Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 511
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Cell Biochem ; 124(5): 743-752, 2023 05.
Article in English | MEDLINE | ID: mdl-36947703

ABSTRACT

Glucose-regulated protein-78 (Grp78) is an endoplasmic reticulum chaperone, which is secreted by cells and associates with cell surfaces, where it functions as a receptor for activated α2 -macroglobulin (α2 M) and tissue-type plasminogen activator (tPA). In macrophages, α2 M and tPA also bind to the transmembrane receptor, LDL receptor-related protein-1 (LRP1), activating a cell-signaling receptor assembly that includes the NMDA receptor (NMDA-R) to suppress innate immunity. Herein, we demonstrate that an antibody targeting Grp78 (N88) inhibits NFκB activation and expression of proinflammatory cytokines in bone marrow-derived macrophages (BMDMs) treated with the toll-like receptor-4 (TLR4) ligand, lipopolysaccharide, or with agonists that activate TLR2, TLR7, or TLR9. Pharmacologic inhibition of the NMDA-R or deletion of the gene encoding LRP1 (Lrp1) in BMDMs neutralizes the activity of N88. The fibrinolysis protease inhibitor, plasminogen activator inhibitor-1 (PAI1), has been implicated in diverse diseases including metabolic syndrome, cardiovascular disease, and type 2 diabetes. Deletion of Lrp1 independently increased expression of PAI1 and PAI2 in BMDMs, as did treatment of wild-type BMDMs with TLR agonists. tPA, α2 M, and N88 inhibited expression of PAI1 and PAI2 in BMDMs treated with TLR-activating agents. Inhibiting Src family kinases blocked the ability of both N88 and tPA to function as anti-inflammatory agents, suggesting that the cell-signaling pathway activated by tPA and N88, downstream of LRP1 and the NMDA-R, may be equivalent. We conclude that targeting cell-surface Grp78 may be effective in suppressing innate immunity by a mechanism that requires LRP1 and the NMDA-R.


Subject(s)
Cytokines , Diabetes Mellitus, Type 2 , Humans , Cytokines/metabolism , Membrane Proteins/metabolism , Plasminogen Inactivators/metabolism , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Chaperone BiP , N-Methylaspartate/metabolism , Macrophages/metabolism , Antibodies , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism
2.
Proc Natl Acad Sci U S A ; 116(13): 6292-6297, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30862734

ABSTRACT

Inflammation in the tumor microenvironment is a strong promoter of tumor growth. Substantial epidemiologic evidence suggests that aspirin, which suppresses inflammation, reduces the risk of cancer. The mechanism by which aspirin inhibits cancer has remained unclear, and toxicity has limited its clinical use. Aspirin not only blocks the biosynthesis of prostaglandins, but also stimulates the endogenous production of anti-inflammatory and proresolving mediators termed aspirin-triggered specialized proresolving mediators (AT-SPMs), such as aspirin-triggered resolvins (AT-RvDs) and lipoxins (AT-LXs). Using genetic and pharmacologic manipulation of a proresolving receptor, we demonstrate that AT-RvDs mediate the antitumor activity of aspirin. Moreover, treatment of mice with AT-RvDs (e.g., AT-RvD1 and AT-RvD3) or AT-LXA4 inhibited primary tumor growth by enhancing macrophage phagocytosis of tumor cell debris and counter-regulating macrophage-secreted proinflammatory cytokines, including migration inhibitory factor, plasminogen activator inhibitor-1, and C-C motif chemokine ligand 2/monocyte chemoattractant protein 1. Thus, the pro-resolution activity of AT-resolvins and AT-lipoxins may explain some of aspirin's broad anticancer activity. These AT-SPMs are active at considerably lower concentrations than aspirin, and thus may provide a nontoxic approach to harnessing aspirin's anticancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , Aspirin/pharmacology , Neoplasms/drug therapy , Neoplasms/prevention & control , Animals , Aspirin/administration & dosage , Chemokine CCL2/metabolism , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Eicosanoids/metabolism , Fatty Acids, Unsaturated/metabolism , Female , Inflammation/drug therapy , Lipoxins/metabolism , Macrophages/drug effects , Macrophages/metabolism , Metabolomics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/prevention & control , Nerve Tissue Proteins/metabolism , Phagocytosis/drug effects , Plasminogen Inactivators/metabolism , Prostaglandins/metabolism
3.
Int J Mol Sci ; 22(5)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33669052

ABSTRACT

Fibrinolytic factors like plasminogen, tissue-type plasminogen activator (tPA), and urokinase plasminogen activator (uPA) dissolve clots. Though mere extracellular-matrix-degrading enzymes, fibrinolytic factors interfere with many processes during primary cancer growth and metastasis. Their many receptors give them access to cellular functions that tumor cells have widely exploited to promote tumor cell survival, growth, and metastatic abilities. They give cancer cells tools to ensure their own survival by interfering with the signaling pathways involved in senescence, anoikis, and autophagy. They can also directly promote primary tumor growth and metastasis, and endow tumor cells with mechanisms to evade myelosuppression, thus acquiring drug resistance. In this review, recent studies on the role fibrinolytic factors play in metastasis and controlling cell-death-associated processes are presented, along with studies that describe how cancer cells have exploited plasminogen receptors to escape myelosuppression.


Subject(s)
Anoikis/genetics , Autophagy , Cellular Senescence , Drug Resistance, Neoplasm , Neoplasms/metabolism , Plasminogen Inactivators/metabolism , Plasminogen/metabolism , Cell Survival , Drug Resistance, Neoplasm/genetics , Exosomes/metabolism , Extracellular Matrix/metabolism , Humans , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Plasminogen/antagonists & inhibitors , Plasminogen Inactivators/genetics , Signal Transduction/genetics
4.
J Dairy Sci ; 99(6): 4778-4794, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27016834

ABSTRACT

Lactation performance is dependent on both the genetic characteristics and the environmental conditions surrounding lactating cows. However, individual variations can still be observed within a given breed under similar environmental conditions. The role of the environment between birth and lactation could be better appreciated in cloned cows, which are presumed to be genetically identical, but differences in lactation performance between cloned and noncloned cows first need to be clearly evaluated. Conflicting results have been described in the literature, so our aim was to clarify this situation. Nine cloned Prim' Holstein cows were produced by the transfer of nuclei from a single fibroblast cell line after cell fusion with enucleated oocytes. The cloned cows and 9 noncloned counterparts were raised under similar conditions. Milk production and composition were recorded monthly from calving until 200d in milk. At 67d in milk, biopsies were sampled from the rear quarter of the udder, their mammary epithelial cell content was evaluated, and mammary cell renewal, RNA, and DNA were then analyzed in relevant samples. The results showed that milk production did not differ significantly between cloned and noncloned cows, but milk protein and fat contents were less variable in cloned cows. Furthermore, milk fat yield and contents were lower in cloned cows during early lactation. At around 67 DIM, milk fat and protein yields, as well as milk fat, protein, and lactose contents, were also lower in cloned cows. These lower yields could be linked to the higher apoptotic rate observed in cloned cows. Apoptosis is triggered by insulin-like factor growth binding protein 5 (IGFBP5) and plasminogen activator inhibitor (PAI), which both interact with CSN1S2. During our experiments, CSN1S2 transcript levels were lower in the mammary gland of cloned cows. The mammary cell apoptotic rate observed in cloned cows may have been related to the higher levels of DNA (cytosine-5-)-methyltransferase 1 (DNMT1) transcripts, coding for products that maintain the epigenetic status of cells. We conclude, therefore, that milk production in cloned cows differs slightly from that of noncloned cows. These differences may be due, in part, to a higher incidence of subclinical mastitis. They were associated with differences in cell apoptosis and linked to variations in DNMT1 mRNA. However, milk protein and fat contents were more similar among cloned cows than among noncloned cows.


Subject(s)
Cloning, Organism , Embryo Transfer/veterinary , Lactation , Mammary Glands, Animal/cytology , Animals , Apoptosis , Cattle , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Dietary Fats/analysis , Epigenesis, Genetic , Female , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Lactose/analysis , Mammary Glands, Animal/metabolism , Milk/chemistry , Milk/metabolism , Milk Proteins/analysis , Plasminogen Inactivators/genetics , Plasminogen Inactivators/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Am J Hematol ; 89(2): 151-5, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24123221

ABSTRACT

Thrombosis contributes to morbidity and mortality in neonates following cardiac surgery. Alterations in hemostatic factors following cardiac surgery have been described, but there is no data correlating these changes with risk of thrombosis in neonates. The aim of this study is to predict thrombosis in neonates undergoing cardiac surgery by assessment of a panel of hypercoagulability markers. Neonates undergoing cardiac surgery were enrolled preoperatively and prospectively followed. Preoperative hypercoagulability panel testing included thrombin generation assay (TGA), immunoassays for antithrombin III, protein C, protein S, factor VIII, thrombin-activatable fibrinolytic inhibitor (TAFI), plasminogen activator inhibitor-1 (PAI-1), and cardiolipin antibody. Postoperative thrombosis was defined by clinical events (shunt thrombosis, limb ischemia, and stroke) or imaging (intravascular or intracardiac thrombus). Risk factors for thrombosis were assessed. One hundred neonates were enrolled in the study over a two-year period. The incidence of postoperative in-hospital thrombosis was 20%. The only significant clinical risk factor associated with thrombosis was the single ventricle physiology. Hypercoagulability factors associated with increased risk of thrombosis by univariate analysis were elevated PAI-1, TAFI, and TGA, and presence of anticardiolipin antibodies. Multivariable logistic regression analysis demonstrated that elevated PAI-1 (P = 0.015), TAFI (P = 0.028), and TGA (P = 0.007) were independent predictors of thrombosis. Hypercoagulability panel testing may help identify neonates at high risk for thrombosis following cardiac surgery. Future studies are warranted to determine if high risk patients benefit from targeted anticoagulation therapies.


Subject(s)
Blood Coagulation Tests , Cardiac Surgical Procedures/adverse effects , Thrombosis/diagnosis , Thrombosis/etiology , Adolescent , Antibodies, Anticardiolipin/blood , Blood Coagulation , Blood Coagulation Factors/metabolism , Blood Coagulation Tests/methods , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Odds Ratio , Plasminogen Inactivators/metabolism , Postoperative Complications , Prognosis , ROC Curve , Risk Factors , Thrombin/biosynthesis , Thrombosis/blood
6.
Planta Med ; 80(12): 969-73, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25089738

ABSTRACT

Postoperative adhesions develop after nearly every abdominal surgery. The formation of adhesions is associated with the inflammatory response, fibrinolytic system, and extracellular matrix deposition in response to injury. Tanshinone IIA is one of the major extracts obtained from Salvia miltiorrhiza, which has anti-inflammatory effects on many diseases. Postoperative adhesions were induced by injuring the parietal peritoneum and cecum in Wistar rats, followed by the administration of various dosages of tanshinone IIA. The adhesion scores for each group were collected seven days after the initial laparotomy. The activity of the tissue-type plasminogen activator in the peritoneal lavage fluid was measured. The messenger ribonucleic acid expression levels of the tissue-type plasminogen activator, plasminogen activator inhibitor-1, and cyclooxygenase-2 in the ischaemic tissues were measured by quantitative real-time polymerase chain reaction. The intraperitoneal administration of tanshinone IIA is effective for the prevention of the formation of postoperative adhesions in rats. Tanshinone IIA increased fibrinolytic activity in the peritoneal lavage fluid and tissue-type plasminogen activator messenger ribonucleic acid expression in ischaemic peritoneal tissues but decreased the plasminogen activator inhibitor and cyclooxygenase-2 messenger ribonucleic acid expression significantly. These results revealed that tanshinone IIA was a potent postoperative adhesion preventer by enhancing fibrinolytic activity and decreasing cyclooxygenase-2 activity.


Subject(s)
Abietanes/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Fibrinolytic Agents/therapeutic use , Peritoneum/pathology , Phytotherapy , Postoperative Complications/prevention & control , Tissue Adhesions/prevention & control , Abietanes/pharmacology , Animals , Cecum/pathology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Fibrinolytic Agents/pharmacology , Injections, Intraperitoneal , Male , Peritoneum/surgery , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plasminogen Inactivators/genetics , Plasminogen Inactivators/metabolism , Postoperative Complications/metabolism , RNA, Messenger/metabolism , Rats, Wistar , Salvia miltiorrhiza/chemistry , Tissue Adhesions/metabolism , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism
7.
Vestn Ross Akad Med Nauk ; (5): 16-27, 2013.
Article in Russian | MEDLINE | ID: mdl-24000664

ABSTRACT

Review of authors' results and the most representative literature data on the role of tumor-associated proteolitic systems involved in invasion, metastasizing and angiogenic processes in diagnostics and prognosis in various oncologic diseases is presented in this paper. The main attention is paid to the key matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) as well as to the plasminogen activation system components (uPA, PAI-1) study in tumor tissues and peripheral blood. Personal results demonstrated an increase of most MMPs, uPA and PAl-I1 expression in the tumors of 70-90% patients with various neoplasms as compared to histologically unchanged adjacent tissues. MMP- 7 was shown to be a promising serologic marker of ovarian and colorectal cancer (CRC): its sensitivity at 70% specificity level comprised about 70% in both diseases. The greatest clinical interest should be paid to the implication of tumor-associated proteases as prognostic factors. Thus, results of 5-years monitoring have demonstrated that high preoperative serum MMP-7 and TIMP- levels were independent unfavorable prognosticfactors for CRC and univariate analysis revealed unfavorable prognostic role of high tumor MMP- 7 in patients with disseminated process. Tumor PAI-1 level was shown to be a valuable prognosticfactorfor stage III CRC. In the final part of the review possibilities and prospects of tumor-associated proteases usage as targets for specific molecular directed therapy are discussed.


Subject(s)
Colorectal Neoplasms/metabolism , Matrix Metalloproteinase 7/metabolism , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/metabolism , Plasminogen Activators/metabolism , Plasminogen Inactivators/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Female , Humans , Male , Molecular Targeted Therapy/methods , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Sensitivity and Specificity
8.
Br J Surg ; 98(11): 1503-16, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21877324

ABSTRACT

BACKGROUND: Current views on the pathogenesis of adhesion formation are based on the 'classical concept of adhesion formation', namely that a reduction in peritoneal fibrinolytic activity following peritoneal trauma is of key importance in adhesion development. METHODS: A non-systematic literature search (1960-2010) was performed in PubMed to identify all original articles on the pathogenesis of adhesion formation. Information was sought on the role of the fibrinolytic, coagulatory and inflammatory systems in the disease process. RESULTS: One unifying concept emerged when assessing 50 years of studies in animals and humans on the pathogenesis of adhesion formation. Peritoneal damage inflicted by surgical trauma or other insults evokes an inflammatory response, thereby promoting procoagulatory and antifibrinolytic reactions, and a subsequent significant increase in fibrin formation. Importantly, peritoneal inflammatory status seems a crucial factor in determining the duration and extent of the imbalance between fibrin formation and fibrin dissolution, and therefore in the persistence of fibrin deposits, determining whether or not adhesions develop. CONCLUSION: Suppression of inflammation, manipulation of coagulation as well as direct augmentation of fibrinolytic activity may be promising antiadhesion treatment strategies.


Subject(s)
Peritoneum/surgery , Postoperative Complications/etiology , Animals , Ascitic Fluid/chemistry , Biopsy , Blood Coagulation/physiology , Fibrinolysis/physiology , Humans , Peritoneum/metabolism , Peritonitis/blood , Peritonitis/metabolism , Peritonitis/pathology , Plasminogen Activators/metabolism , Plasminogen Inactivators/metabolism , Postoperative Complications/blood , Rats , Tissue Adhesions/blood , Tissue Adhesions/etiology
9.
Proc Natl Acad Sci U S A ; 105(25): 8754-9, 2008 Jun 24.
Article in English | MEDLINE | ID: mdl-18559859

ABSTRACT

The amyloid hypothesis states that a variety of neurotoxic beta-amyloid (Abeta) species contribute to the pathogenesis of Alzheimer's disease. Accordingly, a key determinant of disease onset and progression is the appropriate balance between Abeta production and clearance. Enzymes responsible for the degradation of Abeta are not well understood, and, thus far, it has not been possible to enhance Abeta catabolism by pharmacological manipulation. We provide evidence that Abeta catabolism is increased after inhibition of plasminogen activator inhibitor-1 (PAI-1) and may constitute a viable therapeutic approach for lowering brain Abeta levels. PAI-1 inhibits the activity of tissue plasminogen activator (tPA), an enzyme that cleaves plasminogen to generate plasmin, a protease that degrades Abeta oligomers and monomers. Because tPA, plasminogen and PAI-1 are expressed in the brain, we tested the hypothesis that inhibitors of PAI-1 will enhance the proteolytic clearance of brain Abeta. Our data demonstrate that PAI-1 inhibitors augment the activity of tPA and plasmin in hippocampus, significantly lower plasma and brain Abeta levels, restore long-term potentiation deficits in hippocampal slices from transgenic Abeta-producing mice, and reverse cognitive deficits in these mice.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Fibrinolysin/metabolism , Fibrinolytic Agents/metabolism , Animals , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Plasminogen Inactivators/metabolism , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/metabolism
10.
Reprod Biol Endocrinol ; 8: 127, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20977773

ABSTRACT

BACKGROUND: SERPINE2, also known as glia-derived nexin or protease nexin-1, belongs to the serine protease inhibitor (SERPIN) superfamily. It is one of the potent serpins that modulates the activity of the plasminogen activator (PA) and was implicated in tissue remodeling. In this study, we investigated the expression patterns of SERPINE2 in the mouse placenta and uterus during the estrous cycle, pregnancy, and lactation. METHODS: SERPINE2 was purified from mouse seminal vesicle secretion using liquid chromatography (LC) and identified by LC/tandem mass spectrometry. The antiserum against the SERPINE2 protein was raised in rabbits. To reveal the uterine and placental expression of SERPINE2, tissues at various stages were collected for real-time PCR quantification, Western blotting, and immunohistochemical staining. RESULTS: Serpine2 mRNA was the major PA inhibitor in the placenta and uterus during the estrous cycle, pregnancy, and lactation, although Serpine1 mRNA had higher expression levels than Serpine2 mRNA in the placenta. Plat seemed to be the major PA in the mouse uterus and placenta. Antiserum against the SERPINE2 protein specifically recognized two forms of SERPINE2 and an extra 75-kDa protein, which was probably a complex of SERPINE2 with a certain protease, from among thousands of protein components in the tissue extract as demonstrated by Western blotting. In the uterus, SERPINE2 was primarily localized in luminal and glandular epithelial cells but it also was detected in circular and longitudinal smooth muscle cells during the estrous cycle and lactation. It was prominently expressed in decidual stroma cells, the metrial gland, and endometrial epithelium of the pregnant uterus. In the placenta, SERPINE2 was expressed in trophoblasts of the labyrinth and spongiotrophoblasts. However, its expression was remarkably reduced in giant cells which existed in the giant cell-decidual junction zone. In contrast, prominent expression of SERPINE2 seemed to be detected on clusters of glycogen cells near the junction zone. In addition, yolk sac membranes also showed high expression of SERPINE2. CONCLUSIONS: These findings indicate that SERPINE2 is a major PA inhibitor in the placenta and uterus during the estrous cycle, pregnancy, and lactation. It may participate in the PA-modulated tissue remodeling process in the mouse placenta and uterus.


Subject(s)
Estrous Cycle/genetics , Lactation/genetics , Placenta/metabolism , Serpin E2/genetics , Uterus/metabolism , Animals , Estrous Cycle/metabolism , Female , Gene Expression Regulation , Gestational Age , Lactation/metabolism , Male , Mice , Mice, Inbred ICR , Plasminogen Inactivators/genetics , Plasminogen Inactivators/metabolism , Pregnancy , Rabbits , Serpin E2/immunology , Serpin E2/metabolism , Serpin E2/physiology , Time Factors
11.
Cells Tissues Organs ; 191(1): 36-46, 2010.
Article in English | MEDLINE | ID: mdl-19390164

ABSTRACT

Adult human mesenchymal stem cells (hMSC) are involved in wound healing and regeneration of mesodermal tissue, but the underlying homing mechanisms are not well understood. Fibrin clot formation is associated with most wound healing processes and potentially guides the recruitment of hMSC. The objective of this study is the investigation of a fibrinolytic capacity, which is required for hMSC to migrate into a wounded tissue and thus to contribute to tissue regeneration. Using RT-PCR, semiquantitative real-time PCR and ELISA, we detected key components of the fibrinolytic cascade, including the urokinase plasminogen activator (uPA) and its receptor (uPAR), the tissue plasminogen activator (tPA) and the plasminogen activator inhibitor (PAI), suggesting a strong fibrinolytic activity of hMSC. To test this activity in a functional assay, we cultured fibrin-embedded hMSC in vitro for 7 days. The cells efficiently dissolved the surrounding fibrin mesh into the fibrin degradation products, the fibrinopeptides. The fibrinolytic activity of hMSC and human dermal fibroblasts, known to be critically involved in skin wound extracellular matrix remodeling, was similar. Our results suggest that a high intrinsic fibrinolytic capacity of hMSC mediates the invasion into a fibrin clot of a wounded tissue.


Subject(s)
Cell Movement , Fibrin/metabolism , Fibrinolysis/physiology , Mesenchymal Stem Cells/physiology , Adult , Cells, Cultured , Extracellular Matrix/metabolism , Fibrin Fibrinogen Degradation Products/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Mesenchymal Stem Cells/cytology , Plasminogen Inactivators/genetics , Plasminogen Inactivators/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Skin/cytology , Skin/injuries , Skin/physiopathology , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Wound Healing
12.
J Cardiovasc Pharmacol ; 55(3): 276-85, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20075745

ABSTRACT

To overcome the limitations of balloon expandible metal stent-induced neointimal smooth muscle cell proliferation, drug-coated stent devices have been developed. Drug eluting stents release high concentrations of antiproliferative agents, such as paclitaxel, to reduce neointimal hyperplasia. The proinflammatory cytokine, tumor necrosis factor-alpha (TNF-alpha), is known to cause severe endothelial dysfunction and accelerate atherosclerotic lesion progression. The interaction of TNF-alpha and paclitaxel on the release of prothrombotic molecules was examined in endothelial cells. Treatment of endothelial cells with paclitaxel had no direct effect on tissue factor (TF) expression, but TNF-alpha increased TF. Cotreatment of paclitaxel with TNF-alpha markedly augmented the release of TF. TNF-alpha induced release of plasminogen activator inhibitor but no synergism occurred with paclitaxel. Treatment of endothelial cells with paclitaxel and TNF-alpha reduced expression of thrombomodulin and protein C receptor. Tissue factor pathway inhibitor expression was reduced by prolonged treatment with either paclitaxel or TNF-alpha. The adhesion molecule, CD62 E, was induced by TNF-alpha; however, CD31, CD62 P, and CD106 were not affected by paclitaxel and TNF-alpha. Apoptosis was not observed with cotreatment of endothelial cells with paclitaxel and TNF-alpha. CD59-positive microparticles were released in response to TNF-alpha, but the release was not augmented by paclitaxel. Paclitaxel and TNF-alpha increased the nitrotyrosination of proteins. These findings indicate that paclitaxel enhances TNF-alpha-induced release of TF, and downregulated thrombomodulin, increased protein nitration, which may subsequently favor prothrombotic intimal surface.


Subject(s)
Endothelium, Vascular/drug effects , Paclitaxel/toxicity , Tubulin Modulators/toxicity , Tumor Necrosis Factor-alpha/toxicity , Cells, Cultured , Cytokines/drug effects , Cytokines/metabolism , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/physiopathology , Nitrates/metabolism , Paclitaxel/pharmacology , Plasminogen Inactivators/metabolism , Proteins/metabolism , Thrombomodulin/drug effects , Thrombomodulin/genetics , Thromboplastin/drug effects , Thromboplastin/genetics , Tubulin Modulators/pharmacology , Tumor Necrosis Factor-alpha/metabolism
13.
Bioorg Med Chem Lett ; 20(3): 966-70, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20056540

ABSTRACT

Inactivators of plasminogen activator inhibitor-1 (PAI-1) have been identified as possible treatments for a range of conditions, including atherosclerosis, venous thrombosis, and obesity. We describe the synthesis and inhibitory activity of a novel series of compounds based on bis-arylsulfonamide and aryl sulfonimide motifs that show potent and specific activity towards PAI-1. Inhibitors containing short linking units between the sulfonyl moieties and a 3,4-dihydroxy aryl substitution pattern showed the most potent inhibitory activity, and retained high specificity for PAI-1 over the structurally-related serpin anti-thrombin III (ATIII).


Subject(s)
Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Inactivators/chemistry , Plasminogen Inactivators/metabolism , Sulfonamides/chemistry , Sulfonamides/metabolism , Humans
14.
J Obstet Gynaecol ; 30(4): 404-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20455727

ABSTRACT

Increasing evidence indicates that statins increase peritoneal fibrinolysis through RhoA-linked pathway and may play a role in the prevention of postoperative adhesion. This study investigated whether lovastatin perturbs cytoskeletons and cell morphology by RhoA suppression in peritoneal Met5A cells. Subcellular distributions of RhoA protein and actin were assessed by immunocytochemical staining. Exposure to lovastatin caused actin filament reorganisation, decrease in the active (membrane-bound) form of RhoA and subsequently cellular shrinkage in Met5A cells. These lovastatin-induced changes were significantly overcome by the addition of geranylgeranyl pyrophosphate (downstream intermediate of HMG-CoA pathway). A RhoA protein inhibitor C3 transferase mimicked the effects of lovastatin on the Met5A cells. The effects of lovastatin on cytoskeleton alterations were correlated well with inhibition of the membrane localisation of RhoA. These results suggest that lovastatin induced reversible cytoskeleton reorganisation and cellular retraction through the reduction of RhoA geranylgeranylation, which in turn suppressed RhoA signalling-associated cellular events.


Subject(s)
Actin Cytoskeleton/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Peritoneal Diseases/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Line , Humans , Plasminogen Inactivators/metabolism , Signal Transduction/drug effects , Tissue Adhesions/metabolism
15.
Plast Reconstr Surg ; 145(2): 392-401, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31985629

ABSTRACT

BACKGROUND: Sequential compression devices are often considered a mainstay of prophylaxis against deep venous thromboses in surgical patients. The devices are believed to produce a milking action on the deep veins to prevent venous stasis. A systemic fibrinolytic effect has also been proposed, adding a second mechanism of action. The plasma levels of tissue plasminogen activator and plasminogen activator inhibitor-1 reflect fibrinolytic activity. METHODS: A randomized trial was conducted among 50 consecutive plastic surgery outpatients undergoing cosmetic surgery performed by the author under total intravenous anesthesia and without paralysis. Patients were randomized to receive calf-length sequential compression devices or no sequential compression devices during surgery. Blood samples were obtained from the upper extremity preoperatively and at hourly intervals until the patient was discharged from the postanesthesia care unit. Tissue plasminogen activator and plasminogen activator inhibitor-1 levels were measured. Ultrasound surveillance was used in all patients. There was no outside funding for the study. RESULTS: All patients agreed to participate (inclusion rate, 100 percent). No patient developed clinical signs or ultrasound evidence of a deep venous thrombosis. There were no significant changes in tissue plasminogen activator levels or plasminogen activator inhibitor-1 levels from the preoperative measurements at any hourly interval and no differences in levels comparing patients treated with or without sequential compression devices. CONCLUSIONS: No significant change in systemic fibrinolytic activity occurs during outpatient plastic surgery under total intravenous anesthesia. Sequential compression devices do not affect tissue plasminogen activator or plasminogen activator inhibitor-1 levels, suggesting no fibrinolytic benefit. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, I.


Subject(s)
Fibrinolysis/physiology , Intermittent Pneumatic Compression Devices , Adult , Aged , Cosmetic Techniques , Female , Humans , Male , Middle Aged , Operative Time , Plasminogen Inactivators/metabolism , Prospective Studies , Plastic Surgery Procedures/methods , Tissue Plasminogen Activator/metabolism , Venous Thrombosis/blood , Venous Thrombosis/prevention & control , Young Adult
16.
J Cell Biol ; 111(5 Pt 1): 2183-95, 1990 Nov.
Article in English | MEDLINE | ID: mdl-1699954

ABSTRACT

Polyclonal antibodies against plasminogen activator inhibitor type-I (PAI-1) caused rapid retraction and rounding of substrate-attached HT-1080 cells. The kinetics and extent of antibody-mediated cell rounding were not dependent on either urokinase or plasmin activity. Cells adherent to vitronectin-coated substrates detached within 2 h of antibody addition. Cells adherent to fibronectin were unaffected by the antibodies. Immunoblotting of substrate-attached material indicated that HT-1080 cells deposited PAI-1 into vitronectin, but not fibronectin, dependent contacts. These data suggest that the antibody-mediated cell rounding resulted from a steric disruption of vitronectin-dependent adhesions, indicating that the binding site on vitronectin for PAI-1 is near, but does not overlap, the binding site for vitronectin receptor. The accumulation of PAI-1 into vitronectin-dependent adhesion sites correlated temporally with the preferential degradation of fibronectin from the substrate. HT-1080 cells adherent to either fibronectin or vitronectin were able to activate exogenous plasminogen to plasmin. Plasmin levels were increased 200% on cells adherent to fibronectin and 100% on cells adherent to vitronectin. In the presence of a neutralizing antibody against PAI-1, vitronectin adherent cells activated plasminogen to the same extent as fibronectin adherent cells. Plasmin levels of 200% above baseline were associated with retraction of cells from the substrate. The ability of vitronectin adherent cells to activate exogenous plasmin was completely blocked in the presence of neutralizing antibodies against urokinase. These data represent the first demonstration that vitronectin-associated PAI-1 regulates urokinase in focal contact areas.


Subject(s)
Fibrosarcoma/metabolism , Glycoproteins/physiology , Plasminogen Inactivators/metabolism , Antibodies , Cell Adhesion/physiology , Fibrinolysin/metabolism , Fibronectins/metabolism , Humans , Plasminogen Inactivators/isolation & purification , Tumor Cells, Cultured , Vitronectin
17.
J Cell Biol ; 113(6): 1413-23, 1991 Jun.
Article in English | MEDLINE | ID: mdl-2045419

ABSTRACT

Immunogold EM was employed to compare the distribution of type 1 plasminogen activator inhibitor (PAI-1) on the surface of agonist-activated human umbilical vein endothelial cells (HUVECs) with that of control, unactivated cells. As previously observed, (Schleef, R.R., T.J. Podor, E. Dunne, J. Mimuro, and D.J. Loskutoff. J. Cell Biol. 110:155-163), analysis of cross-sections of nonpermeabilized control HUVEC monolayers stained first with affinity-purified rabbit antibodies to PAI-1 and then with gold-conjugated goat anti-rabbit IgG, revealed the presence of relatively few gold particles (less than 1-2% of the total) on the apical cell surface. The majority of gold particles were detected primarily in the extracellular matrix between the culture substratum and the cell membrane. In contrast, treatment of HUVECs with tumor necrosis factor alpha (TNF alpha; 200 U/ml, 24 h) or with lipopolysaccharide (LPS; 10 micrograms/ml, 24 h) resulted in an increased staining of PAI-1 not only in the extracellular matrix, but also on the apical cell surface (10-fold increase). Immunoabsorption of the rabbit anti-PAI-1 with purified PAI-1, or treatment of HUVECs with tissue-type plasminogen activator (2.5 micrograms/ml, 2 h, 4 degrees C) reduced the amount of staining both on the apical surface and in the extracellular matrix of agonist-activated HUVECs by 80-95%. The topographical location of PAI-1 on the cell surface was examined further by coupling immunogold staining with high resolution surface replication. Transmission EM of surface replicas from TNF alpha- or LPS-activated HUVECs revealed a general increase in PAI-1 staining both on planar regions and within indentations of the apical cell surface. Nonactivated HUVECs revealed PAI-1-specific immunogold particles only in areas of exposed extracellular matrix between the cells and occasionally at regions of cell-cell contacts. Analysis of activated bovine aortic endothelial cells by immuno-electron microscopy, immunologic assays, and flow cytometry revealed similar increases in surface PAI-1. These increases in surface PAI-1 could be detected by 3 h and continued over a 24-h period. The expression of PAI-1 on the luminal surface of endothelial cells during immune or inflammatory reactions could reduce endothelial fibrinolytic activity, thus, promoting the localized, pathologic formation of intravascular thrombi.


Subject(s)
Endothelium, Vascular/metabolism , Plasminogen Inactivators/metabolism , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/ultrastructure , Flow Cytometry , Humans , Lipopolysaccharides/pharmacology , Microscopy, Immunoelectron , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins
18.
J Cell Biol ; 115(1): 191-9, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1918136

ABSTRACT

Five out of six human melanoma cell lines tested were able to degrade in vitro a smooth muscle cell extracellular matrix in a plasmin-dependent way. In three of these five cell lines, this process was mediated by tissue-type plasminogen activator (t-PA) and in the other two cell lines by urokinase-type plasminogen activator (u-PA). All melanoma cell lines produced t-PA mRNA and protein, whereas only the two cell lines showing u-PA-mediated matrix degradation produced u-PA mRNA and protein. These latter cell lines also produced plasminogen activator inhibitor type-1 (PAI-1) and type-2 (PAI-2) mRNA and protein. u-PA receptor (u-PA-R) mRNA and binding of radiolabeled u-PA was found in all melanoma cell lines. The metastatic capacity of these cell lines was studied in nude mice. All cell lines were able to develop primary tumors at the subcutaneous inoculation site. The production of plasminogen activators, their inhibitors and urokinase receptor by subcutaneous tumors corresponded with the production by the parental cell lines in vitro. The two u-PA and PAI-1 producing cell lines showed the highest frequency to form spontaneous lung metastases after subcutaneous inoculation, whereas five of the six cell lines formed lung colonies after intravenous inoculation. In conclusion, u-PA mediated matrix degradation in vitro and production of u-PA and PAI-1 by human melanoma cell lines correlated with their ability to form spontaneous lung metastasis in nude mice. No correlation was found with the ability to form lung colonies after intravenous injection. These findings suggest a role for u-PA and PAI-1 in a relatively early stage of melanoma metastasis.


Subject(s)
Extracellular Matrix/metabolism , Melanoma/pathology , Neoplasm Metastasis , Plasminogen Inactivators/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Blotting, Northern , Gene Expression , Humans , In Vitro Techniques , Melanoma/enzymology , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Messenger/genetics , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/genetics
19.
J Cell Biol ; 115(6): 1773-81, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1721912

ABSTRACT

Vitronectin endows plasminogen activator inhibitor 1 (PAI-1), the fast-acting inhibitor of both tissue-type plasminogen activator (t-PA) and urokinase-type plasminogen activator (u-PA), with additional thrombin inhibitory properties. In view of the apparent association between PAI-1 and vitronectin in the endothelial cell matrix (ECM), we analyzed the interaction between PAI-1 and thrombin in this environment. Upon incubating 125I-labeled alpha-thrombin with endothelial cell matrix (ECM), the protease formed SDS-stable complexes exclusively with PAI-1, with subsequent release of these complexes into the supernatant. Vitronectin was required as a cofactor for the association between PAI-1 and thrombin in ECM. Metabolic labeling of endothelial cell proteins, followed by incubation of ECM with t-PA, u-PA, or thrombin, indicated that all three proteases depleted PAI-1 from ECM by complex formation and proteolytic cleavage. Proteolytically inactive thrombin as well as anticoagulant thrombin, i.e., thrombin in complex with its endothelial cell surface receptor thrombomodulin, did not neutralize PAI-1, emphasizing that the procoagulant moiety of thrombin is required for a functional interaction with PAI-1. A physiological implication of our findings may be related to the mutual neutralization of both PAI-1 and thrombin, providing a new link between plasminogen activation and the coagulation system. Evidence is provided that in ECM, procoagulant thrombin may promote plasminogen activator activity by inactivating PAI-1.


Subject(s)
Endothelium, Vascular/metabolism , Glycoproteins/metabolism , Plasminogen Inactivators/metabolism , Thrombin/physiology , Antibodies, Monoclonal/analysis , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Endopeptidases/metabolism , Endothelium, Vascular/cytology , Glycoproteins/immunology , Humans , Immune Sera , Plasminogen Activators/metabolism , Plasminogen Inactivators/immunology , Precipitin Tests , Vitronectin
20.
J Cell Biol ; 111(3): 1283-91, 1990 Sep.
Article in English | MEDLINE | ID: mdl-1697297

ABSTRACT

Bovine aortic endothelial cells (BAEs) were used as a model system to study the nature and origin of protein(s) in the extracellular matrix that bind to type 1 plasminogen activator inhibitor (PAI-1). Matrix samples were fractionated by SDS-PAGE and analyzed by PAI-1 ligand binding and by immunoblotting using antibodies to vitronectin (Vn). PAI-1 bound primarily to two Vn-related polypeptides of Mr 63,000 and 57,000, and both of these partially degraded polypeptides were present in the culture serum. Radiolabeling experiments failed to detect significant Vn biosynthesis by BAEs (less than 0.03% of total), or by human umbilical vein endothelial cells and HT 1080 cells. The binding of PAI-1 to Vn was relatively specific since direct binding studies failed to demonstrate significant interactions between PAI-1 and other matrix proteins (e.g., fibronectin, type IV collagen, laminin, or matrigel). Kinetic studies indicate that PAI-1 rapidly accumulates in the matrix when BAEs are plated on Vn, appearing in the conditioned medium only after a significant lag period (1-2 h). However, no PAI-1 was detected in the matrix when the cells were plated on fibronectin-coated dishes, and there was no lag period for PAI-1 accumulation in the medium. These results indicate that PAI-1 binds specifically to serum-derived Vn in the matrix, and suggest that the composition of both the matrix and serum itself may influence the pericellular distribution of this important inhibitor.


Subject(s)
Extracellular Matrix/physiology , Glycoproteins/physiology , Plasminogen Inactivators/metabolism , Animals , Blood Proteins/physiology , Cattle , Collagen/pharmacology , Collagen/physiology , Culture Media , Drug Combinations/pharmacology , Endothelium, Vascular/metabolism , Fibronectins/physiology , Kinetics , Laminin/pharmacology , Laminin/physiology , Protein Binding/drug effects , Proteoglycans/pharmacology , Vitronectin
SELECTION OF CITATIONS
SEARCH DETAIL