Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 26: 363-96, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19575676

RESUMEN

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.


Asunto(s)
Adhesión Celular , Movimiento Celular , Animales , Moléculas de Adhesión Celular/metabolismo , Humanos , Leucocitos/citología , Leucocitos/metabolismo , Selectinas/metabolismo
2.
J Cell Sci ; 134(18)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34435628

RESUMEN

Selectins and integrins are key players in the adhesion and signaling cascade that recruits leukocytes to inflamed tissues. Selectin binding induces ß2 integrin binding to slow leukocyte rolling. Here, a micropipette was used to characterize neutrophil adhesion to E-selectin and intercellular adhesion molecule-1 (ICAM-1) at room temperature. The time-dependent adhesion frequency displayed two-stage kinetics, with an E-selectin-mediated fast increase to a low plateau followed by a slow increase to a high plateau mediated by intermediate-affinity binding of integrin αLß2 to ICAM-1. The αLß2 activation required more than 5 s contact to E-selectin and spleen tyrosine kinase (Syk) activity. A multi-zone channel was used to analyze αLß2 activation by P-selectin in separate zones of receptors or antibodies, finding an inverse relationship between the rolling velocity on ICAM-1 and P-selectin dose, and a P-selectin dose-dependent change from bent to extended conformations with a closed headpiece that was faster at 37°C than at room temperature. Activation of αLß2 exhibited different levels of cooperativity and persistent times depending on the strength and duration of selectin stimulation. These results define the precise timing and kinetics of intermediate activation of αLß2 by E- and P-selectins.


Asunto(s)
Selectina E , Antígeno-1 Asociado a Función de Linfocito , Antígenos CD18 , Adhesión Celular , Selectina E/genética , Selectina E/metabolismo , Molécula 1 de Adhesión Intercelular , Cinética , Neutrófilos/metabolismo , Selectina-P
3.
Blood ; 137(20): 2756-2769, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-33619517

RESUMEN

During early embryonic development in mammals, including humans and mice, megakaryocytes (Mks) first originate from primitive hematopoiesis in the yolk sac. These embryonic Mks (eMks) circulate in the vasculature with unclear function. Herein, we report that podoplanin (PDPN), the ligand of C-type lectin-like receptor (CLEC-2) on Mks/platelets, is temporarily expressed in neural tissue during midgestation in mice. Loss of PDPN or CLEC-2 resulted in aneurysms and spontaneous hemorrhage, specifically in the lower diencephalon during midgestation. Surprisingly, more eMks/platelets had enhanced granule release and localized to the lower diencephalon in mutant mouse embryos than in wild-type littermates before hemorrhage. We found that PDPN counteracted the collagen-1-induced secretion of angiopoietin-1 from fetal Mks, which coincided with enhanced TIE-2 activation in aneurysm-like sprouts of PDPN-deficient embryos. Blocking platelet activation prevented the PDPN-deficient embryo from developing vascular defects. Our data reveal a new role for PDPN in regulating eMk function during midgestation.


Asunto(s)
Encéfalo/irrigación sanguínea , Aneurisma Intracraneal/etiología , Megacariocitos/patología , Glicoproteínas de Membrana/deficiencia , Aneurisma Roto/embriología , Aneurisma Roto/etiología , Angiopoyetina 1/metabolismo , Animales , Encéfalo/embriología , Células Cultivadas , Hemorragia Cerebral/embriología , Hemorragia Cerebral/etiología , Colágeno/farmacología , Diencéfalo/irrigación sanguínea , Diencéfalo/embriología , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Aneurisma Intracraneal/embriología , Aneurisma Intracraneal/genética , Aneurisma Intracraneal/patología , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Lectinas Tipo C/fisiología , Megacariocitos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Noqueados , Neovascularización Patológica/genética , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Activación Plaquetaria , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Receptor TIE-2/metabolismo
4.
J Immunol ; 204(1): 37-48, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31757864

RESUMEN

During inflammation, both neutrophils and effector T cells use selectins to roll and integrins to arrest in postcapillary venules. In both cell types, chemokines can transduce signals that convert integrin αLß2 to a high-affinity conformation, which interacts with ICAM-1 to mediate arrest. In neutrophils, selectins also trigger an immunoreceptor-like signaling cascade that converts integrin αLß2 to an intermediate-affinity conformation, which interacts with ICAM-1 to slow rolling. It is not known whether selectins induce similar signaling events in T cells. Ag engagement causes phosphorylation of ITAMs on the TCR; these motifs recruit kinases and adaptors that lead to the activation of αLß2. We found that mouse Th1 cells rolling on P- or E-selectin triggered signals that promoted αLß2-dependent slow rolling on ICAM-1 in vitro and in vivo. The selectin signaling cascade resembled that used by the TCR, except that unexpectedly, Th1 cells employed the ITAM-bearing protein DAP12, which was not known to be expressed in these cells. Importantly, outside-in signaling through ligand-occupied αLß2 also required DAP12. Cooperative selectin and chemokine signaling in Th1 cells promoted αLß2-dependent slow rolling and arrest in vitro and in vivo and migration into Ag-challenged tissues in vivo. Our findings reveal an important function for DAP12 in Th1 cells and a new mechanism to recruit effector T cells to sites of inflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Células TH1/inmunología , Animales , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Blood ; 132(13): 1426-1437, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30068506

RESUMEN

Inflammation is a major contributor to deep vein thrombosis (DVT). Flow restriction of the inferior vena cava (IVC) in mice induces DVT like that in humans. In this model, P-selectin-dependent adhesion of neutrophils and monocytes leads to release of neutrophil extracellular traps (NETs) and expression of tissue factor. However, it is not known what signals cause myeloid cells to generate these procoagulant effectors. Using ultrasonography and spinning-disk intravital microscopy in genetically engineered mice, we found that engagement of P-selectin glycoprotein ligand-1 (PSGL-1) and the chemokine receptor CXCR2 on rolling neutrophils propagated signals that cooperated to induce ß2 integrin-dependent arrest in flow-restricted IVCs. Unlike previous reports, PSGL-1 signaling in neutrophils did not require L-selectin, and it used tyrosine 145 rather than tyrosines 112 and 128 on the adaptor Src homology domain-containing leukocyte phosphoprotein of 76 kDa. PSGL-1 and CXCR2 signaling cooperated to increase the frequency and size of thrombi, in part by stimulating release of NETs. Unlike in neutrophils, blocking PSGL-1 or CXCR2 signaling in monocytes did not affect their recruitment into thrombi or their expression of tissue factor. Our results demonstrate that neutrophils cooperatively signal through PSGL-1 and CXCR2 to promote DVT.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Neutrófilos/metabolismo , Receptores de Interleucina-8B/metabolismo , Transducción de Señal , Trombosis de la Vena/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Neutrófilos/patología , Trombosis de la Vena/patología
6.
Proc Natl Acad Sci U S A ; 114(31): 8360-8365, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28716912

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Galactosiltransferasas/genética , Macrófagos del Hígado/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Polisacáridos/metabolismo , Animales , Receptor de Asialoglicoproteína/metabolismo , Hepatocitos/metabolismo , Homeostasis/fisiología , Lectinas Tipo C/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombocitopenia/patología
7.
Blood ; 130(2): 181-191, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28515093

RESUMEN

Leukocyte adhesion to P-selectin on activated platelets and endothelial cells induces shedding of the P-selectin ectodomain into the circulation. Plasma soluble P-selectin (sP-selectin) is elevated threefold to fourfold in patients with cardiovascular disease. Circulating sP-selectin is thought to trigger signaling in leukocytes that directly contributes to inflammation and thrombosis. However, sP-selectin likely circulates as a monomer, and in vitro studies suggest that sP-selectin must dimerize to induce signaling in leukocytes. To address this discrepancy, we expressed the entire ectodomain of mouse P-selectin as a monomer (sP-selectin) or as a disulfide-linked dimer fused to the Fc portion of mouse immunoglobulin G (sP-selectin-Fc). Dimeric sP-selectin-Fc, but not monomeric sP-selectin, triggered integrin-dependent adhesion of mouse leukocytes in vitro. Antibody-induced oligomerization of sP-selectin or sP-selectin-Fc was required to trigger formation of neutrophil extracellular traps. Injecting sP-selectin-Fc, but not sP-selectin, into mice augmented integrin-dependent adhesion of neutrophils in venules, generated tissue factor-bearing microparticles, shortened plasma-clotting times, and increased thrombus frequency in the inferior vena cava. Furthermore, transgenic mice that overexpressed monomeric sP-selectin did not exhibit increased inflammation or thrombosis. We conclude that elevated plasma sP-selectin is a consequence rather than a cause of cardiovascular disease.


Asunto(s)
Trampas Extracelulares/inmunología , Neutrófilos/inmunología , Selectina-P/sangre , Trombosis/genética , Vena Cava Inferior/inmunología , Animales , Anticuerpos/farmacología , Antígenos CD18/genética , Antígenos CD18/inmunología , Células CHO , Adhesión Celular/efectos de los fármacos , Cricetulus , Disulfuros/química , Trampas Extracelulares/efectos de los fármacos , Regulación de la Expresión Génica , Fragmentos Fc de Inmunoglobulinas/sangre , Fragmentos Fc de Inmunoglobulinas/genética , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Selectina-P/química , Selectina-P/genética , Selectina-P/inmunología , Dominios Proteicos , Multimerización de Proteína , Proteínas Recombinantes de Fusión/sangre , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Transducción de Señal , Tromboplastina/genética , Tromboplastina/inmunología , Trombosis/inmunología , Trombosis/patología , Vena Cava Inferior/patología
8.
Nature ; 502(7469): 105-9, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23995678

RESUMEN

Circulating lymphocytes continuously enter lymph nodes for immune surveillance through specialized blood vessels named high endothelial venules, a process that increases markedly during immune responses. How high endothelial venules (HEVs) permit lymphocyte transmigration while maintaining vascular integrity is unknown. Here we report a role for the transmembrane O-glycoprotein podoplanin (PDPN, also known as gp38 and T1α) in maintaining HEV barrier function. Mice with postnatal deletion of Pdpn lost HEV integrity and exhibited spontaneous bleeding in mucosal lymph nodes, and bleeding in the draining peripheral lymph nodes after immunization. Blocking lymphocyte homing rescued bleeding, indicating that PDPN is required to protect the barrier function of HEVs during lymphocyte trafficking. Further analyses demonstrated that PDPN expressed on fibroblastic reticular cells, which surround HEVs, functions as an activating ligand for platelet C-type lectin-like receptor 2 (CLEC-2, also known as CLEC1B). Mice lacking fibroblastic reticular cell PDPN or platelet CLEC-2 exhibited significantly reduced levels of VE-cadherin (also known as CDH5), which is essential for overall vascular integrity, on HEVs. Infusion of wild-type platelets restored HEV integrity in Clec-2-deficient mice. Activation of CLEC-2 induced release of sphingosine-1-phosphate from platelets, which promoted expression of VE-cadherin on HEVs ex vivo. Furthermore, draining peripheral lymph nodes of immunized mice lacking sphingosine-1-phosphate had impaired HEV integrity similar to Pdpn- and Clec-2-deficient mice. These data demonstrate that local sphingosine-1-phosphate release after PDPN-CLEC-2-mediated platelet activation is critical for HEV integrity during immune responses.


Asunto(s)
Endotelio Linfático/metabolismo , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Endotelio Linfático/inmunología , Femenino , Regulación de la Expresión Génica , Uniones Intercelulares/genética , Uniones Intercelulares/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Lisofosfolípidos/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Esfingosina/análogos & derivados , Esfingosina/metabolismo
9.
J Biol Chem ; 292(6): 2510-2518, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28011641

RESUMEN

Selectin interactions with fucosylated glycan ligands mediate leukocyte rolling in the vasculature under shear forces. Crystal structures of P- and E-selectin suggest a two-state model in which ligand binding to the lectin domain closes loop 83-89 around the Ca2+ coordination site, enabling Glu-88 to engage Ca2+ and fucose. This triggers further allostery that opens the lectin/EGF domain hinge. The model posits that force accelerates transition from the bent (low affinity) to the extended (high affinity) state. However, transition intermediates have not been described, and the role of Glu-88 in force-assisted allostery has not been examined. Here we report the structure of the lectin and EGF domains of L-selectin bound to a fucose mimetic; that is, a terminal mannose on an N-glycan attached to a symmetry-related molecule. The structure is a transition intermediate where loop 83-89 closes to engage Ca2+ and mannose without triggering allostery that opens the lectin/EGF domain hinge. We used three complementary assays to compare ligand binding to WT selectins and to E88D selectins that replaced Glu-88 with Asp. Soluble P-selectinE88D bound with an ∼9-fold lower affinity to PSGL-1, a physiological ligand, due to faster dissociation. Adhesion frequency experiments with a biomembrane force probe could not detect interactions of P-selectinE88D with PSGL-1. Cells expressing transmembrane P-selectinE88D or L-selectinE88D detached from immobilized ligands immediately after initiating flow. Cells expressing E-selectinE88D rolled but detached faster. Our data support a two-state model for selectins in which Glu-88 must engage ligand to trigger allostery that stabilizes the high affinity state under force.


Asunto(s)
Ácido Glutámico/metabolismo , Selectina L/metabolismo , Polisacáridos/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Cristalografía por Rayos X , Humanos , Selectina L/química , Glicoproteínas de Membrana/metabolismo , Conformación Proteica
10.
Proc Natl Acad Sci U S A ; 112(28): 8661-6, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26124096

RESUMEN

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.


Asunto(s)
Leucocitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Microdominios de Membrana/metabolismo , Polisacáridos/metabolismo , Animales , Receptores de Hialuranos/metabolismo , Leucosialina/metabolismo , Ligandos , Ratones
11.
J Biol Chem ; 291(3): 1441-7, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26631722

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Selectina-P/metabolismo , Regiones Promotoras Genéticas , Animales , Secuencia de Bases , Cruzamientos Genéticos , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Rodamiento de Leucocito/efectos de los fármacos , Rodamiento de Leucocito/inmunología , Lipopolisacáridos/toxicidad , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Selectina-P/química , Selectina-P/genética , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/metabolismo , Especificidad de la Especie , Organismos Libres de Patógenos Específicos , Factor de Necrosis Tumoral alfa/metabolismo , Vénulas/efectos de los fármacos , Vénulas/inmunología
13.
Arterioscler Thromb Vasc Biol ; 36(6): 1114-21, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27102967

RESUMEN

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.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Selectina-P/metabolismo , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Plaquetas/metabolismo , Trasplante de Médula Ósea , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Selectina-P/genética , Fenotipo , Placa Aterosclerótica , Factores de Tiempo , Trombosis de la Vena/genética , Trombosis de la Vena/metabolismo
14.
J Immunol ; 195(8): 3880-9, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26355151

RESUMEN

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.


Asunto(s)
Rodamiento de Leucocito/inmunología , Neutrófilos/inmunología , Receptor de Adenosina A2A/inmunología , Transducción de Señal/inmunología , Animales , Antígenos CD18/genética , Antígenos CD18/inmunología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/inmunología , Humanos , Rodamiento de Leucocito/efectos de los fármacos , Ratones , Ratones Noqueados , Neutrófilos/citología , Piperidinas/farmacología , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/inmunología , Receptor de Adenosina A2A/genética , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Familia-src Quinasas/genética , Familia-src Quinasas/inmunología
15.
Blood ; 124(24): 3656-65, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25336627

RESUMEN

O-glycosylation of podoplanin (PDPN) on lymphatic endothelial cells is critical for the separation of blood and lymphatic systems by interacting with platelet C-type lectin-like receptor 2 during development. However, how O-glycosylation controls endothelial PDPN function and expression remains unclear. In this study, we report that core 1 O-glycan-deficient or desialylated PDPN was highly susceptible to proteolytic degradation by various proteases, including metalloproteinases (MMP)-2/9. We found that the lymph contained activated MMP-2/9 and incubation of the lymph reduced surface levels of PDPN on core 1 O-glycan-deficient endothelial cells, but not on wild-type ECs. The lymph from mice with sepsis induced by cecal ligation and puncture, which contained bacteria-derived sialidase, reduced PDPN levels on wild-type ECs. The MMP inhibitor, GM6001, rescued these reductions. Additionally, GM6001 treatment rescued the reduction of PDPN level on lymphatic endothelial cells in mice lacking endothelial core 1 O-glycan or cecal ligation and puncture-treated mice. Furthermore, core 1 O-glycan-deficient or desialylated PDPN impaired platelet interaction under physiological flow. These data indicate that sialylated O-glycans of PDPN are essential for platelet adhesion and prevent PDPN from proteolytic degradation primarily mediated by MMPs in the lymph.


Asunto(s)
Plaquetas/metabolismo , Comunicación Celular/fisiología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/fisiología , Glicoproteínas de Membrana/metabolismo , Polisacáridos/biosíntesis , Animales , Plaquetas/citología , Células CHO , Comunicación Celular/efectos de los fármacos , Cricetulus , Dipéptidos/farmacología , Células Endoteliales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Glicosilación/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Adhesividad Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/fisiología , Polisacáridos/genética , Ácidos Siálicos/genética , Ácidos Siálicos/metabolismo
16.
J Biol Chem ; 289(51): 35159-71, 2014 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-25359776

RESUMEN

Interactions of CD44 on neutrophils with E-selectin on activated endothelial cells mediate rolling under flow, a prerequisite for neutrophil arrest and migration into perivascular tissues. How CD44 functions as a rolling ligand despite its weak affinity for E-selectin is unknown. We examined the nanometer scale organization of CD44 on intact cells. CD44 on leukocytes and transfected K562 cells was cross-linked within a 1.14-nm spacer. Depolymerizing actin with latrunculin B reduced cross-linking. Fluorescence resonance energy transfer (FRET) revealed tight co-clustering between CD44 fused to yellow fluorescent protein (YFP) and CD44 fused to cyan fluorescent protein on K562 cells. Latrunculin B reduced FRET-reported co-clustering. Number and brightness analysis confirmed actin-dependent CD44-YFP clusters on living cells. CD44 lacking binding sites for ankyrin and for ezrin/radixin/moesin (ERM) proteins on its cytoplasmic domain (ΔANKΔERM) did not cluster. Unexpectedly, CD44 lacking only the ankyrin-binding site (ΔANK) formed larger but looser clusters. Fluorescence recovery after photobleaching demonstrated increased CD44 mobility by latrunculin B treatment or by deleting the cytoplasmic domain. ΔANKΔERM mobility increased only modestly, suggesting that the cytoplasmic domain engages the cytoskeleton by an additional mechanism. Ex vivo differentiated CD44-deficient neutrophils expressing exogenous CD44 rolled on E-selectin and activated Src kinases after binding anti-CD44 antibody. In contrast, differentiated neutrophils expressing ΔANK had impaired rolling and kinase activation. These data demonstrate that spectrin and actin networks regulate CD44 clustering and suggest that ankyrin enhances CD44-mediated neutrophil rolling and signaling.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Membrana Celular/metabolismo , Selectina E/metabolismo , Receptores de Hialuranos/metabolismo , Animales , Ancirinas/genética , Ancirinas/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Selectina E/genética , Femenino , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Receptores de Hialuranos/genética , Células K562 , Rodamiento de Leucocito/efectos de los fármacos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Microscopía Confocal , Mutación , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Unión Proteica/efectos de los fármacos , Tiazolidinas/farmacología
17.
Blood ; 122(23): 3832-42, 2013 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-24081661

RESUMEN

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.


Asunto(s)
Quimiocina CXCL1/metabolismo , Receptor gp130 de Citocinas/deficiencia , Células Endoteliales/fisiología , Neutrófilos/fisiología , Animales , Permeabilidad Capilar/fisiología , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Quimiocina CXCL1/genética , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/fisiología , Inflamación/fisiopatología , Rodamiento de Leucocito/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Selectina-P/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Regulación hacia Arriba , Vénulas/fisiología
18.
Blood ; 121(14): 2743-52, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23372168

RESUMEN

Three isoforms of phosphatidylinositol-4-phosphate 5-kinase (PIP5KIα, PIP5KIß, and PIP5KIγ) can each catalyze the final step in the synthesis of phosphatidylinositol-4,5-bisphosphate (PIP2), which in turn can be either converted to second messengers or bind directly to and thereby regulate proteins such as talin. A widely quoted model speculates that only p90, a longer splice form of platelet-specific PIP5KIγ, but not the shorter p87 PIP5KIγ, regulates the ligand-binding activity of integrins via talin. However, when we used mice genetically engineered to lack only p90 PIP5KIγ, we found that p90 PIP5KIγ is not critical for integrin activation or platelet adhesion on collagen. However, p90 PIP5KIγ-null platelets do have impaired anchoring of their integrins to the underlying cytoskeleton. Platelets lacking both the p90 and p87 PIP5KIγ isoforms had normal integrin activation and actin dynamics, but impaired anchoring of their integrins to the cytoskeleton. Most importantly, they formed weak shear-resistant adhesions ex vivo and unstable vascular occlusions in vivo. Together, our studies demonstrate that, although PIP5KIγ is essential for normal platelet function, individual isoforms of PIP5KIγ fulfill unique roles for the integrin-dependent integrity of the membrane cytoskeleton and for the stabilization of platelet adhesion.


Asunto(s)
Plaquetas/citología , Plaquetas/enzimología , Integrinas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Adhesividad Plaquetaria/fisiología , Trombosis/enzimología , Citoesqueleto de Actina/fisiología , Empalme Alternativo/genética , Animales , Citoesqueleto/fisiología , Exones/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Isomerismo , Megacariocitos/citología , Megacariocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pinzas Ópticas , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Embarazo , Talina/metabolismo , Trombosis/genética
19.
J Biol Chem ; 287(23): 19585-98, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22511754

RESUMEN

In inflamed venules, neutrophils roll on P- or E-selectin, engage P-selectin glycoprotein ligand-1 (PSGL-1), and signal extension of integrin α(L)ß(2) in a low affinity state to slow rolling on intercellular adhesion molecule-1 (ICAM-1). Cytoskeleton-dependent receptor clustering often triggers signaling, and it has been hypothesized that the cytoplasmic domain links PSGL-1 to the cytoskeleton. Chemokines cause rolling neutrophils to fully activate α(L)ß(2), leading to arrest on ICAM-1. Cytoskeletal anchorage of α(L)ß(2) has been linked to chemokine-triggered extension and force-regulated conversion to the high affinity state. We asked whether PSGL-1 must interact with the cytoskeleton to initiate signaling and whether α(L)ß(2) must interact with the cytoskeleton to extend. Fluorescence recovery after photobleaching of transfected cells documented cytoskeletal restraint of PSGL-1. The lateral mobility of PSGL-1 similarly increased by depolymerizing actin filaments with latrunculin B or by mutating the cytoplasmic tail to impair binding to the cytoskeleton. Converting dimeric PSGL-1 to a monomer by replacing its transmembrane domain did not alter its mobility. By transducing retroviruses expressing WT or mutant PSGL-1 into bone marrow-derived macrophages from PSGL-1-deficient mice, we show that PSGL-1 required neither dimerization nor cytoskeletal anchorage to signal ß(2) integrin-dependent slow rolling on P-selectin and ICAM-1. Depolymerizing actin filaments or decreasing actomyosin tension in neutrophils did not impair PSGL-1- or chemokine-mediated integrin extension. Unlike chemokines, PSGL-1 did not signal cytoskeleton-dependent swing out of the ß(2)-hybrid domain associated with the high affinity state. The cytoskeletal independence of PSGL-1-initiated, α(L)ß(2)-mediated slow rolling differs markedly from the cytoskeletal dependence of chemokine-initiated, α(L)ß(2)-mediated arrest.


Asunto(s)
Rodamiento de Leucocito/fisiología , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Glicoproteínas de Membrana/metabolismo , Neutrófilos/metabolismo , Transducción de Señal/fisiología , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Células CHO , Quimiocinas/genética , Quimiocinas/metabolismo , Cricetinae , Cricetulus , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Neutrófilos/citología , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/fisiología , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos , Tiazolidinas/farmacología
20.
Blood ; 128(4): 467-8, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27471230
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA