Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
Placenta ; 111: 1-9, 2021 08.
Article in English | MEDLINE | ID: mdl-34126415

ABSTRACT

INTRODUCTION: Pregnancy is a state of maternal systemic stress due to inflammation and hypoxic reactions originating from the utero-placental unit. Maternal tolerance to these stresses is a key for successful outcomes. Thrombomodulin (TM), a glycoprotein expressed on cell surface, regulates local inflammatory pathways by inhibiting proinflammatory factor, High-mobility-group box1(HMGB1). Although TM is highly expressed on placental trophoblast cells, biological activities of TM during pregnancy remains unclear. Here, we hypothesized that TM may contribute to the maternal stress coping mechanisms. METHODS: By administering recombinant-TM (rTM) to the pregnant mice, we investigated the influence of TM functions on the placenta and fetal growth. We further examined its effect on trophoblast cells, focusing on HMGB1-regulated inflammatory signalings and hypoxia-inducible factor 1α (HIF1α)-dependent regulation of placental angiogenic factors. RESULTS: Administration of rTM increased fetal weight and fetal/placental-weight ratios, which implies the improvement of placental function. These features were accompanied by maternal serum HMGB1 reduction and suppressed placental proinflammatory cytokine, IL-6 and TNF-α, expressions. In addition, rTM reduced HIF1α protein accumulation and enhanced placental growth factor (PlGF) expression in the placenta, that explains the improvement of maternal features. DISCUSSION: Our study revealed the supportive effect of TM on the placental function in mice. By inhibiting HMGB1, rTM suppresses proinflammatory cytokines, downregulates HIF1α and induces PlFG expression in the placental tissue. Our results have elucidated the novel aspects of TM; the regulation of placental inflammatory cytokines and angiogenic factors, during pregnancy. These findings may reveal potential therapeutic opportunities for the management of maternal complications.


Subject(s)
Adaptation, Physiological , Fetal Development , Placenta/metabolism , Pregnancy, Animal/physiology , Thrombomodulin/physiology , Animals , Cytokines/metabolism , Female , HMGB1 Protein/blood , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Inbred C57BL , Placenta Growth Factor/metabolism , Pregnancy , Vascular Endothelial Growth Factor Receptor-1/metabolism
2.
BMC Anesthesiol ; 21(1): 83, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33740916

ABSTRACT

BACKGROUND: The mechanisms of trauma induced coagulopathy (TIC) are considered multifactorial. Amongst others, however, shedding of the endothelial glycocalyx resulting in increased concentrations of glycocalyx fragments in plasma might also play a role. Thus, we hypothesized that shedded glycocalyx components affect coagulation and may act as humoral mediators of TIC. METHODS: To investigate effects of heparan sulfate, chondroitin sulfate, syndecan-1, versican, and thrombomodulin we added these fragments to in vitro assays of whole blood from healthy volunteers to yield concentrations observed in trauma patients. Platelet function, whole blood coagulation, and fibrinolysis were measured by standard coagulation tests, impedance aggregometry (IA), and viscoelastic tests (VET). To assess dose-response relationships, we performed IA with increasing concentrations of versican and VET with increasing concentrations of thrombomodulin. RESULTS: Intrinsically activated clotting times (i.e., activated partial thromboplastin time and intrinsically activated VET with and without heparinase) were unaffected by any glycocalyx fragment. Thrombomodulin, however, significantly and dose-dependently diminished fibrinolysis as assessed by VET with exogenously added rt-PA, and increased rt-PA-induced lysis Indices after 30 (up to 108% of control, p <  0,0001), 45 (up to 368% of control, p <  0,0001), and 60 min (up to 950% of control, p <  0,0001) in VET. Versican impaired platelet aggregation in response to arachidonic acid (up to - 37,6%, p <  0,0001), ADP (up to - 14,5%, p <  0,0001), and collagen (up to - 31,8%, p <  0,0001) in a dose-dependent manner, but did not affect TRAP-6 induced platelet aggregation. Clotting time in extrinsically activated VET was shortened by heparan sulfate (- 7,2%, p = 0,024), chondroitin sulfate (- 11,6%, p = 0,016), versican (- 13%, p = 0,012%), and when combined (- 7,2%, p = 0,007). CONCLUSIONS: Glycocalyx components exert distinct inhibitory effects on platelet function, coagulation, and fibrinolysis. These data do not support a 'heparin-like auto-anticoagulation' by shed glycosaminoglycans but suggest a possible role of versican in trauma-induced thrombocytopathy and of thrombomodulin in trauma-associated impairment of endogenous fibrinolysis.


Subject(s)
Fibrinolysis/physiology , Glycocalyx/physiology , Partial Thromboplastin Time , Platelet Aggregation/physiology , Adult , Chondroitin Sulfates/physiology , Female , Heparitin Sulfate/physiology , Humans , In Vitro Techniques , Male , Syndecan-1/physiology , Thrombomodulin/physiology , Versicans/physiology
3.
J Pharmacol Sci ; 143(1): 17-22, 2020 May.
Article in English | MEDLINE | ID: mdl-32122774

ABSTRACT

Thrombomodulin (TM) is an integral membrane protein expressed on the surface of vascular endothelial cells that suppresses blood coagulation. Recent studies have shown that TM exhibits anti-inflammatory effects by inhibiting leukocyte recruitment. However, the actual modes of action of TM in vivo remain unclear. Here, we describe the pharmacological effects of recombinant human soluble TM (TM alfa) on leukocyte dynamics in living mice using intravital imaging techniques. Under control conditions, neutrophils exhibited three distinct types of adhesion behavior in vessels: 1) "non-adhesion", in which cells flowed without vessel adhesion; 2) "rolling adhesion", in which cells transiently interacted with the endothelium; and 3) "tight binding", in which cells bound strongly to the endothelial cells. Compared to control conditions, local lipopolysaccharide stimulation resulted in an increased frequency of rolling adhesion that was not homogeneously distributed on vessel walls but occurred at specific endothelial sites. Under inflammatory conditions, TM alfa, particularly the D1 domain which is a lectin-like region of TM, significantly decreased the frequency of rolling adhesion, but did not influence the number of tight bindings. This was the first study to demonstrate that TM alfa exerts anti-inflammatory effects by inhibiting rolling adhesion of neutrophils to vascular endothelial cells in living mice.


Subject(s)
Anti-Inflammatory Agents , Cell Adhesion , Neutrophils/physiology , Thrombomodulin/physiology , Animals , Endothelium, Vascular/cytology , Male , Mice, Inbred C57BL , Molecular Imaging/methods , Recombinant Proteins/pharmacology
4.
Br J Haematol ; 187(4): 518-529, 2019 11.
Article in English | MEDLINE | ID: mdl-31335970

ABSTRACT

Simultaneous evaluation of coagulation and fibrinolysis facilitates an overall understanding of normal and pathological haemostasis. We established an assay for assessing clot formation and fibrinolysis simultaneously using clot waveform analysis by the trigger of a mixture of activated partial thromboplastin time reagent and an optimized concentration of tissue-type plasminogen activator (0·63 µg/ml) to examine the temporal reactions in a short monitoring time (<500 s). The interplay between clot formation and fibrinolysis was confirmed by analysing the effects of argatroban, tranexamic acid and thrombomodulin. Fibrinogen levels positively correlated with coagulation and fibrinolytic potential and initial fibrin clot formation was independent of plasminogen concentration. Plasminogen activator inhibitor-1-deficient (-def) and α2-antiplasmin-def plasmas demonstrated different characteristic hyper-fibrinolytic patterns. For the specificity of individual clotting factor-def plasmas, factor (F)VIII-def and FIX-def plasmas in particular demonstrated shortened fibrinolysis lag-times (FLT) and enhanced endogenous fibrinolysis potential in addition to decreased maximum coagulation velocity, possibly reflecting the fragile formation of fibrin clots. Tranexamic acid depressed fibrinolysis to a similar extent in FVIII-def and FIX-def plasmas. We concluded that the clot-fibrinolysis waveform analysis technique could sensitively monitor both sides of fibrin clot formation and fibrinolysis, and could provide an easy-to-use assay to help clarify the underlying pathogenesis of bleeding disorders in routine clinical practice.


Subject(s)
Fibrin Clot Lysis Time/methods , Fibrin/biosynthesis , Fibrinolysis , Hemorrhagic Disorders/diagnosis , Arginine/analogs & derivatives , Humans , Kinetics , Pipecolic Acids/pharmacology , Sulfonamides , Thrombomodulin/physiology , Tranexamic Acid/pharmacology
5.
J Thromb Thrombolysis ; 48(1): 81-87, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31030323

ABSTRACT

C1-inhibitor (C1INH) was shown to enhance thrombin generation (TG) in the presence of thrombomodulin (TM) by reducing production of activated protein C. Because C1INH is known to inhibit fibrinolytic system proteases, the objective of this study was to evaluate the effect of moderate (3 IU/ml) and high (16 IU/ml) C1INH concentrations on TG and plasmin generation (PG) in the presence of TM. These concentrations were evaluated based on expected maximum plasma levels following C1INH replacement therapy and recently suggested supraphysiologic dosing. TG and PG were investigated in platelet poor plasmas obtained from 21 healthy donors. An assay designed to monitor the continuous generation of the 7-amino-4-methylcoumarin fluorescence from substrates specific to thrombin or plasmin was used to evaluate the impact of C1INH activity. To characterize the C1INH effects on TG and PG, the thrombin and plasmin concentration peaks and production rates were calculated. TM addition to donor plasma shifted the concentration dependence of C1INH on TG parameters from reduction to enhancement. Conversely, PG parameters were significantly reduced by 16 IU/ml in both the presence and absence of TM. Moderate C1INH concentration (3 IU/ml) reduced TG and PG in the absence of TM but did not significantly affect these parameters in the presence of TM. Finally, 3 IU/ml of C1INH reduced PG more so than TG in the absence of TM. The presented results suggest a mechanism by which C1INH could potentiate thrombosis by inhibition of fibrinolysis.


Subject(s)
Complement C1 Inhibitor Protein/pharmacology , Fibrinolysin/antagonists & inhibitors , Thrombin/drug effects , Thrombomodulin/physiology , Blood Coagulation , Blood Specimen Collection , Dose-Response Relationship, Drug , Fibrinolysin/biosynthesis , Fibrinolysis/drug effects , Healthy Volunteers , Humans , Thrombin/metabolism , Thrombosis/chemically induced
6.
Clin Immunol ; 191: 27-33, 2018 06.
Article in English | MEDLINE | ID: mdl-29562205

ABSTRACT

Thrombomodulin (TM, also known as CD141), which functions as an anticoagulant, is widely expressed on cell surface of a variety of cell types, including human blood cells as well as certain immune cells. To determine whether TM could be a potential marker for OSCC diagnosis as well as a molecular target for OSCC therapy, we examined the expression of TM in an oral cancer tissue microarray with 153 oral cancer tissues. Further, we also analyzed the expression of TM on DCs of 36 OSCC patients and 36 healthy donors. The expression of TM was determined using standard immunohistochemistry on a tissue microarray of 153 OSCC patients. Flow cytometric analyses were performed to determine the proportions of CD141+ DCs in the PBMC of 36 OSCC patients and 36 healthy donors. Clinicopathological correlations were performed based on the available clinical data. Our results showed that in the univariate analysis, high TM expression was significantly associated with well differentiation of tumor cells (P=.001), but not correlated with overall survival and disease-free survival (P>.05). In addition, CD141+ DCs were both present in OSCC patients and healthy donors with about 0.04%. There was no significant difference with the percentages of CD141+ DCs in the PBMC of OSCC patients and that of the normal control group (P>.05). This study indicates that TM expression might play the most critical role in the differentiation of OSCC tumors. Functional distinctions of CD141+ DCs in OSCC patients deserve further investigation to provide important therapeutic understandings for future immunotherapy.


Subject(s)
Mouth Neoplasms/immunology , Squamous Cell Carcinoma of Head and Neck/immunology , Thrombomodulin/physiology , Aged , Antigens, Surface/analysis , Cell Differentiation , Dendritic Cells/immunology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/pathology , Thrombomodulin/analysis , Tissue Array Analysis
7.
Thromb Haemost ; 116(6): 1022-1031, 2016 Nov 30.
Article in English | MEDLINE | ID: mdl-27604259

ABSTRACT

Antithrombin (AT) and thrombomodulin (TM) play important roles in the process of natural anticoagulation in vivo. Recently, we reported that the prothrombin Yukuhashi mutation (p.Arg596Leu) was associated with AT and TM resistance-related thrombophilia. To assess the AT and TM resistances associated with other missense mutations by single base substitution in the Arg596 codon, we generated recombinant variants (596Gln, 596Trp, 596Gly, and 596Pro) and investigated the effects on AT and TM anticoagulant functions. All variants except 596Pro were secreted in amounts comparable to that of the wild-type but exhibited variable procoagulant activities. After a 30-minute inactivation by AT, the relative residual activity of wild-type thrombin decreased to 15 ± 4.0 %, in contrast to values of all variants were maintained at above 80 %. The thrombin-AT complex formation, as determined by enzyme-linked immunosorbent assay, was reduced with all tested variants in the presence and absence of heparin. In the presence of soluble TM (sTM), the relative fibrinogen clotting activity of wild-type thrombin decreased to 16 ± 0.12 %, whereas that of tested variants was 37 %-56 %. In a surface plasmon resonance assay, missense Arg596 mutations reduced thrombin-TM affinity to an extent similar to the reduction of fibrinogen clotting inhibition. In the presence of sTM or cultured endothelial-like cells, APC generation was enhanced differently by variant thrombins in a thrombin-TM affinity-dependent manner. These data indicate that prothrombin Arg596 missense mutations lead to AT and TM resistance in the variant thrombins and suggest that prothrombin Arg596 is important for AT- and TM-mediated anticoagulation.


Subject(s)
Antithrombins/physiology , Mutation, Missense , Prothrombin/genetics , Thrombomodulin/physiology , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Thrombin/physiology
8.
PLoS One ; 11(8): e0160550, 2016.
Article in English | MEDLINE | ID: mdl-27512995

ABSTRACT

BACKGROUND: Treatment resistance and metastasis are the major causes of death among patients with colorectal cancer (CRC). Approximately 20% of surgically treated patients ultimately develop metastases during the follow-up period. Currently, the TNM system is the only available prognostic test. Therefore, the identification of new markers for CRC remains important. Thrombomodulin (TM), a glycoprotein, is involved in angiogenesis and has been linked to many malignant diseases. However, the function of TM in CRC remains unclear. METHODS: A total of 170 patients with CRC participated in this study. TM expression was analyzed via immunohistochemistry. Univariate (Kaplan-Meier) analysis was used to analyze patient outcomes, including overall survival (OS) and disease-free survival (DFS). TM expression was manipulated using shRNA or an overexpression system. Transwell migration assays, wound healing migration assays, and the xCELLigence biosensor system were used to detect cell proliferative and migratory capacities. RESULTS: TM expression in the tumor tissues significantly and positively correlated with the DFS and OS of non-metastatic patients with CRC (ps = 0.036 and 0.0218, respectively). Suppression of TM expression increased the proliferation and migration of DLD-1 cells. TM overexpression reduced the cells' proliferative and migratory capacities. Cyclooxygenase (COX)-2 expression was up-regulated following TM silencing. Furthermore, the association between the migration of colon cancer cells and the levels of TM and epithelial-to-mesenchymal transition (EMT) markers (fibronectin, vimentin and ezrin) was confirmed in HT29 and DLD-1 cells. CONCLUSIONS: Our study demonstrates that patients with non-metastatic CRC display low TM expression in their tumors and exhibit reduced DFS and OS. The enhanced expression of mesenchymal markers and COX-2 may be involved in the mechanisms that underlie recurrence in patients with cancer displaying low TM expression.


Subject(s)
Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Thrombomodulin/physiology , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , Cell Movement/genetics , Cell Proliferation/genetics , Cyclooxygenase 2/metabolism , Disease-Free Survival , Humans , Prognosis , Survival Analysis , Thrombomodulin/genetics , Thrombomodulin/metabolism , Tumor Cells, Cultured , Up-Regulation
9.
Biosci Trends ; 10(3): 231-4, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27210597

ABSTRACT

Thrombomodulin (TM) is a transmembrane protein expressed on vascular endothelial cells. TM has anticoagulant and anti-inflammatory properties. It has recently been reported that TM modulates complement, an immune effector system that destroys pathogens and is also involved in inflammation. TM was demonstrated to enhance the degradation of C3b into iC3b by factor I and factor H, indicating that its role is in negative regulation in the alternative pathway of the complement system. In this study, we examined the effects of recombinant human soluble TM protein composed of the extracellular domains (rTM) on the alternative pathway. The degradation of C3b into iC3b by factor I and factor H was enhanced by rTM as assessed by SDS-PAGE, confirming the previous observation. We also found that rTM enhances the cleavage of C3 into C3b as a result of activation of the alternative pathway. These results indicate that TM has both activating and inactivating functions in the alternative pathway.


Subject(s)
Complement Pathway, Alternative , Thrombomodulin/physiology , Complement C3b/metabolism , Complement Factor H/metabolism , Fibrinogen/metabolism , Humans , Protein Domains , Proteolysis , Recombinant Fusion Proteins/pharmacology , Thrombomodulin/chemistry , Thrombomodulin/metabolism
10.
Ann Biol Clin (Paris) ; 74(2): 203-12, 2016.
Article in French | MEDLINE | ID: mdl-26806287

ABSTRACT

Cirrhosis is associated with complex hemostatic modifications. Most coagulation factors, either procoagulants or anticoagulants, are reduced. Conventional coagulation tests (prothrombin time, activated partial thromboplastin time) don't allow to precisely identify the thrombotic risk as they are not sensible to coagulation inhibitors deficiencies. The aim of this study was to evaluate the coagulation in a population of cirrhotic patients using thrombinography. We analyzed the plasma samples from 30 cirrhotic patients (10 Child A, 10 Child B, Child C 10) compared to 10 healthy controls using thrombinography with and without thrombomodulin to sensiblise this test at the activated protein C pathway. The results of endogenous thrombin potential, the main parameter, expressed as a ratio (thrombinography with/without thrombomodulin) were significantly higher in cirrhotic patients (0.69 ± 0.16) than in controls (0.49 ± 0.10) which reflects a low sensibility to the action of thrombomodulin. This resistance increases with the severity of the disease assessed by the Child-Pugh score, demonstrating a potential hypercoagulable state. The results of the thrombinography challenge the dogma that cirrhotic patients are naturally "anticoagulated." These results highlight the potential interest of the thrombinography in the detection and monitoring of hypercoagulability in cirrhotic patient. Increasing hypercoagulability with the severity of the disease seems to be correlated with clinical observations since the occurrence of thrombosis is more common when cirrhosis is at an advanced stage.


Subject(s)
Liver Cirrhosis/blood , Thrombomodulin/physiology , Adult , Aged , Blood Coagulation Tests/methods , Case-Control Studies , Female , Humans , Liver Cirrhosis/pathology , Male , Middle Aged , Severity of Illness Index , Thrombomodulin/blood
11.
Semin Thromb Hemost ; 41(6): 549-55, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26270112

ABSTRACT

Forming an interface with virtually every other organ, endothelium has a strategic role in modulating vascular homeostasis. While its miscellany of functions includes regulation of vasomotor tone, promotion, and prevention of vascular growth, and modulation of inflammatory and hemostatic processes, it functions critically in maintaining the balance of hemostasis in health. Whereas endothelium has been recognized to influence all stages of hemostasis, new evidence suggests it to have a primary role for thrombin generation. Endothelial dysfunction is being increasingly appreciated in several pathological states and particularly, by virtue of its critical role in hemostasis, in causing thrombosis in a multitude of diseases.


Subject(s)
Endothelium, Vascular/physiology , Hemostasis/physiology , Homeostasis/physiology , Thrombosis/blood , Animals , Antigens, CD/physiology , Endothelial Cells/pathology , Endothelial Cells/physiology , Endothelial Protein C Receptor , Endothelium, Vascular/pathology , Hormones/metabolism , Humans , Lipoproteins/physiology , Paracrine Communication , Receptors, Cell Surface/physiology , Thrombin/biosynthesis , Thrombomodulin/physiology , Thrombophilia/blood , Thrombophilia/etiology , Thrombophilia/pathology , Thromboplastin/physiology , Thrombosis/etiology , Thrombosis/pathology , von Willebrand Factor/physiology
12.
Crit Care ; 19: 72, 2015 Feb 23.
Article in English | MEDLINE | ID: mdl-25886801

ABSTRACT

Knowing the pathophysiology of trauma-induced coagulopathy is important for the management of severely injured trauma patients. The aims of this review are to provide a summary of the recent advances in our understanding of thrombosis and hemostasis following trauma and to discuss the pathogenesis of disseminated intravascular coagulation (DIC) at an early stage of trauma. Local hemostasis and thrombosis respectively act to induce physiological wound healing of injuries and innate immune responses to damaged-self following trauma. However, if overwhelmed by systemic inflammation caused by extensive tissue damage and tissue hypoperfusion, both of these processes foster systemic DIC associated with pathological fibrin(ogen)olysis. This is called DIC with the fibrinolytic phenotype, which is characterized by the activation of coagulation, consumption coagulopathy, insufficient control of coagulation, and increased fibrin(ogen)olysis. Irrespective of microvascular thrombosis, the condition shows systemic thrombin generation as well as its activation in the circulation and extensive damage to the microvasculature endothelium. DIC with the fibrinolytic phenotype gives rise to oozing-type non-surgical bleeding and greatly affects the prognosis of trauma patients. The coexistences of hypothermia, acidosis, and dilution aggravate DIC and lead to so-called trauma-induced coagulopathy. He that would know what shall be must consider what has been. The Analects of Confucius.


Subject(s)
Disseminated Intravascular Coagulation/physiopathology , Hemostasis/physiology , Shock, Traumatic/physiopathology , Thrombosis/physiopathology , Wounds and Injuries/physiopathology , Animals , Antithrombins/metabolism , DNA, Mitochondrial/metabolism , Disseminated Intravascular Coagulation/diagnosis , Disseminated Intravascular Coagulation/metabolism , Fibrinolysis/physiology , HMGB1 Protein/metabolism , Histones/metabolism , Humans , Immunity, Innate/physiology , Lipoproteins/metabolism , Nucleosomes/metabolism , Protein C/metabolism , Protein S/metabolism , Shock, Traumatic/metabolism , Thrombin/metabolism , Thrombomodulin/physiology , Thrombosis/metabolism , Wound Healing/physiology , Wounds and Injuries/metabolism
13.
J Invest Dermatol ; 135(6): 1668-1675, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25651160

ABSTRACT

Keratinocyte-expressed thrombomodulin (TM) and the released soluble TM (sTM) have been demonstrated to promote wound healing. However, the effects of high glucose on TM expression in keratinocytes and the role of TM in diabetic ulcer remain unclear. In this study, we demonstrated that expressions of TM and Toll-like receptor 4 (TLR4) were both downregulated in high-glucose cultured human keratinocytes and in skin keratinocytes of diabetic patients. In addition, the wound-triggered upregulation of TM and sTM production was abolished in both high-glucose cultured human keratinocytes and streptozotocin-induced diabetic mouse skin. Furthermore, supplementation of recombinant sTM could increase TLR4 expression and promote cutaneous wound healing in both high-glucose cultured human keratinocytes and diabetic mice. However, in Tlr4-deleted mice, which exhibited delayed wound healing, the therapeutic benefit of recombinant sTM was abrogated. Moreover, our results showed that tumor necrosis factor-α (TNF-α) expression in keratinocytes was dose-dependently upregulated by glucose, and TNF-α treatment downregulated the expression of TM and TLR4. Taken together, high-glucose environment reduces the expression of TM and TLR4 in keratinocytes possibly through the action of TNF-α, and recombinant sTM can increase the TLR4 expression and promote wound healing under diabetic condition.


Subject(s)
Diabetes Complications/metabolism , Diabetes Mellitus, Experimental/metabolism , Thrombomodulin/physiology , Toll-Like Receptor 4/metabolism , Wound Healing , Animals , Cell Line, Tumor , Gene Deletion , Gene Expression Regulation , Glucose/chemistry , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Recombinant Proteins/metabolism , Skin/metabolism , Streptozocin/chemistry , Tumor Necrosis Factor-alpha/metabolism
14.
Cardiovasc Res ; 105(1): 107-17, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25388665

ABSTRACT

AIMS: The recombinant epidermal growth factor-like domain plus the serine/threonine-rich domain of thrombomodulin (rTMD23) promotes angiogenesis and accelerates the generation of activated protein C (APC), which facilitates angiogenesis. The aim of this study was to elucidate the molecular mechanisms underlying the angiogenic activity of rTMD23. METHODS AND RESULTS: We prepared rTMD23 and its mutants that did not possess the ability to promote APC generation and investigated their angiogenic activities in vitro and in vivo. rTMD23 mutants promoted proliferation, migration, and tube formation of human umbilical vein endothelial cells in vitro and induced neovascularization in vivo; these effects were similar to those exerted by wild-type rTMD23. To investigate its interaction with rTMD23, Type I fibroblast growth factor receptor (FGFR1) was precipitated along with syndecan-4 by rTMD23-conjugated Sepharose in human umbilical vein endothelial cells and FGFR1-expressing human embryonic kidney 293 cells. Additionally, the kinetics of the interaction between rTMD23 and FGFR1 were analysed using surface plasmon resonance. rTMD23-induced FGFR1 activation and tube formation were inhibited by an FGFR1-specific tyrosine kinase inhibitor, PD173074, or by knockdown of FGFR1 using siRNA technology. We observed an improvement in rat hindlimb recovery in an ischaemic model following rTMD23 treatment, and this was associated with increased neovascularization and FGFR1 phosphorylation. CONCLUSION: rTMD23 induced angiogenesis via FGFR1, a process that is independent of the APC pathway.


Subject(s)
Neovascularization, Physiologic/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Thrombomodulin/physiology , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/pharmacology , Animals , Corneal Neovascularization/etiology , Gene Knockdown Techniques , HEK293 Cells , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Ischemia/drug therapy , Ischemia/pathology , Ischemia/physiopathology , Mice , Mice, Inbred BALB C , Neovascularization, Physiologic/drug effects , Phosphorylation , Protein C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Syndecan-4/metabolism , Thrombomodulin/chemistry , Thrombomodulin/genetics
15.
Tumour Biol ; 35(6): 6047-54, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24633886

ABSTRACT

Thrombomodulin (TM) is highly expressed in endothelial cells and plays the key role in maintaining physical homeostasis. In addition, many pieces of evidence also show that TM contains the diagnostic value for malignant diseases. TM has been found to correlate with metastatic status in multiple cancers, but its role in prostate cancer progression remains unclear. TM expression was determined in prostate cancer cells (DU-145 and PC-3 cells) using real-time PCR and Western blotting. TM expression was manipulated in prostate cancer cells using TM-specific shRNA and an overexpression system. The proliferation, adhesion, and migratory ability of prostate cancer cells expressing various TM levels were determined using the x'Celligence biosensor system and a transwell migration assay. Higher levels of TM transcription and translation were found in DU-145 cells and were negatively correlated with the low migratory ability of DU-145 cells. After silencing TM expression in DU-145 cells, cell growth decreased, but cell adhesion and migration dramatically increased. TM overexpression in PC-3 cells reduced their metastatic ability. We investigated the possible mechanisms of this phenomenon and determined that the enhanced cell migration was mediated through the expression of E-cadherin and vimentin. TM may be a modulator of hormone-independent prostate cancer (HIPC) metastasis. The downregulation of TM expression enhanced the migratory ability of these cells via an increase in vimentin expression and a decrease in E-cadherin expression.


Subject(s)
Epithelial-Mesenchymal Transition , Prostatic Neoplasms/pathology , Thrombomodulin/physiology , Biomarkers , Cadherins/analysis , Calcium/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Male , Vimentin/analysis
16.
Int J Cardiol ; 172(2): 340-9, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24485615

ABSTRACT

BACKGROUND: Thrombomodulin (TM) is an endothelial cell (EC) membrane-bound anticoagulant protein that has novel direct cellular effects. TM is shed from EC and becomes soluble form (sTM) in plasma. Higher sTM levels in healthy subjects are associated with lower cardiovascular risk, suggesting that sTM possesses a protective role. The purpose of the study was to evaluate the effect of sTM on vascular endothelium. METHODS AND RESULTS: Apoptosis of cultured ECs was induced via serum starvation. EC-bound TM was released into the medium after serum starvation. The medium conditioned by serum-starved EC decreased apoptosis in another set of cultured EC. Direct treatment with sTM reduced EC apoptosis and decreased pro-apoptotic protein expression. TM knockdown in EC exacerbated the rate of serum starvation-induced apoptosis. Treatment of sTM activated the phosphatidylinositol 3-kinase (PI3 kinase)-protein kinase B/Akt survival pathway and suppressed the death pathway, c-Jun N-terminal kinase. We found that sTM also increased growth and reduced apoptosis of endothelial progenitor cells. CONCLUSIONS: EC-bound TM is released during stress-induced EC damage and becomes sTM, a paracrine factor that exerts anti-apoptotic activity. Our data indicate that sTM is not only an endothelial injury biomarker but also has cytoprotective effects on vascular endothelium.


Subject(s)
Apoptosis/drug effects , Endothelium, Vascular/drug effects , Thrombomodulin/physiology , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , In Situ Nick-End Labeling , Signal Transduction/drug effects , Thrombomodulin/metabolism , Umbilical Cord/cytology
18.
Transplantation ; 97(2): 138-47, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24150517

ABSTRACT

BACKGROUND: Among other mismatches between human and pig, incompatibilities in the blood coagulation systems hamper the xenotransplantation of vascularized organs. The provision of the porcine endothelium with human thrombomodulin (hTM) is hypothesized to overcome the impaired activation of protein C by a heterodimer consisting of human thrombin and porcine TM. METHODS: We evaluated regulatory regions of the THBD gene, optimized vectors for transgene expression, and generated hTM expressing pigs by somatic cell nuclear transfer. Genetically modified pigs were characterized at the molecular, cellular, histological, and physiological levels. RESULTS: A 7.6-kb fragment containing the entire upstream region of the porcine THBD gene was found to drive a high expression in a porcine endothelial cell line and was therefore used to control hTM expression in transgenic pigs. The abundance of hTM was restricted to the endothelium, according to the predicted pattern, and the transgene expression of hTM was stably inherited to the offspring. When endothelial cells from pigs carrying the hTM transgene--either alone or in combination with an aGalTKO and a transgene encoding the human CD46-were tested in a coagulation assay with human whole blood, the clotting time was increased three- to four-fold (P<0.001) compared to wild-type and aGalTKO/CD46 transgenic endothelial cells. This, for the first time, demonstrated the anticoagulant properties of hTM on porcine endothelial cells in a human whole blood assay. CONCLUSIONS: The biological efficacy of hTM suggests that the (multi-)transgenic donor pigs described here have the potential to overcome coagulation incompatibilities in pig-to-primate xenotransplantation.


Subject(s)
Animals, Genetically Modified , Endothelial Cells/metabolism , Regulatory Sequences, Nucleic Acid , Swine/genetics , Thrombomodulin/genetics , Animals , Genetic Vectors , Humans , Membrane Cofactor Protein/analysis , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Thrombomodulin/physiology , Transplantation, Heterologous
19.
Chin Med J (Engl) ; 126(14): 2636-40, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23876886

ABSTRACT

BACKGROUND: Stent thrombosis is one of severe complications after sirolimus-eluting stent implantation. Rapamycin (sirolimus) promotes arterial thrombosis in in vivo studies. However, the underlying molecular and transcriptional mechanisms of this adverse effect have not been thoroughly investigated. This study was designed to examine the effects of rapamycin on the expression of the gene, Kruppel-like factor 2 (KLF2), and its transcriptional targets in mice. METHODS: Mice were randomly divided into four groups: the control group (intraperitoneal injection with 2.5% of dimethyl sulfoxide (DMSO) only), rapamycin group (intraperitoneal injection with 2 mg/kg of rapamycin only), Ad-LacZ + rapamycin group (carotid arterial incubation with Ad-LacZ plus intraperitoneal injection with 2 mg/kg of rapamycin 10 days later), and Ad-KLF2 + rapamycin group (carotid arterial incubation with Ad-KLF2 plus intraperitoneal injection with 2 mg/kg rapamycin 10 days later). The carotid arterial thrombosis formation was induced by FeCl3 and the time of arterial thrombosis was determined. Finally, the RNA and protein of carotid arteries were extracted for KLF2, tissue factor (TF), plasminogen activator inhibitor-1 (PAI-1), endothelial nitric oxide synthase (eNOS), thrombomodulin (TM) mRNA and protein analysis. RESULTS: Compared with controls, treatment with rapamycin inhibited KLF2, eNOS and TM mRNA and protein expression, and enhanced TF and PAI-1 mRNA and protein expression, and shortened time to thrombotic occlusion from (1282 ± 347) seconds to (715 ± 120) seconds (P < 0.01) in vivo. Overexpression of KLF2 strongly reversed rapamycin-induced effects on KLF2, eNOS, TM, TF and PAI-1 expression. KLF2 overexpression increased the time to thrombotic occlusion to control levels in vivo. CONCLUSIONS: Rapamycin induced an inhibition of KLF2 expression and an imbalance of anti- and pro-thrombotic gene expression, which promoted arterial thrombosis in vivo. Overexpression of KLF2 increased KLF2 expression and reversed time to thrombosis in vivo.


Subject(s)
Drug-Eluting Stents/adverse effects , Kruppel-Like Transcription Factors/physiology , Sirolimus/pharmacology , Thrombosis/chemically induced , Animals , Carotid Arteries/metabolism , Kruppel-Like Transcription Factors/analysis , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/physiology , Plasminogen Activator Inhibitor 1/physiology , Thrombomodulin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...