Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
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
2.
JCI Insight ; 6(14)2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34291736

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a pandemic. Severe disease is associated with dysfunction of multiple organs, but some infected cells do not express ACE2, the canonical entry receptor for SARS-CoV-2. Here, we report that the C-type lectin receptor L-SIGN interacted in a Ca2+-dependent manner with high-mannose-type N-glycans on the SARS-CoV-2 spike protein. We found that L-SIGN was highly expressed on human liver sinusoidal endothelial cells (LSECs) and lymph node lymphatic endothelial cells but not on blood endothelial cells. Using high-resolution confocal microscopy imaging, we detected SARS-CoV-2 viral proteins within the LSECs from liver autopsy samples from patients with COVID-19. We found that both pseudo-typed virus enveloped with SARS-CoV-2 spike protein and authentic SARS-CoV-2 virus infected L-SIGN-expressing cells relative to control cells. Moreover, blocking L-SIGN function reduced CoV-2-type infection. These results indicate that L-SIGN is a receptor for SARS-CoV-2 infection. LSECs are major sources of the clotting factors vWF and factor VIII (FVIII). LSECs from liver autopsy samples from patients with COVID-19 expressed substantially higher levels of vWF and FVIII than LSECs from uninfected liver samples. Our data demonstrate that L-SIGN is an endothelial cell receptor for SARS-CoV-2 that may contribute to COVID-19-associated coagulopathy.


Asunto(s)
COVID-19 , Capilares , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales , Lectinas Tipo C/metabolismo , Hígado/irrigación sanguínea , Vasos Linfáticos , Receptores de Superficie Celular/metabolismo , SARS-CoV-2/fisiología , COVID-19/metabolismo , COVID-19/patología , COVID-19/virología , Capilares/metabolismo , Capilares/patología , Capilares/virología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Endoteliales/virología , Perfilación de la Expresión Génica/métodos , Humanos , Hígado/patología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Vasos Linfáticos/virología , Glicoproteína de la Espiga del Coronavirus , Internalización del Virus
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.
Science ; 370(6515): 467-472, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-33093110

RESUMEN

Colon mucus segregates the intestinal microbiota from host tissues, but how it organizes to function throughout the colon is unclear. In mice, we found that colon mucus consists of two distinct O-glycosylated entities of Muc2: a major form produced by the proximal colon, which encapsulates the fecal material including the microbiota, and a minor form derived from the distal colon, which adheres to the major form. The microbiota directs its own encapsulation by inducing Muc2 production from proximal colon goblet cells. In turn, O-glycans on proximal colon-derived Muc2 modulate the structure and function of the microbiota as well as transcription in the colon mucosa. Our work shows how proximal colon control of mucin production is an important element in the regulation of host-microbiota symbiosis.


Asunto(s)
Colon/metabolismo , Colon/microbiología , Microbioma Gastrointestinal , Mucina 2/metabolismo , Moco/metabolismo , Animales , Heces/microbiología , Glicosilación , Ratones , Ratones Noqueados , Mucina 2/genética , Transcripción Genética
5.
Blood Adv ; 4(15): 3559-3571, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32761234

RESUMEN

Ezrin/radixin/moesin (ERM) proteins are adaptors that link the actin cytoskeleton to the cytoplasmic domains of membrane proteins. Leukocytes express mostly moesin with lower levels of ezrin but no radixin. When leukocytes are activated, ERMs are postulated to redistribute membrane proteins from microvilli into uropods during polarization and to transduce signals that influence adhesion and other responses. However, these functions have not been tested in leukocytes lacking all ERMs. We used knockout (KO) mice with neutrophils lacking ezrin, moesin, or both proteins (double knockout [DKO]) to probe how ERMs modulate cell shape, adhesion, and signaling in vitro and in vivo. Surprisingly, chemokine-stimulated DKO neutrophils still polarized and redistributed ERM-binding proteins such as PSGL-1 and CD44 to the uropods. Selectin binding to PSGL-1 on moesin KO or DKO neutrophils activated kinases that enable integrin-dependent slow rolling but not those that generate neutrophil extracellular traps. Flowing neutrophils of all genotypes rolled normally on selectins and, upon chemokine stimulation, arrested on integrin ligands. However, moesin KO and DKO neutrophils exhibited defective integrin outside-in signaling and reduced adhesion strength. In vivo, DKO neutrophils displayed normal directional crawling toward a chemotactic gradient, but premature detachment markedly reduced migration from venules into inflamed tissues. Our results demonstrate that stimulated neutrophils do not require ERMs to polarize or to move membrane proteins into uropods. They also reveal an unexpected contribution of moesin to integrin outside-in signaling and adhesion strengthening.


Asunto(s)
Proteínas de la Membrana , Neutrófilos , Animales , Proteínas del Citoesqueleto , Proteínas de la Membrana/genética , Ratones , Proteínas de Microfilamentos
6.
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
7.
Blood Adv ; 3(2): 168-183, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30670533

RESUMEN

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.


Asunto(s)
Endotelio Vascular/metabolismo , Neutrófilos/metabolismo , Oncostatina M/metabolismo , Selectina-P/metabolismo , Trombosis/etiología , Trombosis/metabolismo , Vasculitis/etiología , Vasculitis/metabolismo , Animales , Biomarcadores , Adhesión Celular , Comunicación Celular , Vesículas Cubiertas por Clatrina/metabolismo , Receptor gp130 de Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Células Endoteliales/metabolismo , Humanos , Inmunofenotipificación , Rodamiento de Leucocito , Ratones , Ratones Noqueados , Modelos Biológicos , Neutrófilos/inmunología , Selectina-P/genética , Unión Proteica , Transducción de Señal , Trombosis/patología , Vasculitis/patología
8.
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
9.
JCI Insight ; 3(14)2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30046013

RESUMEN

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.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/metabolismo , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Transporte de Proteínas , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Apoptosis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Enfermedades del Desarrollo Óseo/fisiopatología , Técnicas de Cultivo de Célula , Preescolar , Condrocitos/metabolismo , Colágeno/metabolismo , Retículo Endoplásmico/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Enfermedades Genéticas Congénitas , Aparato de Golgi/metabolismo , Homeostasis , Humanos , Lipogénesis , Lisosomas/metabolismo , Manosafosfatos , Mutación
10.
Blood Adv ; 2(7): 731-744, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29592875

RESUMEN

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.


Asunto(s)
Antígenos CD18/metabolismo , Quimiocinas/fisiología , Rodamiento de Leucocito , Neutrófilos/metabolismo , Selectinas/fisiología , Animales , Quimiocinas/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Humanos , Ratones , Neutrófilos/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Selectinas/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rap1/metabolismo
11.
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
12.
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
13.
Nat Commun ; 8: 15196, 2017 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-28497779

RESUMEN

Circulating neutrophils must avoid premature activation to prevent tissue injury. The leukocyte adhesion receptor L-selectin forms bonds with P-selectin glycoprotein ligand-1 (PSGL-1) on other leukocytes and with peripheral node addressin (PNAd) on high endothelial venules. Mechanical forces can strengthen (catch) or weaken (slip) bonds between biological molecules. How these mechanochemical processes influence function in vivo is unexplored. Here we show that mice expressing an L-selectin mutant (N138G) have altered catch bonds and prolonged bond lifetimes at low forces. Basal lymphocyte homing and neutrophil recruitment to inflamed sites are normal. However, circulating neutrophils form unstable aggregates and are unexpectedly primed to respond robustly to inflammatory mediators. Priming requires signals transduced through L-selectin N138G after it engages PSGL-1 or PNAd. Priming enhances bacterial clearance but increases inflammatory injury and enlarges venous thrombi. Thus, L-selectin mechanochemistry limits premature activation of neutrophils. Our results highlight the importance of probing how mechanochemistry functions in vivo.


Asunto(s)
Fenómenos Biomecánicos/inmunología , Selectina L/metabolismo , Glicoproteínas de Membrana/metabolismo , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Animales , Antígenos de Superficie/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Escherichia coli/inmunología , Técnicas de Sustitución del Gen , Selectina L/genética , Linfocitos/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombosis de la Vena/inmunología
14.
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
15.
Blood ; 128(4): 467-8, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27471230
16.
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
18.
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
19.
J Clin Invest ; 125(12): 4349-64, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26571402

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/farmacología , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Péptidos/farmacología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Secuencias de Aminoácidos , Animales , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Péptidos/genética , Péptidos/metabolismo , Estructura Terciaria de Proteína , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA