Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood Adv ; 3(2): 168-183, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30670533

RESUMO

In the earliest phase of inflammation, histamine and other agonists rapidly mobilize P-selectin to the apical membranes of endothelial cells, where it initiates rolling adhesion of flowing neutrophils. Clustering of P-selectin in clathrin-coated pits facilitates rolling. Inflammatory cytokines typically signal by regulating gene transcription over a period of hours. We found that neutrophils rolling on P-selectin secreted the cytokine oncostatin M (OSM). The released OSM triggered signals through glycoprotein 130 (gp130)-containing receptors on endothelial cells that, within minutes, further clustered P-selectin and markedly enhanced its adhesive function. Antibodies to OSM or gp130, deletion of the gene encoding OSM in hematopoietic cells, or conditional deletion of the gene encoding gp130 in endothelial cells inhibited neutrophil rolling on P-selectin in trauma-stimulated venules of the mouse cremaster muscle. In a mouse model of P-selectin-dependent deep vein thrombosis, deletion of OSM in hematopoietic cells or of gp130 in endothelial cells markedly inhibited adhesion of neutrophils and monocytes and the rate and extent of thrombus formation. Our results reveal a paracrine-signaling mechanism by which neutrophil-released OSM rapidly influences endothelial cell function during physiological and pathological inflammation.


Assuntos
Endotélio Vascular/metabolismo , Neutrófilos/metabolismo , Oncostatina M/metabolismo , Selectina-P/metabolismo , Trombose/etiologia , Trombose/metabolismo , Vasculite/etiologia , Vasculite/metabolismo , Animais , Biomarcadores , Adesão Celular , Comunicação Celular , Vesículas Revestidas por Clatrina/metabolismo , Receptor gp130 de Citocina/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Células Endoteliais/metabolismo , Humanos , Imunofenotipagem , Migração e Rolagem de Leucócitos , Camundongos , Camundongos Knockout , Modelos Biológicos , Neutrófilos/imunologia , Selectina-P/genética , Ligação Proteica , Transdução de Sinais , Trombose/patologia , Vasculite/patologia
2.
JCI Insight ; 3(14)2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30046013

RESUMO

Site-1 protease (S1P), encoded by MBTPS1, is a serine protease in the Golgi. S1P regulates lipogenesis, endoplasmic reticulum (ER) function, and lysosome biogenesis in mice and in cultured cells. However, how S1P differentially regulates these diverse functions in humans has been unclear. In addition, no human disease with S1P deficiency has been identified. Here, we report a pediatric patient with an amorphic and a severely hypomorphic mutation in MBTPS1. The unique combination of these mutations results in a frequency of functional MBTPS1 transcripts of approximately 1%, a finding that is associated with skeletal dysplasia and elevated blood lysosomal enzymes. We found that the residually expressed S1P is sufficient for lipid homeostasis but not for ER and lysosomal functions, especially in chondrocytes. The defective S1P function specifically impairs activation of the ER stress transducer BBF2H7, leading to ER retention of collagen in chondrocytes. S1P deficiency also causes abnormal secretion of lysosomal enzymes due to partial impairment of mannose-6-phosphate-dependent delivery to lysosomes. Collectively, these abnormalities lead to apoptosis of chondrocytes and lysosomal enzyme-mediated degradation of the bone matrix. Correction of an MBTPS1 variant or reduction of ER stress mitigated collagen-trafficking defects. These results define a new congenital human skeletal disorder and, more importantly, reveal that S1P is particularly required for skeletal development in humans. Our findings may also lead to new therapies for other genetic skeletal diseases, as ER dysfunction is common in these disorders.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/metabolismo , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Transporte Proteico , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Apoptose , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Doenças do Desenvolvimento Ósseo/fisiopatologia , Técnicas de Cultura de Células , Pré-Escolar , Condrócitos/metabolismo , Colágeno/metabolismo , Retículo Endoplasmático/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Doenças Genéticas Inatas , Complexo de Golgi/metabolismo , Homeostase , Humanos , Lipogênese , Lisossomos/metabolismo , Manosefosfatos , Mutação
3.
Blood Adv ; 2(7): 731-744, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29592875

RESUMO

Rolling neutrophils receive signals while engaging P- and E-selectin and chemokines on inflamed endothelium. Selectin signaling activates ß2 integrins to slow rolling velocities. Chemokine signaling activates ß2 integrins to cause arrest. Despite extensive study, key aspects of these signaling cascades remain unresolved. Using complementary in vitro and in vivo assays, we found that selectin and chemokine signals in neutrophils triggered Rap1a-dependent and phosphatidylinositol-4-phosphate 5-kinase γ (PIP5Kγ90)-dependent pathways that induce integrin-dependent slow rolling and arrest. Interruption of both pathways, but not either pathway alone, blocked talin-1 recruitment to and activation of integrins. An isoform of PIP5Kγ90 lacking the talin-binding domain (PIP5Kγ87) could not activate integrins. Chemokines, but not selectins, used phosphatidylinositol-4,5-bisphosphate 3-kinase γ (PI3Kγ) in cooperation with Rap1a to mediate integrin-dependent slow rolling (at low chemokine concentrations), as well as arrest (at high chemokine concentrations). High levels of chemokines activated ß2 integrins without selectin signals. When chemokines were limiting, they synergized with selectins to activate ß2 integrins.


Assuntos
Antígenos CD18/metabolismo , Quimiocinas/fisiologia , Migração e Rolagem de Leucócitos , Neutrófilos/metabolismo , Selectinas/fisiologia , Animais , Quimiocinas/metabolismo , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Humanos , Camundongos , Neutrófilos/química , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Selectinas/metabolismo , Transdução de Sinais/fisiologia , Proteínas rap1 de Ligação ao GTP/metabolismo
4.
Proc Natl Acad Sci U S A ; 114(31): 8360-8365, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28716912

RESUMO

Most platelet membrane proteins are modified by mucin-type core 1-derived glycans (O-glycans). However, the biological importance of O-glycans in platelet clearance is unclear. Here, we generated mice with a hematopoietic cell-specific loss of O-glycans (HC C1galt1-/- ). These mice lack O-glycans on platelets and exhibit reduced peripheral platelet numbers. Platelets from HC C1galt1-/- mice show reduced levels of α-2,3-linked sialic acids and increased accumulation in the liver relative to wild-type platelets. The preferential accumulation of HC C1galt1-/- platelets in the liver was reduced in mice lacking the hepatic asialoglycoprotein receptor [Ashwell-Morell receptor (AMR)]. However, we found that Kupffer cells are the primary cells phagocytosing HC C1galt1-/- platelets in the liver. Our results demonstrate that hepatic AMR promotes preferential adherence to and phagocytosis of desialylated and/or HC C1galt1-/- platelets by the Kupffer cell through its C-type lectin receptor CLEC4F. These findings provide insights into an essential role for core 1 O-glycosylation of platelets in their clearance in the liver.


Assuntos
Plaquetas/metabolismo , Galactosiltransferases/genética , Células de Kupffer/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Polissacarídeos/metabolismo , Animais , Receptor de Asialoglicoproteína/metabolismo , Hepatócitos/metabolismo , Homeostase/fisiologia , Lectinas Tipo C/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Trombocitopenia/patologia
5.
Arterioscler Thromb Vasc Biol ; 36(6): 1114-21, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27102967

RESUMO

OBJECTIVE: During inflammation, P-selectin expressed on activated endothelial cells and platelets mediates rolling adhesion of leukocytes. Atherosclerosis-prone mice crossed with P-selectin-deficient (Selp(-/-)) mice develop smaller lesions. Cytokines, such as tumor necrosis factor-α, increase Selp transcripts and augment atherosclerosis in mice. However, they decrease SELP transcripts in humans, challenging assumptions that human P-selectin is atherogenic. We used mice expressing a human SELP transgene to examine the atherogenic role of P-selectin. APPROACH AND RESULTS: We crossed apolipoprotein E-deficient (Apoe(-/-)) mice with Selp(-/-) mice or transgenic mice expressing the entire human SELP gene (TgSELP(+/-)). Aortas developed larger, macrophage-rich atheromas in Apoe(-/-)Selp(-/-)TgSELP(+/-) mice than in Apoe(-/-)Selp(-/-) mice after 8 or 16 weeks on a Western diet. Confocal microscopy of Apoe(-/-)Selp(-/-)TgSELP(+/-) aortas revealed staining for human P-selectin in endothelial cells overlying atheromas but not in lesional macrophages. We also observed staining for human P-selectin in aortic endothelial cells of 3- to 4-week-old Apoe(-/-)Selp(-/-)TgSELP(+/-) weanlings before atheromas developed. Furthermore, human SELP transcripts were ≈3-fold higher in aortas of Apoe(-/-)Selp(+/-)TgSELP(+/-) weanlings than in Selp(+/-)TgSELP(+/-) weanlings, whereas murine Selp and Sele transcripts were equivalent in weanlings of both genotypes. Human SELP transcripts in aortas of Apoe(-/-)Selp(+/-)TgSELP(+/-) mice remained nearly constant during 16 weeks on a Western diet, whereas murine Selp and Sele transcripts progressively increased. Bone marrow transplantation in Apoe(-/-)Selp(-/-) and Apoe(-/-)Selp(-/-)TgSELP(+/-) mice demonstrated that both platelets and endothelial cells must express human P-selectin to promote atherogenesis. CONCLUSIONS: P-selectin expressed by human SELP is atherogenic in Apoe(-/-) mice, suggesting that P-selectin contributes to atherogenesis in humans.


Assuntos
Aorta/metabolismo , Doenças da Aorta/metabolismo , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Selectina-P/metabolismo , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Plaquetas/metabolismo , Transplante de Medula Óssea , Dieta Hiperlipídica , Modelos Animais de Doenças , Progressão da Doença , Células Endoteliais/metabolismo , Predisposição Genética para Doença , Humanos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Selectina-P/genética , Fenótipo , Placa Aterosclerótica , Fatores de Tempo , Trombose Venosa/genética , Trombose Venosa/metabolismo
7.
J Biol Chem ; 291(3): 1441-7, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26631722

RESUMO

In humans and mice, megakaryocytes/platelets and endothelial cells constitutively synthesize P-selectin and mobilize it to the plasma membrane to mediate leukocyte rolling during inflammation. TNF-α, interleukin 1ß, and LPS markedly increase P-selectin mRNA in mice but decrease P-selectin mRNA in humans. Transgenic mice bearing the entire human SELP gene recapitulate basal and inducible expression of human P-selectin and reveal human-specific differences in P-selectin function. Differences in the human SELP and murine Selp promoters account for divergent expression in vitro, but their significance in vivo is not known. Here we generated knockin mice that replace the 1.4-kb proximal Selp promoter with the corresponding SELP sequence (Selp(KI)). Selp(KI) (/) (KI) mice constitutively expressed more P-selectin on platelets and more P-selectin mRNA in tissues but only slightly increased P-selectin mRNA after injection of TNF-α or LPS. Consistent with higher basal expression, leukocytes rolled more slowly on P-selectin in trauma-stimulated venules of Selp(KI) (/) (KI) mice. However, TNF-α did not further reduce P-selectin-dependent rolling velocities. Blunted up-regulation of P-selectin mRNA during contact hypersensitivity reduced P-selectin-dependent inflammation in Selp(KI) (/-) mice. Higher basal P-selectin in Selp(KI) (/) (KI) mice compensated for this defect. Therefore, divergent sequences in a short promoter mediate most of the functionally significant differences in expression of human and murine P-selectin in vivo.


Assuntos
Regulação da Expressão Gênica , Selectina-P/metabolismo , Regiões Promotoras Genéticas , Animais , Sequência de Bases , Cruzamentos Genéticos , Dermatite de Contato/imunologia , Dermatite de Contato/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Migração e Rolagem de Leucócitos/imunologia , Lipopolissacarídeos/toxicidade , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos , Selectina-P/química , Selectina-P/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Especificidade da Espécie , Organismos Livres de Patógenos Específicos , Fator de Necrose Tumoral alfa/metabolismo , Vênulas/efeitos dos fármacos , Vênulas/imunologia
8.
J Clin Invest ; 125(12): 4349-64, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26571402

RESUMO

Tumor angiogenesis is critical for cancer progression. In multiple murine models, endothelium-specific epsin deficiency abrogates tumor progression by shifting the balance of VEGFR2 signaling toward uncontrolled tumor angiogenesis, resulting in dysfunctional tumor vasculature. Here, we designed a tumor endothelium-targeting chimeric peptide (UPI) for the purpose of inhibiting endogenous tumor endothelial epsins by competitively binding activated VEGFR2. We determined that the UPI peptide specifically targets tumor endothelial VEGFR2 through an unconventional binding mechanism that is driven by unique residues present only in the epsin ubiquitin-interacting motif (UIM) and the VEGFR2 kinase domain. In murine models of neoangiogenesis, UPI peptide increased VEGF-driven angiogenesis and neovascularization but spared quiescent vascular beds. Further, in tumor-bearing mice, UPI peptide markedly impaired functional tumor angiogenesis, tumor growth, and metastasis, resulting in a notable increase in survival. Coadministration of UPI peptide with cytotoxic chemotherapeutics further sustained tumor inhibition. Equipped with localized tumor endothelium-specific targeting, our UPI peptide provides potential for an effective and alternative cancer therapy.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/farmacologia , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Peptídeos/farmacologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Motivos de Aminoácidos , Animais , Camundongos , Camundongos Knockout , Metástase Neoplásica , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Peptídeos/genética , Peptídeos/metabolismo , Estrutura Terciária de Proteína , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
J Immunol ; 195(8): 3880-9, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26355151

RESUMO

A2A adenosine receptor (A2AAR) signaling negatively regulates inflammatory responses in many disease models, but the detailed mechanisms remain unclear. We used the selective A2AAR agonist, ATL313, to examine how A2AAR signaling affects human and murine neutrophil adhesion under flow. Treating neutrophils with ATL313 inhibited selectin-induced, ß2 integrin-dependent slow rolling and chemokine-induced, ß2 integrin-dependent arrest on ICAM-1. ATL313 inhibited selectin-induced ß2 integrin extension, which supports slow rolling, and chemokine-induced hybrid domain "swing-out," which supports arrest. Furthermore, ATL313 inhibited integrin outside-in signaling as revealed by reduced neutrophil superoxide production and spreading on immobilized anti-ß2 integrin Ab. ATL313 suppressed selectin-triggered activation of Src family kinases (SFKs) and p38 MAPK, chemokine-triggered activation of Ras-related protein 1, and ß2 integrin-triggered activation of SFKs and Vav cytoskeletal regulatory proteins. ATL313 activated protein kinase A and its substrate C-terminal Src kinase, an inhibitor of SFKs. Treating neutrophils with a protein kinase A inhibitor blocked the actions of ATL313. In vivo, ATL313-treated neutrophils rolled faster and arrested much less frequently in postcapillary venules of the murine cremaster muscle after TNF-α challenge. Furthermore, ATL313 markedly suppressed neutrophil migration into the peritoneum challenged with thioglycollate. ATL313 did not affect A2AAR-deficient neutrophils, confirming its specificity. Our findings provide new insights into the anti-inflammatory mechanisms of A2AAR signaling and the potential utility of A2AAR agonists in inflammatory diseases.


Assuntos
Migração e Rolagem de Leucócitos/imunologia , Neutrófilos/imunologia , Receptor A2A de Adenosina/imunologia , Transdução de Sinais/imunologia , Animais , Antígenos CD18/genética , Antígenos CD18/imunologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Humanos , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Neutrófilos/citologia , Piperidinas/farmacologia , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/imunologia , Receptor A2A de Adenosina/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Quinases da Família src/genética , Quinases da Família src/imunologia
10.
Proc Natl Acad Sci U S A ; 112(28): 8661-6, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26124096

RESUMO

Palmitoylated cysteines typically target transmembrane proteins to domains enriched in cholesterol and sphingolipids (lipid rafts). P-selectin glycoprotein ligand-1 (PSGL-1), CD43, and CD44 are O-glycosylated proteins on leukocytes that associate with lipid rafts. During inflammation, they transduce signals by engaging selectins as leukocytes roll in venules, and they move to the raft-enriched uropods of polarized cells upon chemokine stimulation. It is not known how these glycoproteins associate with lipid rafts or whether this association is required for signaling or for translocation to uropods. Here, we found that loss of core 1-derived O-glycans in murine C1galt1(-/-) neutrophils blocked raft targeting of PSGL-1, CD43, and CD44, but not of other glycosylated proteins, as measured by resistance to solubilization in nonionic detergent and by copatching with a raft-resident sphingolipid on intact cells. Neuraminidase removal of sialic acids from wild-type neutrophils also blocked raft targeting. C1galt1(-/-) neutrophils or neuraminidase-treated neutrophils failed to activate tyrosine kinases when plated on immobilized anti-PSGL-1 or anti-CD44 F(ab')2. Furthermore, C1galt1(-/-) neutrophils incubated with anti-PSGL-1 F(ab')2 did not generate microparticles. In marked contrast, PSGL-1, CD43, and CD44 moved normally to the uropods of chemokine-stimulated C1galt1(-/-) neutrophils. These data define a role for core 1-derived O-glycans and terminal sialic acids in targeting glycoprotein ligands for selectins to lipid rafts of leukocytes. Preassociation of these glycoproteins with rafts is required for signaling but not for movement to uropods.


Assuntos
Leucócitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Microdomínios da Membrana/metabolismo , Polissacarídeos/metabolismo , Animais , Receptores de Hialuronatos/metabolismo , Leucossialina/metabolismo , Ligantes , Camundongos
11.
Nat Commun ; 6: 6915, 2015 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25892652

RESUMO

Myeloid-related proteins (MRPs) 8 and 14 are cytosolic proteins secreted from myeloid cells as proinflammatory mediators. Currently, the functional role of circulating extracellular MRP8/14 is unclear. Our present study identifies extracellular MRP8/14 as an autocrine player in the leukocyte adhesion cascade. We show that E-selectin-PSGL-1 interaction during neutrophil rolling triggers Mrp8/14 secretion. Released MRP8/14 in turn activates a TLR4-mediated, Rap1-GTPase-dependent pathway of rapid ß2 integrin activation in neutrophils. This extracellular activation loop reduces leukocyte rolling velocity and stimulates adhesion. Thus, we identify Mrp8/14 and TLR4 as important modulators of the leukocyte recruitment cascade during inflammation in vivo.


Assuntos
Antígenos CD18/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Adesão Celular/fisiologia , Migração e Rolagem de Leucócitos/fisiologia , Neutrófilos/fisiologia , Animais , Antígenos CD18/genética , Calgranulina A/genética , Calgranulina B/genética , Regulação da Expressão Gênica , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Ligação Proteica
12.
Blood ; 122(23): 3832-42, 2013 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-24081661

RESUMO

Neutrophils emigrate from venules to sites of infection or injury in response to chemotactic gradients. How these gradients form is not well understood. Some IL-6 family cytokines stimulate endothelial cells to express adhesion molecules and chemokines that recruit leukocytes. Receptors for these cytokines share the signaling subunit gp130. We studied knockout mice lacking gp130 in endothelial cells. Unexpectedly, gp130-deficient endothelial cells constitutively expressed more CXCL1 in vivo and in vitro, and even more upon stimulation with tumor necrosis factor-α. Mobilization of this increased CXCL1 from intracellular stores to the venular surface triggered ß2 integrin-dependent arrest of neutrophils rolling on selectins but impaired intraluminal crawling and transendothelial migration. Superfusing CXCL1 over venules promoted neutrophil migration only after intravenously injecting mAb to CXCL1 to diminish its intravascular function or heparinase to release CXCL1 from endothelial proteoglycans. Remarkably, mice lacking gp130 in endothelial cells had impaired histamine-induced venular permeability, which was restored by injecting anti-P-selectin mAb to prevent neutrophil rolling and arrest. Thus, excessive CXCL1 expression in gp130-deficient endothelial cells augments neutrophil adhesion but hinders migration, most likely by disrupting chemotactic gradients. Our data define a role for endothelial cell gp130 in regulating integrin-dependent adhesion and de-adhesion of neutrophils during inflammation.


Assuntos
Quimiocina CXCL1/metabolismo , Receptor gp130 de Citocina/deficiência , Células Endoteliais/fisiologia , Neutrófilos/fisiologia , Animais , Permeabilidade Capilar/fisiologia , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Quimiocina CXCL1/genética , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/fisiologia , Inflamação/fisiopatologia , Migração e Rolagem de Leucócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Selectina-P/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Regulação para Cima , Vênulas/fisiologia
13.
Blood ; 118(26): 6743-51, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-22021370

RESUMO

Reversible interactions of glycoconjugates on leukocytes with P- and E-selectin on endothelial cells mediate tethering and rolling of leukocytes in inflamed vascular beds, the first step in their recruitment to sites of injury. Although selectin ligands on hematopoietic precursors have been identified, here we review evidence that PSGL-1, CD44, and ESL-1 on mature leukocytes are physiologic glycoprotein ligands for endothelial selectins. Each ligand has specialized adhesive functions during tethering and rolling. Furthermore, PSGL-1 and CD44 induce signals that activate the ß2 integrin LFA-1 and promote slow rolling, whereas ESL-1 induces signals that activate the ß2 integrin Mac-1 in adherent neutrophils. We also review evidence for glycolipids, CD43, L-selectin, and other glycoconjugates as potential physiologic ligands for endothelial selectins on neutrophils or lymphocytes. Although the physiologic characterization of these ligands has been obtained in mice, we also note reported similarities and differences with human selectin ligands.


Assuntos
Selectina E/imunologia , Receptores de Hialuronatos/imunologia , Leucócitos/imunologia , Glicoproteínas de Membrana/imunologia , Selectina-P/imunologia , Receptores de Fatores de Crescimento de Fibroblastos/imunologia , Sialoglicoproteínas/imunologia , Animais , Selectina E/metabolismo , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Glicoconjugados/imunologia , Glicoconjugados/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Migração e Rolagem de Leucócitos/imunologia , Leucócitos/metabolismo , Ligantes , Glicoproteínas de Membrana/metabolismo , Camundongos , Selectina-P/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Sialoglicoproteínas/metabolismo , Transdução de Sinais/imunologia , Estresse Mecânico
14.
Blood ; 118(15): 4015-23, 2011 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-21860019

RESUMO

Trousseau syndrome is classically defined as migratory, heparin-sensitive but warfarin-resistant microthrombi in patients with occult, mucinous adenocarcinomas. Injecting carcinoma mucins into mice generates platelet-rich microthrombi dependent on P- and L-selectin but not thrombin. Heparin prevents mucin binding to P- and L-selectin and mucin-induced microthrombi. This model of Trousseau syndrome explains resistance to warfarin, which inhibits fluid-phase coagulation but not selectins. Here we found that carcinoma mucins do not generate microthrombi in mice lacking P-selectin glycoprotein ligand-1 (PSGL-1), the leukocyte ligand for P- and L-selectin. Furthermore, mucins did not activate platelets in blood from PSGL-1-deficient mice. Mucins induced microthrombi in radiation chimeras lacking endothelial P-selectin but not in chimeras lacking platelet P-selectin. Mucins caused leukocytes to release cathepsin G, but only if platelets were present. Mucins failed to generate microthrombi in cathepsin G-deficient mice. Mucins did not activate platelets in blood from mice lacking cathepsin G or protease-activated receptor-4 (PAR4), indicating that cathepsin G activates platelets through PAR4. Using knockout mice and blocking antibodies, we found that mucin-triggered cathepsin G release requires L-selectin and PSGL-1 on neutrophils, P-selectin on platelets, and Src family kinases in both cell types. Thus, carcinoma mucins promote thrombosis through adhesion-dependent, bidirectional signaling in neutrophils and platelets.


Assuntos
Adenocarcinoma Mucinoso/metabolismo , Plaquetas/metabolismo , Mucinas/metabolismo , Proteínas de Neoplasias/metabolismo , Ativação de Neutrófilo , Neutrófilos/metabolismo , Ativação Plaquetária , Trombose/metabolismo , Adenocarcinoma Mucinoso/complicações , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Mucinoso/patologia , Animais , Anticorpos Antineoplásicos/farmacologia , Anticorpos Neutralizantes/farmacologia , Plaquetas/patologia , Catepsina G/genética , Catepsina G/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo , Modelos Animais de Doenças , Humanos , Selectina L/genética , Selectina L/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mucinas/antagonistas & inibidores , Mucinas/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Neutrófilos/patologia , Selectina-P/genética , Selectina-P/metabolismo , Receptores de Trombina/genética , Receptores de Trombina/metabolismo , Síndrome , Trombose/etiologia , Trombose/genética , Trombose/patologia
15.
J Exp Med ; 207(13): 2975-87, 2010 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-21149548

RESUMO

Leukocytes roll on P-selectin after its mobilization from secretory granules to the surfaces of platelets and endothelial cells. Tumor necrosis factor (TNF), IL-1ß, and lipopolysaccharide increase synthesis of P-selectin in murine but not in human endothelial cells. To explore the physiological significance of this difference in gene regulation, we made transgenic mice bearing the human Selp gene and crossed them with mice lacking murine P-selectin (Selp(-/-)). The transgenic mice constitutively expressed human P-selectin in platelets, endothelial cells, and macrophages. P-selectin mediated comparable neutrophil migration into the inflamed peritoneum of transgenic and wild-type (WT) mice. Leukocytes rolled similarly on human or murine P-selectin on activated murine platelets and in venules of the cremaster muscle subjected to trauma. However, TNF increased murine P-selectin in venules, slowing rolling and increasing adhesion, whereas it decreased human P-selectin, accelerating rolling and decreasing adhesion. Both P- and E-selectin mediated basal rolling in the skin of WT mice, but E-selectin dominated rolling in transgenic mice. During contact hypersensitivity, murine P-selectin messenger (m) RNA was up-regulated and P-selectin was essential for leukocyte recruitment. However, human P-selectin mRNA was down-regulated and P-selectin contributed much less to leukocyte recruitment. These findings reveal functionally significant differences in basal and inducible expression of human and murine P-selectin in vivo.


Assuntos
Plaquetas/metabolismo , Perfilação da Expressão Gênica , Macrófagos Peritoneais/metabolismo , Selectina-P/genética , Animais , Western Blotting , Células Cultivadas , Células Endoteliais/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Migração e Rolagem de Leucócitos , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Neutrófilos/metabolismo , Selectina-P/metabolismo , Peritonite/induzido quimicamente , Peritonite/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tioglicolatos , Fator de Necrose Tumoral alfa/farmacologia , Vênulas/metabolismo
16.
Proc Natl Acad Sci U S A ; 107(20): 9204-9, 2010 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-20439727

RESUMO

Neutrophils roll on E-selectin in inflamed venules through interactions with cell-surface glycoconjugates. The identification of physiologic E-selectin ligands on neutrophils has been elusive. Current evidence suggests that P-selectin glycoprotein ligand-1 (PSGL-1), E-selectin ligand-1 (ESL-1), and CD44 encompass all glycoprotein ligands for E-selectin; that ESL-1 and CD44 use N-glycans to bind to E-selectin; and that neutrophils lacking core 2 O-glycans have partially defective interactions with E-selectin. These data imply that N-glycans on ESL-1 and CD44 and O-glycans on PSGL-1 constitute all E-selectin ligands, with neither glycan subset having a dominant role. The enzyme T-synthase transfers Gal to GalNAcalpha1-Ser/Thr to form the core 1 structure Galbeta1-3GalNAcalpha1-Ser/Thr, a precursor for core 2 and extended core 1 O-glycans that might serve as selectin ligands. Here, using mice lacking T-synthase in endothelial and hematopoietic cells, we found that E-selectin bound to CD44 and ESL-1 in lysates of T-synthase-deficient neutrophils. However, the cells exhibited markedly impaired rolling on E-selectin in vitro and in vivo, failed to activate beta2 integrins while rolling, and did not emigrate into inflamed tissues. These defects were more severe than those of neutrophils lacking PSGL-1, CD44, and the mucin CD43. Our results demonstrate that core 1-derived O-glycans are essential E-selectin ligands; that some of these O-glycans are on protein(s) other than PSGL-1, CD44, and CD43; and that PSGL-1, CD44, and ESL-1 do not constitute all glycoprotein ligands for E-selectin.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Selectina E/metabolismo , Neutrófilos/metabolismo , Polissacarídeos/metabolismo , Animais , Citometria de Fluxo , Receptores de Hialuronatos/metabolismo , Ligantes , Glicoproteínas de Membrana/metabolismo , Camundongos , Neutrófilos/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sialoglicoproteínas/metabolismo
17.
Blood ; 116(3): 485-94, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20299514

RESUMO

In inflamed venules, neutrophils rolling on E-selectin induce integrin alpha(L)beta(2)-dependent slow rolling on intercellular adhesion molecule-1 by activating Src family kinases (SFKs), DAP12 and Fc receptor-gamma (FcRgamma), spleen tyrosine kinase (Syk), and p38. E-selectin signaling cooperates with chemokine signaling to recruit neutrophils into tissues. Previous studies identified P-selectin glycoprotein ligand-1 (PSGL-1) as the essential E-selectin ligand and Fgr as the only SFK that initiate signaling to slow rolling. In contrast, we found that E-selectin engagement of PSGL-1 or CD44 triggered slow rolling through a common, lipid raft-dependent pathway that used the SFKs Hck and Lyn as well as Fgr. We identified the Tec kinase Bruton tyrosine kinase as a key signaling intermediate between Syk and p38. E-selectin engagement of PSGL-1 was dependent on its cytoplasmic domain to activate SFKs and slow rolling. Although recruiting phosphoinositide-3-kinase to the PSGL-1 cytoplasmic domain was reported to activate integrins, E-selectin-mediated slow rolling did not require phosphoinositide-3-kinase. Studies in mice confirmed the physiologic significance of these events for neutrophil slow rolling and recruitment during inflammation. Thus, E-selectin triggers common signals through distinct neutrophil glycoproteins to induce alpha(L)beta(2)-dependent slow rolling.


Assuntos
Selectina E/fisiologia , Receptores de Hialuronatos/fisiologia , Migração e Rolagem de Leucócitos/fisiologia , Antígeno-1 Associado à Função Linfocitária/fisiologia , Glicoproteínas de Membrana/fisiologia , Tirosina Quinase da Agamaglobulinemia , Animais , Humanos , Receptores de Hialuronatos/genética , Técnicas In Vitro , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Microdomínios da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Neutrófilos/fisiologia , Selectina-P/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-hck/fisiologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Quinases da Família src/fisiologia
18.
Annu Rev Cell Dev Biol ; 26: 363-96, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19575676

RESUMO

Rolling adhesion on vascular surfaces is the first step in recruiting circulating leukocytes, hematopoietic progenitors, or platelets to specific organs or to sites of infection or injury. Rolling requires the rapid yet balanced formation and dissociation of adhesive bonds in the challenging environment of blood flow. This review explores how structurally distinct adhesion receptors interact through mechanically regulated kinetics with their ligands to meet these challenges. Remarkably, increasing force applied to adhesive bonds first prolongs their lifetimes (catch bonds) and then shortens their lifetimes (slip bonds). Catch bonds mediate the counterintuitive phenomenon of flow-enhanced rolling adhesion. Force-regulated disruptions of receptor interdomain or intradomain interactions remote from the ligand-binding surface generate catch bonds. Adhesion receptor dimerization, clustering in membrane domains, and interactions with the cytoskeleton modulate the forces applied to bonds. Both inside-out and outside-in cell signals regulate these processes.


Assuntos
Adesão Celular , Movimento Celular , Animais , Moléculas de Adesão Celular/metabolismo , Humanos , Leucócitos/citologia , Leucócitos/metabolismo , Selectinas/metabolismo
19.
J Immunol ; 180(9): 6288-96, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18424752

RESUMO

After an inflammatory stimulus, lymphocyte migration into draining lymph nodes increases dramatically to facilitate the encounter of naive T cells with Ag-loaded dendritic cells. In this study, we show that CD73 (ecto-5'-nucleotidase) plays an important role in regulating this process. CD73 produces adenosine from AMP and is expressed on high endothelial venules (HEV) and subsets of lymphocytes. Cd73(-/-) mice have normal sized lymphoid organs in the steady state, but approximately 1.5-fold larger draining lymph nodes and 2.5-fold increased rates of L-selectin-dependent lymphocyte migration from the blood through HEV compared with wild-type mice 24 h after LPS administration. Migration rates of cd73(+/+) and cd73(-/-) lymphocytes into lymph nodes of wild-type mice are equal, suggesting that it is CD73 on HEV that regulates lymphocyte migration into draining lymph nodes. The A(2B) receptor is a likely target of CD73-generated adenosine, because it is the only adenosine receptor expressed on the HEV-like cell line KOP2.16 and it is up-regulated by TNF-alpha. Furthermore, increased lymphocyte migration into draining lymph nodes of cd73(-/-) mice is largely normalized by pretreatment with the selective A(2B) receptor agonist BAY 60-6583. Adenosine receptor signaling to restrict lymphocyte migration across HEV may be an important mechanism to control the magnitude of an inflammatory response.


Assuntos
5'-Nucleotidase/imunologia , Adenosina/imunologia , Movimento Celular/imunologia , Endotélio Vascular/imunologia , Linfonodos/imunologia , Linfócitos T/imunologia , 5'-Nucleotidase/genética , 5'-Nucleotidase/metabolismo , Adenosina/genética , Adenosina/metabolismo , Agonistas do Receptor A2 de Adenosina , Monofosfato de Adenosina/genética , Monofosfato de Adenosina/imunologia , Monofosfato de Adenosina/metabolismo , Aminopiridinas/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Endotélio Vascular/enzimologia , Inflamação/enzimologia , Inflamação/genética , Inflamação/imunologia , Selectina L/imunologia , Selectina L/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Receptor A2B de Adenosina/imunologia , Receptor A2B de Adenosina/metabolismo , Linfócitos T/enzimologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Regulação para Cima/imunologia , Vênulas/enzimologia , Vênulas/imunologia
20.
J Immunol ; 180(5): 3091-102, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18292532

RESUMO

Galectin-1 (Gal-1) and galectin-3 (Gal-3) exhibit profound but unique immunomodulatory activities in animals but their molecular mechanisms are incompletely understood. Early studies suggested that Gal-1 inhibits leukocyte function by inducing apoptotic cell death and removal, but recent studies show that some galectins induce exposure of the common death signal phosphatidylserine (PS) independently of apoptosis. In this study, we report that Gal-3, but not Gal-1, induces both PS exposure and apoptosis in primary activated human T cells, whereas both Gal-1 and Gal-3 induce PS exposure in neutrophils in the absence of cell death. Gal-1 and Gal-3 bind differently to the surfaces of T cells and only Gal-3 mobilizes intracellular Ca2+ in these cells, although Gal-1 and Gal-3 bind their respective T cell ligands with similar affinities. Although Gal-1 does not alter T cell viability, it induces IL-10 production and attenuates IFN-gamma production in activated T cells, suggesting a mechanism for Gal-1-mediated immunosuppression in vivo. These studies demonstrate that Gal-1 and Gal-3 induce differential responses in T cells and neutrophils, and identify the first factor, Gal-3, capable of inducing PS exposure with or without accompanying apoptosis in different leukocytes, thus providing a possible mechanism for galectin-mediated immunomodulation in vivo.


Assuntos
Citocinas/metabolismo , Galectina 1/fisiologia , Galectina 3/fisiologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Morte Celular/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/imunologia , Humanos , Interferon gama/biossíntese , Interleucina-10/biossíntese , Ativação Linfocitária/imunologia , Ativação de Neutrófilo/imunologia , Neutrófilos/citologia , Fosfatidilserinas/fisiologia , Transdução de Sinais/imunologia , Linfócitos T/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA