Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33836597

RESUMO

Thrombomodulin (TM) is a thrombin receptor on endothelial cells that is involved in promoting activation of the anticoagulant protein C pathway during blood coagulation. TM also exerts protective anti-inflammatory properties through a poorly understood mechanism. In this study, we investigated the importance of TM signaling to cellular functions by deleting it from endothelial cells by CRISPR-Cas9 technology and analyzed the resultant phenotype of TM-deficient (TM-/- ) cells. Deficiency of TM in endothelial cells resulted in increased basal permeability and hyperpermeability when stimulated by thrombin and TNF-α. The loss of the basal barrier permeability function was accompanied by increased tyrosine phosphorylation of VE-cadherin and reduced polymerization of F-actin filaments at cellular junctions. A significant increase in basal NF-κB signaling and expression of inflammatory cell adhesion molecules was observed in TM-/- cells that resulted in enhanced adhesion of leukocytes to TM-/- cells in flow chamber experiments. There was also a marked increase in expression, storage, and release of the von Willebrand factor (VWF) and decreased storage and release of angiopoietin-2 in TM-/- cells. In a flow chamber assay, isolated platelets adhered to TM-/- cells, forming characteristic VWF-platelet strings. Increased VWF levels and inflammatory foci were also observed in the lungs of tamoxifen-treated ERcre-TMf/f mice. Reexpression of the TM construct in TM-/- cells, but not treatment with soluble TM, normalized the cellular phenotype. Based on these results, we postulate cell-bound TM endows a quiescent cellular phenotype by tightly regulating expression of procoagulant, proinflammatory, and angiogenic molecules in vascular endothelial cells.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Trombomodulina/metabolismo , Angiopoietina-2/metabolismo , Animais , Plaquetas/citologia , Permeabilidade Capilar , Adesão Celular , Células Endoteliais/citologia , Endotélio Vascular/citologia , Humanos , Inflamação , Leucócitos/citologia , Pulmão/metabolismo , Camundongos , Receptor PAR-1/metabolismo , Trombina/metabolismo , Trombomodulina/deficiência , Trombomodulina/genética , Fator de von Willebrand/metabolismo
2.
Blood ; 137(7): 977-982, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32870264

RESUMO

Excess platelet activation by extracellular vesicles (EVs) results in trophoblast inflammasome activation, interleukin 1ß (IL-1ß) activation, preeclampsia (PE), and partial embryonic lethality. Embryonic thrombomodulin (TM) deficiency, which causes embryonic lethality hallmarked by impaired trophoblast proliferation, has been linked with maternal platelet activation. We hypothesized that placental TM loss, platelet activation, and embryonic lethality are mechanistically linked to trophoblast inflammasome activation. Here, we uncover unidirectional interaction of placental inflammasome activation and reduced placental TM expression: although inflammasome inhibition did not rescue TM-null embryos from lethality, the inflammasome-dependent cytokine IL-1ß reduced trophoblast TM expression and impaired pregnancy outcome. EVs, known to induce placental inflammasome activation, reduced trophoblast TM expression and proliferation. Trophoblast TM expression correlated negatively with IL-1ß expression and positively with platelet numbers and trophoblast proliferation in human PE placentae, implying translational relevance. Soluble TM treatment or placental TM restoration ameliorated the EV-induced PE-like phenotype in mice, preventing placental thromboinflammation and embryonic death. The lethality of TM-null embryos is not a consequence of placental NLRP3 inflammasome activation. Conversely, EV-induced placental inflammasome activation reduces placental TM expression, promoting placental and embryonic demise. These data identify a new function of placental TM in PE and suggest that soluble TM limits thromboinflammatory pregnancy complications.


Assuntos
Morte Fetal/etiologia , Inflamassomos/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Trombomodulina/deficiência , Animais , Divisão Celular , Regulação para Baixo , Vesículas Extracelulares , Feminino , Genes Letais , Humanos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Placenta/irrigação sanguínea , Ativação Plaquetária , Plasma Rico em Plaquetas , Gravidez , Resultado da Gravidez , Receptores de Trombina , Proteínas Recombinantes/farmacologia , Trombomodulina/antagonistas & inibidores , Trombomodulina/biossíntese , Trombomodulina/genética , Trofoblastos/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 40(12): 2837-2844, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33028093

RESUMO

OBJECTIVE: THBD (thrombomodulin) is part of the anticoagulant protein C-system that acts at the endothelium and is involved in anti-inflammatory and barrier-stabilizing processes. A recombinant soluble form of THBD was shown to have protective effects in different organs, but how the endogenous THBD is regulated during ischemia, particularly in the brain is not known to date. The aim of this study was to investigate the role of THBD, especially in brain endothelial cells, during ischemic stroke. Approach and Results: To induce ischemic brain damage, we occluded the middle cerebral artery of mice. We found an increased endothelial expression of Thbd in the peri-infarct area, whereas in the core of the ischemic tissue Thbd expression was decreased compared with the contralateral side. We generated a novel Cre/loxP-based mouse line that allows for the inducible deletion of Thbd specifically in brain endothelial cells, which worsened stroke outcome 48 hours after middle cerebral artery occlusion. Unexpectedly, we found no signs of increased coagulation, thrombosis, or inflammation in the brain but decreased vessel diameters and impaired angiogenesis in the peri-infarct area that led to a reduced overall vessel length 1 week after stroke induction. CONCLUSIONS: Endogenous THBD acts as a protective factor in the brain during ischemic stroke and enhances vessel diameter and proliferation. These previously unknown properties of THBD could offer new opportunities to affect vessel function after ischemia and thereby improve stroke outcome.


Assuntos
Encéfalo/irrigação sanguínea , Células Endoteliais/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Neovascularização Fisiológica , Trombomodulina/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Trombomodulina/deficiência
4.
Arterioscler Thromb Vasc Biol ; 35(11): 2412-22, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26338301

RESUMO

OBJECTIVE: Thrombomodulin (TM), a glycoprotein constitutively expressed in the endothelium, is well known for its anticoagulant and anti-inflammatory properties. Paradoxically, we recently found that monocytic membrane-bound TM (ie, endogenous TM expression in monocytes) triggers lipopolysaccharide- and gram-negative bacteria-induced inflammatory responses. However, the significance of membrane-bound TM in chronic sterile vascular inflammation and the development of abdominal aortic aneurysm (AAA) remains undetermined. APPROACH AND RESULTS: Implicating a potential role for membrane-bound TM in AAA, we found that TM signals were predominantly localized to macrophages and vascular smooth muscle cells in human aneurysm specimens. Characterization of the CaCl2-induced AAA in mice revealed that during aneurysm development, TM expression was mainly localized in infiltrating macrophages and vascular smooth muscle cells. To investigate the function of membrane-bound TM in vivo, transgenic mice with myeloid- (LysMcre/TM(flox/flox)) and vascular smooth muscle cell-specific (SM22-cre(tg)/TM(flox/flox)) TM ablation and their respective wild-type controls (TM(flox/flox) and SM22-cre(tg)/TM(+/+)) were generated. In the mouse CaCl2-induced AAA model, deficiency of myeloid TM, but not vascular smooth muscle cell TM, inhibited macrophage accumulation, attenuated proinflammatory cytokine and matrix metalloproteinase-9 production, and finally mitigated elastin destruction and aortic dilatation. In vitro TM-deficient monocytes/macrophages, versus TM wild-type counterparts, exhibited attenuation of proinflammatory mediator expression, adhesion to endothelial cells, and generation of reactive oxygen species. Consistently, myeloid TM-deficient hyperlipidemic mice (ApoE(-/-)/LysMcre/TM(flox/flox)) were resistant to AAA formation induced by angiotensin II infusion, along with reduced macrophage infiltration, suppressed matrix metalloproteinase activities, and diminished oxidative stress. CONCLUSIONS: Membrane-bound TM in macrophages plays an essential role in the development of AAA by enhancing proinflammatory mediator elaboration, macrophage recruitment, and oxidative stress.


Assuntos
Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/metabolismo , Aortite/metabolismo , Membrana Celular/metabolismo , Macrófagos Peritoneais/metabolismo , Trombomodulina/metabolismo , Angiotensina II , Animais , Aorta Abdominal/imunologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/imunologia , Aortite/induzido quimicamente , Aortite/genética , Aortite/imunologia , Cloreto de Cálcio , Membrana Celular/imunologia , Células Cultivadas , Quimiotaxia , Modelos Animais de Doenças , Elastina/metabolismo , Células Endoteliais da Veia Umbilical Humana/imunologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos Peritoneais/imunologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/imunologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/metabolismo , Estresse Oxidativo , Interferência de RNA , Estudos Retrospectivos , Transdução de Sinais , Trombomodulina/deficiência , Trombomodulina/genética , Fatores de Tempo , Transfecção
5.
Crit Care Med ; 42(3): e221-30, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24399241

RESUMO

OBJECTIVE: Thrombomodulin is a multidomain receptor primarily expressed by vascular endothelium. The lectin-like domain of thrombomodulin has anti-inflammatory properties. In this study, we investigated the role of the thrombomodulin lectin-like domain in the host response to Gram-negative sepsis caused by Burkholderia pseudomallei, a "Tier 1" biothreat agent and the causative agent of melioidosis, a common form of community-acquired sepsis in Southeast Asia. DESIGN: Animal study. SETTING: University research laboratory. SUBJECTS: Wild-type mice and mice lacking the lectin-like domain of thrombomodulin. INTERVENTIONS: Mice were intranasally infected with live B. pseudomallei and killed after 24, 48, or 72 hours for harvesting of lungs, liver, spleen, and blood. Additionally, survival studies were performed. MEASUREMENTS AND MAIN RESULTS: Following exposure to B. pseudomallei, mice lacking the lectin-like domain of thrombomodulin showed a survival advantage, accompanied by decreased bacterial loads in the blood, lungs, liver, and spleen. Although lung histopathology did not differ between groups, mice lacking the lectin-like domain of thrombomodulin displayed strongly attenuated systemic inflammation, as reflected by lower plasma cytokine levels, maintenance of normal kidney and liver function, histologic evidence of reduced organ damage, and damage to the spleen. CONCLUSIONS: This study reveals for the first time a detrimental role for the thrombomodulin lectin-like domain in the host response to sepsis caused by a clinically relevant Gram-negative pathogen.


Assuntos
Pulmão/patologia , Melioidose/patologia , Melioidose/prevenção & controle , Pneumonia Bacteriana/prevenção & controle , Trombomodulina/metabolismo , Animais , Carga Bacteriana , Biópsia por Agulha , Líquido da Lavagem Broncoalveolar/microbiologia , Burkholderia pseudomallei/patogenicidade , Citocinas/metabolismo , Modelos Animais de Doenças , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Estimativa de Kaplan-Meier , Lectinas/metabolismo , Pulmão/microbiologia , Masculino , Melioidose/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Bacteriana/fisiopatologia , Distribuição Aleatória , Estatísticas não Paramétricas , Taxa de Sobrevida , Trombomodulina/deficiência
6.
Curr Med Chem ; 21(18): 2025-34, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24372210

RESUMO

Thrombomodulin (TM) is a membrane protein mainly expressed by endothelial cells. It is part of the anticoagulant protein C system but recently several effects were discovered which occur independently of protein C activation. TM binds thrombin and promotes the cleavage of protein C and the thrombin activatable fibrinolysis inhibitor (TAFI), thereby inhibiting coagulation and fibrinolysis. Additionally, it interferes with inflammation, stabilizes barrier function, and increases blood flow under pathological conditions. Recombinant soluble TM protects against tissue damage and partially restores normal function after ischemia in several organs. Recently, it was shown to reduce the infarct size in stroke models. Compared to other anticoagulant compounds the risk of bleeding seems to be smaller in animals and humans treated with soluble TM. With its multiple actions TM represents a new candidate for stroke treatment. In this review we focus on the effects of TM in coagulation, inflammation, and on its protective roles in the prevention of ischemic brain damage.


Assuntos
Coagulação Sanguínea , Acidente Vascular Cerebral/metabolismo , Trombomodulina/deficiência , Animais , Anticoagulantes/uso terapêutico , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Trombomodulina/metabolismo
7.
J Immunol ; 189(7): 3661-8, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22942429

RESUMO

Shiga toxin (Stx)-producing Escherichia coli is a primary cause of diarrhea-associated hemolytic uremic syndrome (HUS), a disorder of thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure. The pathophysiology of renal microvascular thrombosis in Stx-HUS is still ill-defined. Based on evidence that abnormalities in thrombomodulin (TM), an anticoagulant endothelial glycoprotein that modulates complement and inflammation, predispose to atypical HUS, we assessed whether impaired TM function may adversely affect evolution of Stx-HUS. Disease was induced by coinjection of Stx2/LPS in wild-type mice (TM(wt/wt)) and mice that lack the lectin-like domain of TM (TM(LeD/LeD)), which is critical for its anti-inflammatory and cytoprotective properties. After Stx2/LPS, TM(LeD/LeD) mice exhibited more severe thrombocytopenia and renal dysfunction than TM(wt/wt) mice. Lack of lectin-like domain of TM resulted in a stronger inflammatory reaction after Stx2/LPS with more neutrophils and monocytes/macrophages infiltrating the kidney, associated with PECAM-1 and chemokine upregulation. After Stx2/LPS, intraglomerular fibrin(ogen) deposits were detected earlier in TM(LeD/LeD) than in TM(wt/wt) mice. More abundant fibrin(ogen) deposits were also found in brain and lungs. Under basal conditions, TM(LeD/LeD) mice exhibited excess glomerular C3 deposits, indicating impaired complement regulation in the kidney that could lead to local accumulation of proinflammatory products. TM(LeD/LeD) mice with HUS had a higher mortality rate than TM(wt/wt) mice. If applicable to humans, these findings raise the possibility that genetic or acquired TM defects might have an impact on the severity of microangiopathic lesions after exposure to Stx-producing E. coli infections and raise the potential for using soluble TM in the treatment of Stx-HUS.


Assuntos
Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/imunologia , Lectinas/deficiência , Toxina Shiga/administração & dosagem , Trombomodulina/deficiência , Trombomodulina/genética , Animais , Síndrome Hemolítico-Urêmica/induzido quimicamente , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Estrutura Terciária de Proteína/genética , Índice de Gravidade de Doença , Trombomodulina/fisiologia
8.
Thromb Haemost ; 108(6): 1141-53, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23014597

RESUMO

Coagulation and complement regulators belong to two interactive systems constituting emerging mechanisms of diabetic nephropathy. Thrombomodulin (TM) regulates both coagulation and complement activation, in part through discrete domains. TM's lectin like domain dampens complement activation, while its EGF-like domains independently enhance activation of the anti-coagulant and cytoprotective serine protease protein C (PC). A protective effect of activated PC in diabetic nephropathy is established. We hypothesised that TM controls diabetic nephropathy independent of PC through its lectin-like domain by regulating complement. Diabetic nephropathy was analysed in mice lacking TM's lectin-like domain (TMLeD/LeD) and controls (TMwt/wt). Albuminuria (290 µg/mg vs. 166 µg/mg, p=0.03) and other indices of experimental diabetic nephropathy were aggravated in diabetic TMLeD/LeD mice. Complement deposition (C3 and C5b-9) was markedly increased in glomeruli of diabetic TMLeD/LeD mice. Complement inhibition with enoxaparin ameliorated diabetic nephropathy in TMLeD/LeD mice (e.g. albuminuria 85 µg/mg vs. 290 µg/mg, p<0.001). In vitro TM's lectin-like domain cell-autonomously prevented glucose-induced complement activation on endothelial cells and - notably - on podocytes. Podocyte injury, which was enhanced in diabetic TMLeD/LeD mice, was reduced following complement inhibition with enoxaparin. The current study identifies a novel mechanism regulating complement activation in diabetic nephropathy. TM's lectin-like domain constrains glucose-induced complement activation on endothelial cells and podocytes and ameliorates albuminuria and glomerular damage in mice.


Assuntos
Nefropatias Diabéticas/etiologia , Trombomodulina/química , Trombomodulina/fisiologia , Animais , Linhagem Celular , Ativação do Complemento/fisiologia , Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/deficiência , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/fisiologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/fisiopatologia , Nefropatias Diabéticas/prevenção & controle , Células Endoteliais/imunologia , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Glomérulos Renais/imunologia , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Podócitos/imunologia , Podócitos/patologia , Podócitos/fisiologia , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombomodulina/deficiência , Trombomodulina/genética
9.
Blood ; 112(3): 585-91, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18490515

RESUMO

Absence of the blood coagulation inhibitor thrombomodulin (Thbd) from trophoblast cells of the mouse placenta causes a fatal arrest of placental morphogenesis. The pathogenesis of placental failure requires tissue factor, yet is not associated with increased thrombosis and persists in the absence of fibrinogen. Here, we examine the role of alternative targets of coagulation that might contribute to the placental failure and death of Thbd(-/-) embryos. We demonstrate that genetic deficiency of the protease-activated receptors, Par1 or Par2, in the embryo and trophoblast cells does not prevent the death of Thbd(-/-) embryos. Similarly, genetic ablation of the complement pathway or of maternal immune cell function does not decrease fetal loss. In contrast, Par4 deficiency of the mother, or the absence of maternal platelets, restores normal development in one-third of Thbd-null embryos. This finding generates new evidence implicating increased procoagulant activity and thrombin generation in the demise of thrombomodulin-null embryos, and suggests that platelets play a more prominent role in placental malfunction associated with the absence of thrombomodulin than fibrin formation. Our findings demonstrate that fetal prothrombotic mutations can cause localized activation of maternal platelets at the feto-maternal interface in a mother with normal hemostatic function.


Assuntos
Plaquetas/fisiologia , Doenças Placentárias/etiologia , Receptores Ativados por Proteinase/fisiologia , Trombomodulina/deficiência , Animais , Coagulação Sanguínea , Embrião de Mamíferos , Feminino , Troca Materno-Fetal , Camundongos , Camundongos Knockout , Mães , Gravidez , Receptores Ativados por Proteinase/deficiência , Trombofilia
10.
J Thromb Haemost ; 6(1): 111-7, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17973652

RESUMO

BACKGROUND AND OBJECTIVE: Tissue factor pathway inhibitor (TFPI) and thrombomodulin (TM) are endothelial-associated anticoagulant proteins thought to control hemostasis in specific vascular beds. Here, we have examined the consequences of TFPI deficiency in the presence of a compounding procoagulant state caused by reduced TM function. METHODS AND RESULTS: TFPI(+/-)/TM(pro/pro) mice are born at less than expected frequency in either TFPI(+/-)/TM(pro/+) or TM(pro/pro) mothers but are born at near the expected frequency in TM(pro/+) mothers. Adult TFPI(+/-)/TM(pro/pro) mice have elevated thrombin-antithrombin complex and increased thrombus volume in an electrical injury model of venous thrombosis. In striking contrast to mice with single deficiency of TFPI or TM, TFPI(+/-)/TM(pro/pro) mice exhibit augmented fibrin deposition not only in the liver, but also in the cerebral microvasculature. CONCLUSIONS: TFPI(+/-)/TM(pro/pro) mice exhibit partial intrauterine lethality when carried by mothers with an underlying prothrombotic state, providing the first experimental evidence in an animal model that TFPI-dependent control of hemostasis in the vascular bed of the placenta fulfills a critical role for successful pregnancy outcome. In addition to the placenta, partial TFPI deficiency interacts with decreased TM function in an organ selective manner to produce fibrin deposition in other specific vascular beds, the liver and brain.


Assuntos
Fibrina/metabolismo , Lipoproteínas/deficiência , Trombomodulina/deficiência , Trombofilia/etiologia , Animais , Circulação Cerebrovascular , Feminino , Genótipo , Lipoproteínas/genética , Fígado/irrigação sanguínea , Camundongos , Microcirculação , Especificidade de Órgãos , Placenta/irrigação sanguínea , Gravidez , Resultado da Gravidez , Trombomodulina/genética
11.
Thromb Haemost ; 92(3): 467-77, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15351842

RESUMO

This review describes animal models of TM-deficiency that cause thrombosis in mice. Thrombomodulin (TM) is a key component of the protein C anticoagulant pathway by facilitating the activation of protein C by thrombin. In addition, TM integrates fibrinolytic and anti-inflammatory responses in a manner that is in part independent of protein C and thrombin. A series of genetically modified mouse strains is available in which the various and distinct functions of TM have been altered by means of site-directed mutagenesis of the TM gene locus (Thbd). The focus of the current review is the pathological activation of the hemostatic mechanism in mice with altered TM function (the pathologic activation of the hemostatic mechanism). The analysis of these mouse models has revealed novel and in part organ-specific functions of TM, most notably in the vascular bed of the placenta. In these mouse models, the severity and phenotypic expression of thrombosis is highly variable and is dependent on interaction with secondary genetic or environmental modifiers. The mutant mouse strains replicate important aspects of thrombophilia and thrombosis in humans, and provide a valuable resource to validate existing, and develop novel concepts of disease mechanisms in human patients.


Assuntos
Modelos Animais de Doenças , Trombomodulina/fisiologia , Trombose/etiologia , Animais , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Feminino , Camundongos , Placenta/irrigação sanguínea , Placenta/patologia , Gravidez , Proteína C/genética , Proteína C/metabolismo , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/patologia , Trombomodulina/deficiência
12.
Basic Res Cardiol ; 98(6): 347-52, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14556079

RESUMO

Thrombomodulin (TM) bound thrombin initiates the protein C anticoagulant pathway and defects in TM result in enhanced coagulation. Recent studies suggest a role for TM in arterial vascular disease. In order to corroborate this association we studied arterial thrombus formation in mice with a functional TM defect. We used mice homozygous for a (404)Glu-to-Pro mutation in the TM gene (TM(pro/pro)) and compared these with wildtype littermates in a model of FeCl(3) induced carotid artery thrombosis. Time-to-occlusion (TTO) was assessed by arterial blood flow measurement, using a Doppler flow probe. Complete occlusion occurred in 8/10 (80%) TM(pro/pro) mice and in 3/11 (27%) littermate controls. Mean time to occlusion (TTO) [+/- SE] was 767 +/- 196 s in the F2-TM(pro/pro) mice, versus 1507 +/- 159 s in controls (p = 0.007, Mann Whitney U test). Histology and immunostaining for tissue factor did not reveal any differences in thrombus morphology or thrombogenicity between the two groups. These data confirm and extend the finding that a functional deficiency in TM results in enhanced thrombus formation in a murine model of carotid artery thrombosis and support a role for TM defects in arterial thrombotic disease.


Assuntos
Artérias Carótidas/patologia , Trombose das Artérias Carótidas/patologia , Modelos Animais de Doenças , Trombomodulina/deficiência , Animais , Hemodinâmica , Camundongos , Camundongos Transgênicos
13.
Am J Pathol ; 160(6): 2063-72, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12057911

RESUMO

Microvascular injury is believed to be mechanistically involved in radiation fibrosis, but direct molecular links between endothelial dysfunction and radiation fibrosis have not been established in vivo. We examined radiation-induced changes in endothelial thrombomodulin (TM) and protease-activated receptor-1 (PAR-1) in irradiated intestine, and their relationship to structural, cellular, and molecular aspects of radiation injury. Rat small intestine was locally exposed to fractionated X-radiation. Structural injury was assessed 24 hours and 2, 6, and 26 weeks after the last radiation fraction using quantitative histology and morphometry. TM, neutrophils, transforming growth factor-beta, and collagens I and III were assessed by quantitative immunohistochemistry. PAR-1 protein was localized immunohistochemically, and cells expressing TM or PAR-1 transcript were identified by in situ hybridization. Steady-state PAR-1 mRNA levels in intestinal smooth muscle were determined using laser capture microdissection and competitive reverse transcriptase-polymerase chain reaction. Radiation caused a sustained, dose-dependent decrease in microvascular TM. The number of TM-positive vessels correlated with all parameters of radiation enteropathy and, after adjusting for radiation dose and observation time in a statistical model, remained independently associated with neutrophil infiltration, intestinal wall thickening, and collagen I accumulation. PAR-1 immunoreactivity and transcript increased in vascular and intestinal smooth muscle cells in irradiated intestine. PAR-1 mRNA increased twofold in irradiated intestinal smooth muscle. Intestinal irradiation up-regulates PAR-1 and causes a dose-dependent, sustained deficiency of microvascular TM that is independently associated with the severity of radiation toxicity. Interventions aimed at preserving or restoring endothelial TM or blocking PAR-1 should be explored as strategies to increase the therapeutic ratio in clinical radiation therapy.


Assuntos
Enteropatias/fisiopatologia , Intestinos/efeitos da radiação , Lesões Experimentais por Radiação/fisiopatologia , Receptores de Trombina/biossíntese , Trombomodulina/deficiência , Regulação para Cima , Animais , Doença Crônica , Endotélio/fisiopatologia , Fibrose , Hibridização In Situ , Masculino , Microcirculação , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor PAR-1
14.
Diabetes ; 51(6): 1957-63, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12031986

RESUMO

Human diabetic neuropathy is multifactorial in etiology, with ischemia as a final common pathology. Although impaired vascular endothelial cell function in diabetic microvascular injury is established, the role of thrombomodulin (TM)-dependent protein C antithrombotic mechanism in the pathogenesis of neuropathy is unclear. This neuropathologic case-control study investigated whether vascular endothelial TM expression is deficient in peripheral nerve microvessels in diabetic neuropathy. Sural nerve biopsies from 7 patients with diabetic neuropathy and 10 with axonal neuropathy without vasculopathy were immunostained with anti-TM and anti-von Willebrand factor (vWF; an endothelial cell marker) antibodies. The proportion of TM-positive microvessels was expressed relative to total vWF-staining vessels, according to vessel caliber and regional distribution within the nerve. In diabetic nerves compared with reference controls, the proportion of TM-positive endoneurial microvessels was 15-fold lower (0.02 vs. 0.30 in diabetic nerves vs. controls, P < 0.004), and the proportion of small-caliber epineurial microvessels was 10-fold lower (0.04 vs. 0.43, P < 0.001). No TM expression was detected at the perineurium in diabetic or control nerves. We demonstrate a substantial reduction of vascular endothelial TM expression throughout human diabetic neuropathy. These findings suggest that an impaired native TM-dependent protein C antithrombotic mechanism may contribute to microvascular ischemia in the pathogenesis of diabetic neuropathy.


Assuntos
Neuropatias Diabéticas/etiologia , Microcirculação/química , Nervos Periféricos/irrigação sanguínea , Trombomodulina/deficiência , Adulto , Idoso , Axônios/patologia , Biópsia , Estudos de Casos e Controles , Neuropatias Diabéticas/patologia , Endotélio Vascular/química , Feminino , Humanos , Isquemia/complicações , Masculino , Microcirculação/patologia , Pessoa de Meia-Idade , Proteína C/fisiologia , Nervo Sural/patologia , Trombomodulina/fisiologia , Trombose/prevenção & controle , Fator de von Willebrand/análise
15.
Circ Res ; 90(2): 205-12, 2002 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-11834714

RESUMO

Thrombosis is the major cause of early vein graft failure. Our aim was to determine whether alterations in the expression of the anticoagulant proteins, thrombomodulin (TM) and the endothelial cell protein C receptor (EPCR), impair endothelial thromboresistance that may contribute to vein graft failure. Immunohistochemical staining of autologous rabbit vein graft sections revealed that the expression of TM, but not EPCR, was reduced significantly early after graft implantation. Western blot analysis revealed that TM expression was reduced by >95% during the first 2 weeks after implantation, with gradual but incomplete recovery by 42 days. This resulted in up to a 95% reduction in the capacity of the grafts to activate protein C and was associated with an increase in bound thrombin activity, which peaked on day 7 at 28.7 +/- 3.8 mU/cm(2) and remained elevated for more than 14 days. Restoration of TM expression using adenovirus vector-mediated gene transfer significantly enhanced the capacity of grafts to activate protein C and reduced bound thrombin activity on day 7 to levels comparable to that of normal veins (5.7 +/- 0.4 versus 5.2 +/- 1.1 mU/cm(2), respectively, P=0.74). Surprisingly, neointima formation was not affected by this inhibition of local thrombin activity. These data suggest that the early loss of TM expression significantly impairs vein graft thromboresistance and results in enhanced local thrombin generation. Although enhanced local thrombin generation may predispose to early vein graft failure due to thrombosis, it does not seem to contribute significantly to late vein graft failure due to neointimal hyperplasia.


Assuntos
Fatores de Coagulação Sanguínea , Veias Jugulares/metabolismo , Receptores de Superfície Celular/metabolismo , Trombomodulina/metabolismo , Trombose Venosa/metabolismo , Animais , Western Blotting , Artérias Carótidas/cirurgia , Modelos Animais de Doenças , Fibrinolíticos/farmacologia , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Imuno-Histoquímica , Veias Jugulares/efeitos dos fármacos , Veias Jugulares/patologia , Veias Jugulares/transplante , Masculino , Proteína C/metabolismo , Coelhos , Trombina/antagonistas & inibidores , Trombina/metabolismo , Trombomodulina/deficiência , Trombomodulina/genética , Transdução Genética , Transplante Autólogo/efeitos adversos , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/patologia , Procedimentos Cirúrgicos Vasculares/efeitos adversos , Trombose Venosa/etiologia , Trombose Venosa/patologia
16.
Thromb Haemost ; 87(1): 86-91, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11848462

RESUMO

Thrombomodulin is an endothelial cell surface receptor for thrombin. It plays an important role in the regulation of blood coagulation by decreasing thrombin activity and activating protein C. This study examined the possible association between the thrombomodulin -33G/A polymorphism and acute myocardial infarction. We recruited 278 patients (mean age 57.5 years, 241 men) with documented myocardial infarction and 450 age- and sex-matched control subjects. Polymerase chain reaction and single-strand conformation polymorphism was used to define the thrombomodulin -33G/A polymorphism. The frequency of the thrombomodulin GA+AA genotype among patients with myocardial infarction was higher than that in control subjects (22.7% vs. 16.2%, odds ratio [OR] 1.5, 95% confidence interval [CI] 1.0 to 2.2). The -33G/A polymorphism (GA+AA genotype) was significantly associated with myocardial infarction (OR 1.6, 95% CI 1.1 to 2.5) as was hypertension, diabetes mellitus and smoking. Among young myocardial infarction patients (age < or =45 years, n = 72), the frequency of -33G/A polymorphism was more significantly higher than that in control subjects (29.2% vs. 16.2%, OR 2.1, 95% CI 1.2 to 3.8). The -33G/A polymorphism (OR 2.3, 95% CI 1.3 to 4.1) and smoking (OR 4.5, 95% CI 2.5 to 7.9) were the only independent risk factors for young myocardial infarction. Furthermore, among patients who did not smoke, the polymorphism was associated with a nonsignificant increase in the risk of young myocardial infarction (OR 1.9, 95% CI 0.6 to 5.6); whereas, in the presence of smoking, the increase was statistically significant (OR 2.3, 95% CI 1.2 to 4.7). Smoking carriers of the thrombomodulin -33G/A polymorphism had a nearly 10-fold increased risk of young myocardial infarction (OR 9.8, 95% CI 4.3 to 22.4) when compared with nonsmoking non-carriers. We concluded that there was a significant association between the thrombomodulin -33G/A polymorphism and myocardial infarction in our population, especially in young patients. The clinical effect of this genetic factor was enhanced by smoking.


Assuntos
Infarto do Miocárdio/epidemiologia , Regiões Promotoras Genéticas/genética , Fumar/efeitos adversos , Trombomodulina/genética , Adulto , Estudos de Casos e Controles , Comorbidade , Estenose Coronária/epidemiologia , Diabetes Mellitus/epidemiologia , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Hipercolesterolemia/epidemiologia , Hipertensão/epidemiologia , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/sangue , Infarto do Miocárdio/genética , Polimorfismo Genético , Polimorfismo Conformacional de Fita Simples , Fatores de Risco , Trombomodulina/deficiência , Trombomodulina/fisiologia
17.
J Clin Invest ; 108(4): 537-46, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11518727

RESUMO

The thrombomodulin (TM) gene was ablated in mice in a cell type-restricted manner from vascular endothelium by Cre-recombinase-mediated excision controlled by the endothelial cell lineage-specific Tie2 promoter. Forty percent of mutant (TMLox-) mice display a distinct lethal embryonic phenotype not observed in completely TM-deficient embryos. The remaining 60% of TMLox mice survive beyond birth, but invariably succumb to a severe hypercoagulable state and massive thrombosis after 3 weeks, terminating in a lethal consumptive coagulopathy. The progression of thrombosis was age- and sex-dependent. Disruption of the TM/protein C pathway was not associated with a latent proinflammatory state. Disease onset and progression could be prevented by warfarin anticoagulation. These results show that in mice, loss of endothelial cell TM function causes spontaneous and fatal thrombosis in the arterial and venous circulation, resulting from unfettered activation of the coagulation system. The combination of complete disease penetrance, uniform disease onset at young age, large vessel thrombosis of the extremities and multiple organ systems, and consumptive coagulopathy as the disease end-point provides a unique mouse model of human thrombotic disease.


Assuntos
Coagulação Sanguínea/fisiologia , Coagulação Intravascular Disseminada/etiologia , Endotélio Vascular/metabolismo , Proteína C/fisiologia , Trombomodulina/deficiência , Trombose/etiologia , Fatores Etários , Animais , Anticoagulantes/uso terapêutico , Cardiomegalia/etiologia , Modelos Animais de Doenças , Progressão da Doença , Coagulação Intravascular Disseminada/tratamento farmacológico , Feminino , Regulação da Expressão Gênica , Marcação de Genes , Genes Letais , Genes Sintéticos , Humanos , Integrases/genética , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/patologia , Especificidade de Órgãos , Receptores Proteína Tirosina Quinases/genética , Receptor TIE-2 , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Recombinação Genética , Maturidade Sexual , Trombomodulina/genética , Trombomodulina/fisiologia , Trombofilia/tratamento farmacológico , Trombofilia/etiologia , Trombose/prevenção & controle , Proteínas Virais/genética , Varfarina/uso terapêutico
18.
Development ; 128(6): 827-38, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11222138

RESUMO

The endothelial cell surface receptor thrombomodulin (TM) inhibits blood coagulation by forming a complex with thrombin, which then converts protein C into the natural anticoagulant, activated protein C. In mice, a loss of TM function causes embryonic lethality at day 8.5 p.c. (post coitum) before establishment of a functional cardiovascular system. At this developmental stage, TM is expressed in the developing vasculature of the embryo proper, as well as in non-endothelial cells of the early placenta, giant trophoblast and parietal endoderm. Here, we show that reconstitution of TM expression in extraembryonic tissue by aggregation of tetraploid wild-type embryos with TM-null embryonic stem cells rescues TM-null embryos from early lethality. TM-null tetraploid embryos develop normally during midgestation, but encounter a secondary developmental block between days 12.5 and 16.5 p.c. Embryos lacking TM develop lethal consumptive coagulopathy during this period, and no live embryos are retrieved at term. Morphogenesis of embryonic blood vessels and other organs appears normal before E15. These findings demonstrate a dual role of TM in development, and that a loss of TM function disrupts mouse embryogenesis at two different stages. These two functions of TM are exerted in two distinct tissues: expression of TM in non-endothelial extraembryonic tissues is required for proper function of the early placenta, while the absence of TM from embryonic blood vessel endothelium causes lethal consumptive coagulopathy.


Assuntos
Vasos Sanguíneos/embriologia , Desenvolvimento Embrionário e Fetal/fisiologia , Placenta/fisiologia , Trombomodulina/fisiologia , Animais , Desenvolvimento Embrionário e Fetal/genética , Endoderma/fisiologia , Feminino , Morte Fetal/genética , Fibrinogênio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Pulmão/embriologia , Camundongos , Camundongos Knockout , Morfogênese , Gravidez , Circulação Pulmonar/fisiologia , Trombomodulina/deficiência , Trombomodulina/genética , Trofoblastos/fisiologia
19.
Acta Neuropathol ; 100(6): 718-22, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11078226

RESUMO

We found a microvascular endothelial abnormality in a biopsy specimen from the gastrocnemius muscle of a patient with gastric cancer, who had severe myalgia and angialgia in the calf region with the symptoms of thrombophlebitis. There were no definite findings of inflammatory myopathy in histochemical and immunohistochemical studies. Electron microscopic examination revealed the accumulation of abnormal mitochondria in the subsarcolemmal area, and a fair number of degenerating capillaries. Immunohistochemical analysis of procoagulant or anticoagulant factors revealed marked reduction of thrombomodulin (TM) expression on small vessels and capillaries. Although a reduction of TM on small vessels has been observed around perifascicular atrophic fibers in patients with dermatomyositis, histochemical findings of the present patient showed no perifascicular atrophy or severely degenerating fibers. These pathological findings in the patient may be related to a malignant neoplasm and may be one of the causes of disseminated intravascular coagulation (DIC), which is the main complication of malignant neoplasms. Further studies are necessary to determine whether the reduction of TM on the small vessels and capillaries in skeletal muscle is a predictor of some severe condition such as DIC or a rare pathological finding in some special condition such as scirrhous carcinoma with thrombophlebitis.


Assuntos
Endotélio Vascular/patologia , Músculo Esquelético/patologia , Dor/etiologia , Síndromes Paraneoplásicas/patologia , Neoplasias Gástricas/complicações , Trombomodulina/deficiência , Biópsia , Causalidade , Progressão da Doença , Coagulação Intravascular Disseminada/etiologia , Coagulação Intravascular Disseminada/patologia , Coagulação Intravascular Disseminada/fisiopatologia , Endotélio Vascular/metabolismo , Endotélio Vascular/ultraestrutura , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Dor/patologia , Dor/fisiopatologia , Síndromes Paraneoplásicas/fisiopatologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/fisiopatologia , Tromboflebite/etiologia , Tromboflebite/patologia , Tromboflebite/fisiopatologia
20.
Haemostasis ; 30 Suppl 2: 1-10, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11251335

RESUMO

Venous thromboembolism is a multifactorial disease that depends on variable combinations of acquired and genetic risk factors. The genetic risk factors include loss-of-function mutations in the genes that encode proteins with clot-restraining function, and gain-of-function mutations in procoagulant factors. The loss-of-function mutations are heterogeneous and comprise any mutation that impairs gene function. On the whole, these mutations are rare, with fewer than 1/200--500 individuals affected. This low prevalence in the population is probably caused by the loss of mutant alleles from the gene pool through critically ill homozygous subjects. The gain-of-function mutations in procoagulant proteins differ from the loss-of-function mutations in at least three important respects: these are more homogeneous (factor V Leiden and PT 20210), homozygous individuals are relatively mildly affected, and these are relatively prevalent (3--15%) in Caucasian populations.


Assuntos
Trombofilia/genética , Regiões 3' não Traduzidas/genética , Resistência à Proteína C Ativada/genética , Antitrombinas/deficiência , Antitrombinas/genética , Fatores de Coagulação Sanguínea/genética , Fator V/genética , Fator XIII/genética , Deficiência do Fator XIII/genética , Frequência do Gene , Heterogeneidade Genética , Predisposição Genética para Doença , Humanos , Mutação , Mutação de Sentido Incorreto , Prevalência , Proteína C/genética , Deficiência de Proteína C/genética , Proteína S/genética , Deficiência de Proteína S/genética , Protrombina/genética , Trombomodulina/deficiência , Trombofilia/classificação , Trombofilia/epidemiologia , População Branca/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA