Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Transl Res ; 232: 150-162, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33737161

RÉSUMÉ

Deleterious hyper-inflammation resulting from macrophage activation may aggravate sepsis and lead to lethality. Tumor endothelial marker 1 (TEM1), a type I transmembrane glycoprotein containing six functional domains, has been implicated in cancer and chronic sterile inflammatory disorders. However, the role of TEM1 in acute sepsis remains to be determined. Herein we explored the functional significance of the TEM1 lectin-like domain (TEM1D1) in monocyte/macrophage activation and sepsis using TEM1D1-deleted (TEM1LeD/LeD) transgenic mice and recombinant TEM1D1 (rTEM1D1) protein. Under stimulation with lipopolysaccharides (LPS) or several other toll-like receptor agonists, TEM1LeD/LeD macrophages produced lower levels of tumor necrosis factor (TNF)-α and interleukin (IL)-6 than wild-type TEM1wt/wt macrophages. Compared with TEM1wt/wt macrophages, LPS-macrophage binding and intracellular mitogen-activated protein kinase (MAPK)/nuclear factor (NF)-κB activation were suppressed in TEM1LeD/LeD macrophages. In vivo, TEM1D1 deletion improved survival in LPS-challenged mice with reduction of circulating TNF-α and IL-6 and alleviation of lung injury and pulmonary leukocyte accumulation. In contrast, rTEM1D1 could bind to LPS and markedly suppress LPS-macrophage binding, MAPK/NF-κB signaling in macrophages and proinflammatory cytokine production. Treatment with rTEM1D1 improved survival and attenuated circulating TNF-α and IL-6, lung injury and pulmonary accumulation of leukocytes in LPS-challenged mice. These findings demonstrated differential roles for the TEM1 lectin-like domain in macrophages and soluble TEM1 lectin-like domain in sepsis. TEM1 in macrophages mediates LPS-induced inflammation via its lectin-like domain, whereas rTEM1D1 interferes with LPS-induced macrophage activation and sepsis.


Sujet(s)
Antigènes CD/physiologie , Lectines/composition chimique , Lipopolysaccharides/pharmacologie , Activation des macrophages/effets des médicaments et des substances chimiques , Protéines tumorales/physiologie , Sepsie/étiologie , Animaux , Antigènes CD/composition chimique , Antigènes CD/génétique , Antigènes néoplasiques/génétique , Délétion de gène , Humains , Inflammation/induit chimiquement , Lipopolysaccharides/métabolisme , Activation des macrophages/physiologie , Macrophages/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Protéines tumorales/composition chimique , Protéines recombinantes/génétique , Sepsie/physiopathologie
2.
J Invest Dermatol ; 139(10): 2204-2214.e7, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-30986375

RÉSUMÉ

Tumor endothelial marker 1 (TEM1), also known as endosialin or CD248, is a type I transmembrane glycoprotein containing a C-type lectin-like domain. It is highly expressed in pericytes and fibroblasts. Dermal fibroblasts play a pivotal role during cutaneous wound healing, especially in the proliferative phase. However, the physiological function of TEM1 in wound healing is still undetermined. During the process of wound healing, the expression of both TEM1 and platelet-derived growth factor (PDGF) receptor α was highly upregulated in myofibroblasts. In vivo, fibroblast activation and collagen deposition in granulation tissues were attenuated, and wound healing was retarded in TEM1-deleted mice. In vitro, the migration, adhesion, and proliferation of NIH3T3 cells were suppressed following TEM1 knockdown by short hairpin RNA. In PDGF-BB-treated NIH3T3 cells, the downstream signal and mitogenic, and chemoattractive effects were inhibited by TEM1 knockdown. In addition, TEM1 and PDGF receptor α were colocalized in subcellular organelles in fibroblasts, and the association of TEM1 and PDGF receptor α was demonstrated by coimmunoprecipitation. In summary, these findings suggested that TEM1, in combination with PDGF receptor α, plays a critical role in wound healing by enhancing the mitogenic and chemoattractive effects of PDGF-BB and collagen deposition in myofibroblasts.


Sujet(s)
Antigènes CD/génétique , Régulation de l'expression des gènes , Protéines tumorales/génétique , Récepteurs aux facteurs de croissance dérivés des plaquettes/génétique , Cicatrisation de plaie/génétique , Plaies et blessures/anatomopathologie , Animaux , Technique de Western/méthodes , Modèles animaux de maladie humaine , Humains , Souris , Souris transgéniques , Répartition aléatoire , Réaction de polymérisation en chaine en temps réel/méthodes , Facteurs temps , Résultat thérapeutique , Régulation positive , Cicatrisation de plaie/physiologie , Plaies et blessures/métabolisme
3.
Immunol Cell Biol ; 95(4): 372-379, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-27808085

RÉSUMÉ

The leukocyte adhesion cascade involves multiple events that efficiently localize circulating leukocytes into the injured sites to mediate inflammatory responses. From rolling to firm adhesion, the interactions between adhesion molecules have pivotal roles in increasing the avidity of leukocytes to endothelial cells. Thrombomodulin (TM), an essential anticoagulant protein in the vasculature, is also expressed on leukocytes. We previously demonstrated that Lewisy (Ley), a specific ligand of TM, is upregulated in inflamed endothelium and is involved in leukocyte adhesion. The current study aimed to investigate whether leukocyte-expressed TM promotes cell adhesion by interacting with Ley. Using human monocytic THP-1 cells as an in vitro cell model, we showed that TM increases THP-1 cell adhesion to inflamed endothelium as well as to Ley-immobilized surface. When THP-1 adhered to activated endothelium and Ley-immobilized surface, the TM distribution became polarized. Addition of soluble Ley to a suspension of THP-1 cells with TM expression triggered an increase in the level of phosphorylated p38 mitogen-activated protein kinase (MAPK), which enabled THP-1 to adhere firmly to intercellular adhesion molecule (ICAM)-1 by activating ß2 integrins. In vivo, macrophage infiltration and neointima formation following arterial ligation-induced vascular injury were higher in wild-type TM (TMflox/flox) than in myeloid-specific TM-deficient (LysMcre/TMflox/flox) mice. Taken together, these results suggest a novel function for TM as an adhesion molecule in monocytes, where it enhances cell adhesion by binding Ley, leading to ß2 integrin activation via p38 MAPK.


Sujet(s)
Cellules endothéliales/immunologie , Inflammation/immunologie , Monocytes/immunologie , Néointima/immunologie , Thrombomoduline/métabolisme , Animaux , Antigènes CD18/métabolisme , Adhérence cellulaire , Modèles animaux de maladie humaine , Humains , Molécule-1 d'adhérence intercellulaire/métabolisme , /métabolisme , Ligands , Souris , Souris knockout , Petit ARN interférent/génétique , Transduction du signal , Cellules THP-1 , Thrombomoduline/agonistes , Thrombomoduline/génétique , Régulation positive , p38 Mitogen-Activated Protein Kinases/métabolisme
4.
Oncotarget ; 7(42): 68122-68139, 2016 Oct 18.
Article de Anglais | MEDLINE | ID: mdl-27602495

RÉSUMÉ

Angiogenesis promotes tumor growth and metastasis. Cell adhesion molecules interact with the extracellular matrix (ECM) and increase cell adhesion and migration during angiogenesis. Thrombomodulin (TM) is a cell surface transmembrane glycoprotein expressed in endothelial cells. However, the function and significance of TM in cell-matrix interactions and angiogenesis remain unclear. Here, we first demonstrated that recombinant lectin-like domain of TM interacts with an ECM protein, fibronectin, and identified the N-terminal 70-kDa domain of fibronectin as the TM-binding site. Exogenous expression of TM in TM-deficient A2058 melanoma cells enhanced cell adhesion and migration on fibronectin and invasion on Matrigel. In addition, TM increased focal adhesion kinase (FAK) phosphorylation and matrix metalloproteinase-9 production. In mice bearing subcutaneous B16F10 melanoma tumors, immunofluorescence analysis indicated that TM was highly expressed and co-localized with fibronectin on the tumor vasculature. The interaction between TM and fibronectin in tumor blood vessels was also validated by the proximity ligation assay. In human umbilical vein endothelial cells, up-regulation of TM by vascular endothelial growth factor (VEGF), a tumor angiogenic factor, promoted cell adhesion and tube formation, whereas TM knockdown by RNA interference attenuated VEGF-induced cell adhesion and tube formation. In summary, TM promotes angiogenesis by enhancing cell adhesion, migration, and FAK activation through interaction with fibronectin. TM may represent a novel target for inhibiting tumor angiogenesis.


Sujet(s)
Fibronectines/métabolisme , Focal adhesion protein-tyrosine kinases/métabolisme , Mélanome expérimental/métabolisme , Thrombomoduline/métabolisme , Animaux , Adhérence cellulaire/effets des médicaments et des substances chimiques , Adhérence cellulaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/génétique , Cellules cultivées , Activation enzymatique , Humains , Mélanome expérimental/génétique , Mélanome expérimental/anatomopathologie , Souris de lignée C57BL , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Néovascularisation physiologique/génétique , Liaison aux protéines , Interférence par ARN , Thrombomoduline/génétique , Facteur de croissance endothéliale vasculaire de type A/pharmacologie
5.
PLoS One ; 11(1): e0146565, 2016.
Article de Anglais | MEDLINE | ID: mdl-26741694

RÉSUMÉ

Toll-like receptor (TLR) family plays a key role in innate immunity and various inflammatory responses. TLR4, one of the well-characterized pattern-recognition receptors, can be activated by endogenous damage-associated molecular pattern molecules such as high mobility group box 1 (HMGB1) to sustain sterile inflammation. Evidence suggested that blockade of TLR4 signaling may confer protection against abdominal aortic aneurysm (AAA). Herein we aimed to obtain further insight into the mechanism by which TLR4 might promote aneurysm formation. Characterization of the CaCl2-induced AAA model in mice revealed that upregulation of TLR4 expression, localized predominantly to vascular smooth muscle cells (VSMCs), was followed by a late decline during a 28-day period of AAA development. In vitro, TLR4 expression was increased in VSMCs treated with HMGB1. Knockdown of TLR4 by siRNA attenuated HMGB1-enhanced production of proinflammatory cytokines, specifically interleukin-6 and monocyte chemoattractant protein-1 (MCP-1), and matrix-degrading matrix metalloproteinase (MMP)-2 from VSMCs. In vivo, two different strains of TLR4-deficient (C57BL/10ScNJ and C3H/HeJ) mice were resistant to CaCl2-induced AAA formation compared to their respective controls (C57BL/10ScSnJ and C3H/HeN). Knockout of TLR4 reduced interleukin-6 and MCP-1 levels and HMGB1 expression, attenuated macrophage accumulation, and eventually suppressed MMP production, elastin destruction and VSMC loss. Finally, human AAA exhibited higher TLR4 expression that was localized to VSMCs. These data suggest that TLR4 signaling contributes to AAA formation by promoting a proinflammatory status of VSMCs and by inducing proteinase release from VSMCs during aneurysm initiation and development.


Sujet(s)
Anévrysme de l'aorte abdominale/métabolisme , Récepteur de type Toll-4/physiologie , Animaux , Anévrysme de l'aorte abdominale/induit chimiquement , Chlorure de calcium , Études cas-témoins , Cellules cultivées , Cytokines/biosynthèse , Protéine HMGB1/métabolisme , Humains , Mâle , Matrix metalloproteinases/métabolisme , Souris de lignée C3H , Souris de lignée C57BL , Muscles lisses vasculaires/enzymologie , Myocytes du muscle lisse/enzymologie , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Régulation positive
6.
Arterioscler Thromb Vasc Biol ; 35(11): 2412-22, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26338301

RÉSUMÉ

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.


Sujet(s)
Aorte abdominale/métabolisme , Anévrysme de l'aorte abdominale/métabolisme , Aortite/métabolisme , Membrane cellulaire/métabolisme , Macrophages péritonéaux/métabolisme , Thrombomoduline/métabolisme , Angiotensine-II , Animaux , Aorte abdominale/immunologie , Anévrysme de l'aorte abdominale/induit chimiquement , Anévrysme de l'aorte abdominale/génétique , Anévrysme de l'aorte abdominale/immunologie , Aortite/induit chimiquement , Aortite/génétique , Aortite/immunologie , Chlorure de calcium , Membrane cellulaire/immunologie , Cellules cultivées , Chimiotaxie , Modèles animaux de maladie humaine , Élastine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/immunologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Humains , Médiateurs de l'inflammation/métabolisme , Macrophages péritonéaux/immunologie , Matrix metalloproteinase 9/métabolisme , Souris de lignée C57BL , Souris knockout , Muscles lisses vasculaires/immunologie , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/immunologie , Myocytes du muscle lisse/métabolisme , Stress oxydatif , Interférence par ARN , Études rétrospectives , Transduction du signal , Thrombomoduline/déficit , Thrombomoduline/génétique , Facteurs temps , Transfection
7.
J Immunol ; 194(4): 1905-15, 2015 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-25609841

RÉSUMÉ

CD14, a multiligand pattern-recognition receptor, is involved in the activation of many TLRs. Thrombomodulin (TM), a type I transmembrane glycoprotein, originally was identified as an anticoagulant factor that activates protein C. Previously, we showed that the recombinant TM lectin-like domain binds to LPS and inhibits LPS-induced inflammation, but the function of the recombinant epidermal growth factor-like domain plus serine/threonine-rich domain of TM (rTMD23) in LPS-induced inflammation remains unknown. In the current study, we found that rTMD23 markedly suppressed the activation of intracellular signaling pathways and the production of inflammatory cytokines induced by LPS. The anti-inflammatory activity of rTMD23 was independent of activated protein C. We also found that rTMD23 interacted with the soluble and membrane forms of CD14 and inhibited the CD14-mediated inflammatory response. Knockdown of CD14 in macrophages suppressed the production of inflammatory cytokines induced by LPS, and rTMD23 inhibited LPS-induced IL-6 production in CD14-knockdown macrophages. rTMD23 suppressed the binding of LPS to macrophages by blocking the association between monocytic membrane-bound TM and CD14. The administration of rTMD23 in mice, both pretreatment and posttreatment, significantly increased the survival rate and reduced the inflammatory response to LPS. Notably, the serine/threonine-rich domain is essential for the anti-inflammatory activity of rTMD23. To summarize, we show that rTMD23 suppresses the LPS-induced inflammatory response in mice by targeting CD14 and that the serine/threonine-rich domain is crucial for the inhibitory effect of rTMD23 on LPS-induced inflammation.


Sujet(s)
Inflammation/immunologie , Antigènes CD14/immunologie , Macrophages/immunologie , Transduction du signal/immunologie , Thrombomoduline/immunologie , Animaux , Modèles animaux de maladie humaine , Cellules endothéliales/immunologie , Cytométrie en flux , Technique d'immunofluorescence , Humains , Immunoprécipitation , Lipopolysaccharides/immunologie , Souris , Souris de lignée C57BL , Structure tertiaire des protéines , Protéines recombinantes/immunologie , Résonance plasmonique de surface , Veines ombilicales
8.
Ann Surg ; 258(6): 1103-10, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-23295319

RÉSUMÉ

OBJECTIVE: To investigate whether recombinant thrombomodulin containing all the extracellular domains (rTMD123) has therapeutic potential against aneurysm development. SUMMARY BACKGROUND DATA: The pathogenesis of abdominal aortic aneurysm (AAA) is characterized by chronic inflammation and proteolytic degradation of extracellular matrix. Thrombomodulin, a transmembrane glycoprotein, exerts anti-inflammatory activities such as inhibition of cytokine production and sequestration of proinflammatory high-mobility group box 1 (HMGB1) to prevent it from engaging the receptor for advanced glycation end product (RAGE) that may sustain inflammation and tissue damage. METHODS: The in vivo effects of treatment and posttreatment with rTMD123 on aortic dilatation were measured using the CaCl2-induced AAA model in mice. RESULTS: Characterization of the CaCl2-induced model revealed that HMGB1 and RAGE, both localized mainly to macrophages, were persistently upregulated during a 28-day period of AAA development. In vitro, rTMD123-HMGB1 interaction prevented HMGB1 binding to macrophages, thereby prohibiting activation of HMGB1-RAGE signaling in macrophages. In vivo, short-term treatment with rTMD123 upon AAA induction suppressed the levels of proinflammatory cytokines, HMGB1, and RAGE in the aortic tissue; reduced the infiltrating macrophage number; and finally attenuated matrix metalloproteinase production, extracellular matrix destruction, and AAA formation without disturbing vascular calcification. Consistently, posttreatment with rTMD123 seven days after AAA induction alleviated vascular inflammation and retarded AAA progression. CONCLUSIONS: These data suggest that rTMD123 confers protection against AAA development. The mechanism of action may be associated with reduction of proinflammatory mediators, blockade of macrophage recruitment, and suppression of HMGB1-RAGE signaling involved in aneurysm formation and downstream macrophage activation.


Sujet(s)
Anévrysme de l'aorte abdominale/prévention et contrôle , Thrombomoduline/usage thérapeutique , Animaux , Anévrysme de l'aorte abdominale/induit chimiquement , Chlorure de calcium/administration et posologie , Souris , Souris de lignée C57BL , Récepteur spécifique des produits finaux de glycosylation avancée , Récepteurs immunologiques/physiologie , Protéines recombinantes/usage thérapeutique
9.
J Immunol ; 188(12): 6328-37, 2012 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-22573811

RÉSUMÉ

Sepsis results from the host hyperinflammatory response to bacterial infection, causing multiple organ failure and high mortality. We previously demonstrated that LPS binds to monocytic membrane-bound thrombomodulin (TM), but the role of monocytic TM in LPS-induced inflammation remains unknown. In this study, we demonstrated that TM knockdown in human monocytic cells attenuated LPS-induced signaling pathways and cytokine production. Coimmunoprecipitation and immunofluorescence assays showed that monocytic TM interacted with the LPS receptors, CD14 and TLR4/myeloid differentiation factor-2 (MD-2) complex, indicating that it binds to LPS and triggers an LPS-induced inflammatory response by interacting with the CD14/TLR4/MD-2 complex. We also found that monocytic TM knockdown reduced cytokine production induced by gram-negative bacteria Klebsiella pneumoniae, suggesting that monocytic TM plays an important role in gram-negative bacteria-induced inflammation. To further investigate the function of monocytic TM in vivo, myeloid-specific TM-deficient mice were established and were found to display improved survival that resulted from the attenuation of septic syndrome, including reduced systemic inflammatory response and resistance to bacterial dissemination, after K. pneumoniae infection or cecal ligation and puncture surgery. The inhibition of bacterial dissemination in mice with a deficiency of myeloid TM may be caused by the early increase in neutrophil infiltration. Therefore, we conclude that monocytic TM is a novel component in the CD14/TLR4/MD-2 complex and participates in the LPS- and gram-negative bacteria-induced inflammatory response.


Sujet(s)
Inflammation/immunologie , Monocytes/immunologie , Sepsie/immunologie , Thrombomoduline/immunologie , Animaux , Lignée cellulaire , Cytokines/biosynthèse , Cytométrie en flux , Technique d'immunofluorescence , Techniques de knock-down de gènes , Infections bactériennes à Gram négatif/immunologie , Humains , Immunoprécipitation , Inflammation/métabolisme , Lipopolysaccharides/immunologie , Souris , Monocytes/métabolisme , RT-PCR , Sepsie/métabolisme , Thrombomoduline/métabolisme
10.
FASEB J ; 26(8): 3440-52, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22593542

RÉSUMÉ

Adhesive interactions between cells are needed to maintain tissue architecture during development, tissue renewal and wound healing. Thrombomodulin (TM) is an integral membrane protein that participates in cell-cell adhesion through its extracellular lectin-like domain. However, the molecular basis of TM-mediated cell-cell adhesion is poorly understood. Here, we demonstrate that TM is linked to the actin cytoskeleton via ezrin. In vitro binding assays showed that the TM cytoplasmic domain bound directly to the N-terminal domain of ezrin. Mutational analysis of the TM cytoplasmic domain identified (522)RKK(524) as important ezrin-binding residues. In epidermal epithelial A431 cells, TM colocalized with ezrin and actin filaments at cell-cell contacts. Knockdown of endogenous TM expression by RNA interference induced morphological changes and accelerated cell migration in A431 cells. Moreover, epidermal growth factor, upstream of ezrin activation, stimulated the interaction between ezrin and TM. In skin wound healing of mice, TM and ezrin were highly expressed in neoepidermis, implying that both proteins are key molecules in reepithelialization that requires collective cell migration of epithelial cells. Finally, exogenous expression of TM in TM-deficient melanoma A2058 cells promoted collective cell migration. In summary, TM, which associates with ezrin and actin filaments, maintains epithelial morphology and promotes collective cell migration.


Sujet(s)
Mouvement cellulaire , Protéines du cytosquelette/métabolisme , Thrombomoduline/physiologie , Actines/métabolisme , Animaux , Sites de fixation , Adhérence cellulaire , Lignée cellulaire , Cellules épidermiques , Facteur de croissance épidermique , Humains , Souris , Structure tertiaire des protéines , Thrombomoduline/génétique , Cicatrisation de plaie/physiologie
11.
Blood ; 112(9): 3661-70, 2008 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-18711002

RÉSUMÉ

Thrombomodulin (TM), a widely expressing glycoprotein originally identified in vascular endothelium, is an important cofactor in the protein C anticoagulant system. TM appears to exhibit anti-inflammatory ability through both protein C-dependent and -independent pathways. We presently have demonstrated that recombinant N-terminal lectinlike domain of TM (rTMD1) functions as a protective agent against sepsis caused by Gram-negative bacterial infections. rTMD1 caused agglutination of Escherichia coli and Klebsiella pneumoniae and enhanced the macrophage phagocytosis of these Gram-negative bacteria. Moreover, rTMD1 bound to the Klebsiella pneumoniae and lipopolysaccharide (LPS) by specifically interacting with Lewis Y antigen. rTMD1 inhibited LPS-induced inflammatory mediator production via interference with CD14 and LPS binding. Furthermore, rTMD1 modulated LPS-induced mitogen-activated protein kinase and nuclear factor-kappaB signaling pathway activations and inducible nitric oxide synthase expression in macrophages. Administration of rTMD1 protected the host by suppressing inflammatory responses induced by LPS and Gram-negative bacteria, and enhanced LPS and bacterial clearance in sepsis. Thus, rTMD1 can be used to defend against bacterial infection and inhibit LPS-induced inflammatory responses, suggesting that rTMD1 may be valuable in the treatment of severe inflammation in sepsis, especially in Gram-negative bacterial infections.


Sujet(s)
/métabolisme , Thrombomoduline/composition chimique , Thrombomoduline/métabolisme , Animaux , Sites de fixation , Lignée cellulaire , Infections bactériennes à Gram négatif/traitement médicamenteux , Humains , Inflammation/traitement médicamenteux , Inflammation/prévention et contrôle , Klebsiella pneumoniae/métabolisme , Klebsiella pneumoniae/pathogénicité , Ligands , Lipopolysaccharides/métabolisme , Lipopolysaccharides/toxicité , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Mâle , Souris , Facteur de transcription NF-kappa B/métabolisme , Structure tertiaire des protéines , Protéines recombinantes/composition chimique , Protéines recombinantes/métabolisme , Protéines recombinantes/pharmacologie , Sepsie/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Thrombomoduline/administration et posologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE