a peptide antiserum to this domain is a potent and specific antagonist of thrombin-induced platelet activation. This study demonstrates that the cloned platelet thrombin receptor is necessary for platelet activation by thrombin, and provides a strategy for developing blocking monoclonal antibodies of potential therapeutic value.
Collapse
Affiliation(s)
- D T Hung
- Department of Laboratory Medicine, University of California, San Francisco 94143
| | | | | | | | | |
Collapse
|
37
|
Hott JW, Sparks JA, Godbey SW, Antony VB. Mesothelial cell response to pleural injury: thrombin-induced proliferation and chemotaxis of rat pleural mesothelial cells. Am J Respir Cell Mol Biol 1992; 6:421-5. [PMID: 1550687 DOI: 10.1165/ajrcmb/6.4.421] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Injury to the pleura ultimately results in either repair with fibrosis or repair without fibrosis and a reestablishment of the normal mesothelial monolayer. The role of the mesothelial cell, and of local mediators, in these repair processes remains essentially undefined. In order for repair without fibrosis to occur, mesothelial cells, in response to local mediators, must be capable of migration and/or proliferation to cover the injured and denuded mesothelium. We hypothesized that rat pleural mesothelial cells were capable of both chemotaxis and proliferation in response to thrombin. In an in vitro assay, mesothelial cells demonstrated directed migration in response to a known chemoattractant, formylmethionylleucylphenylalanine. In addition, mesothelial cells demonstrated chemotaxis in a dose-dependent manner in response to thrombin, with a maximal response at a concentration of 10(-8) M. Finally, this chemotaxis was blocked by a specific blocker of thrombin, antithrombin 3. Thrombin also stimulated mesothelial cell proliferation, which was measured both in a [3H]thymidine incorporation assay and by direct cell counts. Again, the response was dose dependent, with the maximal response at 10(-8) M causing the same amount of [3H]thymidine incorporation as 10% fetal bovine serum. As before, this response was completely blocked by antithrombin 3. These results demonstrate that mesothelial cells are capable of both chemotaxis and proliferation in response to thrombin. Thrombin may play an important role in the regulation of pleural repair without fibrosis and the re-establishment of the mesothelial monolayer.
Collapse
Affiliation(s)
- J W Hott
- Department of Medicine, Veterans Administration Medical Center, Indianapolis, IN 46202
| | | | | | | |
Collapse
|
38
|
Hung DT, Vu TH, Nelken NA, Coughlin SR. Thrombin-induced events in non-platelet cells are mediated by the unique proteolytic mechanism established for the cloned platelet thrombin receptor. J Cell Biol 1992; 116:827-32. [PMID: 1309820 PMCID: PMC2289317 DOI: 10.1083/jcb.116.3.827] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
We recently isolated a cDNA clone encoding a functional platelet thrombin receptor that defined a unique mechanism of receptor activation. Thrombin cleaves its receptor's extracellular amino terminal extension, unmasking a new amino terminus that functions as a tethered peptide ligand and activates the receptor. A novel peptide mimicking this new amino terminus was a full agonist for platelet secretion and aggregation, suggesting that this unusual mechanism accounts for platelet activation by thrombin. Does this mechanism also mediate thrombin's assorted actions on non-platelet cells? We now report that the novel thrombin receptor agonist peptide reproduces thrombin-induced events (specifically, phosphoinositide hydrolysis and mitogenesis) in CCL-39 hamster lung fibroblasts, a naturally thrombin-responsive cell line. Moreover, these thrombin-induced events could be recapitulated in CV-1 cells, normally poorly responsive to thrombin, after transfection with human platelet thrombin receptor cDNA. Our data show that important thrombin-induced cellular events are mediated by the same unusual mechanism of receptor activation in both platelets and fibroblasts, very likely via the same or very similar receptors.
Collapse
Affiliation(s)
- D T Hung
- Department of Laboratory Medicine, University of California, San Francisco 94143-0524
| | | | | | | |
Collapse
|
39
|
Coughlin SR, Vu TK, Hung DT, Wheaton VI. Characterization of a functional thrombin receptor. Issues and opportunities. J Clin Invest 1992; 89:351-5. [PMID: 1310691 PMCID: PMC442859 DOI: 10.1172/jci115592] [Citation(s) in RCA: 224] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- S R Coughlin
- Cardiovascular Research Institute, University of California, San Francisco 94143
| | | | | | | |
Collapse
|
40
|
Vu TK, Hung DT, Wheaton VI, Coughlin SR. Molecular cloning of a functional thrombin receptor reveals a novel proteolytic mechanism of receptor activation. Cell 1991; 64:1057-68. [PMID: 1672265 DOI: 10.1016/0092-8674(91)90261-v] [Citation(s) in RCA: 2253] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We isolated a cDNA encoding a functional human thrombin receptor by direct expression cloning in Xenopus oocytes. mRNA encoding this receptor was detected in human platelets and vascular endothelial cells. The deduced amino acid sequence revealed a new member of the seven transmembrane domain receptor family with a large amino-terminal extracellular extension containing a remarkable feature. A putative thrombin cleavage site (LDPR/S) resembling the activation cleavage site in the zymogen protein C (LDPR/I) was noted 41 amino acids carboxyl to the receptor's start methionine. A peptide mimicking the new amino terminus created by cleavage at R41 was a potent agonist for both thrombin receptor activation and platelet activation. "Uncleavable" mutant thrombin receptors failed to respond to thrombin but were responsive to the new amino-terminal peptide. These data reveal a novel signaling mechanism in which thrombin cleaves its receptor's amino-terminal extension to create a new receptor amino terminus that functions as a tethered ligand and activates the receptor.
Collapse
Affiliation(s)
- T K Vu
- Department of Medicine, University of California, San Francisco 94143-0524
| | | | | | | |
Collapse
|
41
|
Prokesová L, Porwit-Bóbr Z, Baran K, Potempa J, Pospísil M, John C. Effect of metalloproteinase from Staphylococcus aureus on in vitro stimulation of human lymphocytes. Immunol Lett 1991; 27:225-30. [PMID: 2060975 DOI: 10.1016/0165-2478(91)90156-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Metalloproteinase (MP) produced by the majority of Staphylococcus aureus strains exerts, in a wide concentration range (0.1-100 micrograms/ml), no cytotoxic action on mononuclear leukocytes of human peripheral blood. The enzyme itself does not appreciably stimulate proliferation and differentiation of lymphocytes in culture, but affects the stimulation of both T and B lymphocytes by polyclonal activators. The action is dose-dependent. High doses of MP (100 micrograms/ml) lower the blastic transformation after stimulation with Con A, SpA, NDCM, S. aureus strain Wood 46 and with suboptimal doses of PWM. Optimal concentrations of the enzyme potentiate the stimulation of lymphocytes by PWM, PHA, S. aureus strains Cowan 1 and Wood 46, and by NDCM. The same potentiation effect was achieved whether the enzyme was added concurrently with the mitogen or 18 h later. This implies that the beginning of cell activation is not affected. A high MP concentration decreases the production of Ig in culture after stimulation with PWM whereas lower concentrations of MP enhance this production. Production of Ig after stimulation with NDCM and Wood 46 is decreased by MP. The possible action of exoproteinase from S. Aureus on the immune response during infection is discussed.
Collapse
Affiliation(s)
- L Prokesová
- Laboratory of Special Medical Microbiology and Immunology, 1st Medical Faculty, Charles University, Prague, Czechoslovakia
| | | | | | | | | | | |
Collapse
|
42
|
Niinobu T, Ogawa M, Murata A, Nishijima J, Mori T. Identification and characterization of receptors specific for human pancreatic secretory trypsin inhibitor. J Exp Med 1990; 172:1133-42. [PMID: 2170560 PMCID: PMC2188617 DOI: 10.1084/jem.172.4.1133] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Specific binding sites for human pancreatic secretory trypsin inhibitor (PSTI) on 3T3 Swiss albino cells were studied using radioiodinated recombinant PSTI. Some ion species, pH, and temperature significantly influenced the binding of 125I-PSTI. Kinetic studies showed that the binding of 125I-PSTI to 3T3 Swiss albino cells reached the maximum level within 120 min at 4 degrees C, with a slow dissociation rate. The half-maximal inhibition (ID50) of 125I-PSTI binding by unlabeled PSTI occurred at 1.0 x 10(-10) M. On Scatchard analysis of the competitive binding data, linear plots indicated a single class of receptors with high affinity (Kd = 5.3 x 10(-10) M) on 3T3 Swiss albino cells, the number of receptors being 5,400 per cell. Treatment of surface-bound radiolabeled PSTI with a chemical crosslinker (disuccinimidyl suberate) led to the identification of a membrane polypeptide of Mr 140,000 to which PSTI was crosslinked. The formation was inhibited by an excess amount of unlabeled PSTI in a dose-dependent manner. The binding of 125I-PSTI to 3T3 Swiss albino cells was competitively inhibited by unlabeled PSTI but not by other peptide hormones, such as epidermal growth factor (EGF), bovine fibroblast growth factor, insulin-like growth factor, transforming growth factor alpha, platelet-derived growth factor, and tumor necrosis factor, indicating the presence of receptors specific for PSTI. Various protease inhibitors had no or only a little effect, and mercaptoethanol and dithiothreitol strongly decreased the binding of 125I-PSTI. Incubation at 37 degrees C resulted in rapid internalization of cell-bound 125I-PSTI, followed by the appearance of trichloroacetic acid-soluble 125I-radioactivity in the culture medium, due to degradation of internalized PSTI. In addition, PSTI stimulated [3H]thymidine incorporation into DNA on 3T3 Swiss albino cells in a dose-dependent manner. The combined addition of PSTI and EGF stimulated [3H]thymidine incorporation to an extent greater than that seen with either agent alone. These results indicated that the biological effect of PSTI was mediated by high affinity plasma membrane receptors, which were not a cell-surface proteinase(s). Specific binding of 125I-PSTI was noted with the following cells: WI-38, 3T3 Swiss albino, HUVE, BDC-1, and H4-II-E-C3.
Collapse
Affiliation(s)
- T Niinobu
- Second Department of Surgery, Osaka University Medical School, Japan
| | | | | | | | | |
Collapse
|
43
|
Oliveira CM, Banerjee DK. Role of extracellular signaling on endothelial cell proliferation and protein N-glycosylation. J Cell Physiol 1990; 144:467-72. [PMID: 2202743 DOI: 10.1002/jcp.1041440314] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In vitro studies of angiogenic phenomenon have been limited due to nonavailability of a simple and biologically relevant model of the capillary wall. Recent development of a capillary endothelial cell line from the vascular bed of bovine adrenal medulla made us to study the effect of heparin, thrombin, thyroxine, glucagon, insulin, and phorbol myristate acetate (PMA) on the proliferative and metabolic activities such as glycosylation of asparagine-linked glycoproteins of these cells in culture. Out of six different agents studied here, only heparin, thrombin, and thyroxine reduced the doubling time of these cells by 24 hr with no observed morphological abnormality. Glucagon, showed marginal reduction in the cell doubling time. By contrast, insulin and PMA enhanced the doubling time. Insulin treatment though induced the S phase of cell cycle but it blocked the cells entry into the G2 + M phase. PMA arrested the cells in G0/G1 phase. The cellular response to protein N-glycosylation is increased in the presence of thyroxine, insulin, and thrombin and the effect is dose dependent. Further analysis on SDS-PAGE indicated that glycosylation of 80-120 kDa and 43 kDa glycoprotein species are enhanced when these cells are treated with insulin and thrombin. Glycopeptide generated from these glycoproteins suggested that they all carry "high mannose" and "complex" type oligosaccharide chains attached to their protein core.
Collapse
Affiliation(s)
- C M Oliveira
- Department of Biochemistry and Nutrition, School of Medicine, University of Puerto Rico, San Juan
| | | |
Collapse
|
44
|
Shen XY, Hamilton TA, DiCorleto PE. Thrombin-induced expression of the KC gene in cultured aortic endothelial cells. Involvement of proteolytic activity and protein kinase C. BIOCHIMICA ET BIOPHYSICA ACTA 1990; 1049:145-50. [PMID: 2194575 DOI: 10.1016/0167-4781(90)90034-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The KC gene, first identified in platelet-derived growth factor-stimulated BALB/c 3T3 cells, shares structural similarities with a new family of genes that code for secreted cytokines which appear to be involved in wound healing and inflammation. Thrombin is a coagulation system proteinase likely to be present in vivo at sites of tissue injury. This enzyme is known to stimulate multiple responses in cultured endothelial cells (EC), including the production of eicosanoids, the expression of growth factor genes and the adhesion of leukocytes. The present experiments were designed to examine the effect of thrombin on KC mRNA expression in EC and to explore the molecular mechanisms involved. Thrombin caused a marked concentration-dependent increase in the steady state level of KC mRNA in confluent porcine aortic EC. The level of KC mRNA reached a peak 2 h after thrombin treatment and returned to near control levels by 8 h. Thrombin that was pretreated with phenylmethylsulfonyl fluoride (PMSF) to block proteolytic activity did not stimulate KC gene expression. Trypsin (2 micrograms/ml) but not PSMF-trypsin also caused a substantial increase in the level of KC mRNA. We postulated a role for protein kinase C in thrombin-induced KC gene expression since previous work had demonstrated a similar EC response to phorbol esters. This hypothesis was further supported by the finding that thrombin-induced KC expression was suppressed by the C kinase inhibitor 1-(5-isoquinolinesulfonyl)-2-methylpiperazine, but not by its structural analogue. The results of the present study demonstrate that thrombin augments KC mRNA expression by vascular EC in a process that requires intact proteinase activity. The activation of protein kinase C may be a necessary component of the intracellular signalling pathway involved in this response.
Collapse
Affiliation(s)
- X Y Shen
- Department of Vascular Cell Biology and Atherosclerosis Research, Cleveland Clinic Foundation, OH 44195
| | | | | |
Collapse
|
45
|
|
46
|
Schöne A, Zimmermann E, Knop J. Induction of procoagulant activity in human epidermal cells. Arch Dermatol Res 1989; 281:316-20. [PMID: 2802662 DOI: 10.1007/bf00412974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We have studied the induction of procoagulant activity (PCA) by lipopolysaccharide (LPS) in cultured human epidermal cells. Single cell suspensions of epidermal cells were prepared from surgical specimens and stimulated for 24 h with LPS (100 micrograms/ml). PCA was determined by one-stage clotting assay. Stimulation of the epidermal cells with LPS resulted in a significant reduction of the clotting time (approx. 30%) as compared with the nonstimulated controls. Further analysis of the induced PCA showed that it did not require factors of the intrinsic pathway of the clotting cascade (factors XI and XII). Similarly, PCA was not affected by factor IX-deficient plasma but required factors II, VII, and X for its full expression. PCA was inactivated by treatment with phospholipase C but not by heating to 56 degrees C. These data indicate that the epidermal cell PCA resembles tissue factor-like activity, activating the extrinsic clotting pathway. Elimination of Langerhans' cells from the epidermal cell suspension by antibody and complement-mediated lysis did not result in a reduction of PCA in the remaining epidermal cells, indicating that keratinocytes were most likely the producer cells. Induction of PCA on the cell membrane surface of epidermal cells may be an early event resulting in the initiation of a local inflammatory reaction.
Collapse
Affiliation(s)
- A Schöne
- Department of Dermatology, University of Münster, FRG
| | | | | |
Collapse
|
47
|
Takahashi S, Ohishi Y, Kato H, Noguchi T, Naito H, Aoyagi T. The effects of bestatin, a microbial aminopeptidase inhibitor, on epidermal growth factor-induced DNA synthesis and cell division in primary cultured hepatocytes of rats. Exp Cell Res 1989; 183:399-412. [PMID: 2548886 DOI: 10.1016/0014-4827(89)90400-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We investigated the effects of microbial protease inhibitors, in particular the aminopeptidase inhibitor bestatin, on DNA synthesis and cell division induced by epidermal growth factor (EGF) in hepatocytes. Although bestatin did not significantly affect binding of EGF to hepatocytes, it inhibited EGF-induced DNA synthesis and cell division. DNA synthesis in rat hepatocytes was maximal 24-26 h after EGF addition to the medium. The time required for maximal DNA synthesis was not affected if bestatin was removed less than 12 h after addition, but synthesis was partially inhibited if bestatin was added to the medium several hours after EGF addition, depending on the time of bestatin addition. Our results suggest that bestatin arrests the new cell cycle induced by EGF at about 12 h after the initiation. Considering also our results obtained by employing other protease inhibitors, we concluded that specific proteases play important roles in hepatocyte DNA synthesis and cell division induced by EGF.
Collapse
Affiliation(s)
- S Takahashi
- Department of Agricultural Chemistry, Faculty of Agriculture, University of Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Prokesová L, Porwit-Bóbr Z, Baran K, Potempa J, John C. Effect of serine proteinase from Staphylococcus aureus on in vitro stimulation of human lymphocytes. Immunol Lett 1988; 19:127-32. [PMID: 3235112 DOI: 10.1016/0165-2478(88)90131-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In a broad concentration range (0.1-100 micrograms/ml) the serine proteinase (SP) from Staphylococcus aureus has no cytotoxic effect on human peripheral blood lymphocytes and does not stimulate them in culture. However, it affects the action of a number of polyclonal activators. In a concentration of 100 micrograms/ml SP completely eliminates blastic transformation after stimulation with B cell mitogens (NDCM, S. aureus and Escherichia coli), lowers the blastic transformation after stimulation with PWM and SPA, and does not affect the blastic transformation after stimulation with PHA. SP (100 micrograms/ml) reduces the concentration of Ig in stimulated cultures (stimulation with PWM, NDCM, S. aureus and E. coli) far below the Ig level of unstimulated controls. This effect can be ascribed to an influence on cell stimulation, not to the proteolytic cleavage of secreted Ig, although SP can partially digest Ig. The effect on lymphocyte stimulation takes place when the SP is added to the culture together with the mitogen, or 18 h after the mitogen. This implies that SP does not affect the first stage of stimulation.
Collapse
Affiliation(s)
- L Prokesová
- Department of Medical Microbiology and Immunology, Charles University, Prague, Czechoslovakia
| | | | | | | | | |
Collapse
|
49
|
Cook JR, Chen JK. Enhancement of transformed cell growth in agar by serine protease inhibitors. J Cell Physiol 1988; 136:188-93. [PMID: 2456292 DOI: 10.1002/jcp.1041360125] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We investigated the effects of three serine protease inhibitors (leupeptin, soybean trypsin inhibitor, and aprotinin) on the serum-free growth of two transformed cell lines in soft agar. Aprotinin markedly enhanced the growth of rat embryo fibroblasts that had been transformed by polyoma middle T antigen (PyMLV-REF52), while having only a slight effect on the colonial growth of SV40 transformed Balb/c 3T3 cells (SV3T3-Aga). Leupeptin and soybean trypsin inhibitor, on the other hand, significantly enhanced the growth of SV3T3-Aga cells while having little effect on PyMLV-REF52 growth. We observed no stimulatory effect of any of the protease inhibitors on serum-free monolayer growth. Under conditions of excess aprotinin, PyMLV-REF52 cells were found to be unresponsive to epidermal growth factor (EGF) at a concentration that would normally stimulate agar colony growth. However, aprotinin was not capable of supporting colony formation with transforming growth factor-beta. These results indicate that aprotinin acts primarily as a protease inhibitor in spite of its structural homology to EGF and that EGF may promote the soft agar growth of these cell lines either by inhibiting proteolysis directly or by enhancing the synthesis of a serine protease inhibitor.
Collapse
Affiliation(s)
- J R Cook
- W. Alton Jones Cell Science Center, Inc., Lake Placid, New York 12946
| | | |
Collapse
|
50
|
Simon HG, Fruth U, Geiger R, Kramer MD, Simon MM. The T cell specific serine proteinase TSP-1: biochemical characterization, genetic analysis, and functional role. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1988; 240:535-45. [PMID: 3266709 DOI: 10.1007/978-1-4613-1057-0_69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- H G Simon
- Max-Planck-Institut für Immunobiologie, Koblenz, FRG
| | | | | | | | | |
Collapse
|