Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Pharm ; 17(10): 3941-3951, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32931292

RESUMEN

In cancer photodynamic therapy (PDT), a photosensitizer taken up by cancer cells can generate reactive oxygen species upon near-infrared light activation to induce cancer cell death. To increase PDT potency and decrease its adverse effect, one approach is to conjugate the photosensitizer with an antibody that specifically targets cancer cells. In the present study, IR700, a hydrophilic phthalocyanine photosensitizer, was conjugated to the humanized monoclonal antibody ARB102, which binds specifically cadherin-17 (CDH17 aka CA17), a cell surface marker highly expressed in gastrointestinal cancer to produce ARB102-IR700. Photoimmunotherapy (PIT) of gastrointestinal cancer cell lines was conducted by ARB102-IR700 treatment and near-infrared light irradiation. The results showed that ARB102-IR700 PIT could induce cell death in CDH17-positive cancer cells with high potency. In a co-culture model, CDH17-negative and CDH17-overexpressing SW480 cells were labeled with distinct fluorescent dyes and cultured together prior to PIT treatment. The results confirmed that ARB102-IR700 PIT could kill CDH17-positive cells specifically, while leaving the adjacent CDH17-negative cells unaffected. An in vivo efficacy study was conducted using a pancreatic adenocarcinoma AsPC-1 xenograft tumor model in nude mice. Fluorescence scanning indicated that ARB102-IR700 accumulated specifically in the tumor sites. To perform PIT, at 24 and 48 h postinjection, mice were irradiated with a 680 nm laser at the tumor site to activate the photosensitizer. It was shown that ARB102-IR700 PIT could inhibit tumor growth significantly. In summary, this study demonstrated that the novel ARB102-IR700 is a promising agent for PIT in gastrointestinal cancers.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Cadherinas/antagonistas & inhibidores , Neoplasias Gastrointestinales/tratamiento farmacológico , Fotoquimioterapia/métodos , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos Inmunológicos/farmacología , Cadherinas/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Neoplasias Gastrointestinales/patología , Humanos , Rayos Infrarrojos , Inyecciones Intravenosas , Ratones , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Hepatology ; 49(5): 1545-53, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19291788

RESUMEN

UNLABELLED: The early stages of alcohol-induced liver injury involve chronic inflammation. Whereas mechanisms by which this effect is mediated are not completely understood, it is hypothesized that enhanced sensitivity to circulating lipopolysaccharide (LPS) contributes to this process. It has recently been shown that ethanol induces activation of plasminogen activator inhibitor-1 (PAI-1). PAI-1 causes fibrin accumulation in liver by inhibiting degradation of fibrin (fibrinolysis). LPS also enhances fibrin accumulation by activating the coagulation cascade. It was therefore hypothesized that ethanol will synergistically increase fibrin accumulation caused by LPS, enhancing liver damage. Accordingly, the effect of ethanol pretreatment on LPS-induced liver injury and fibrin deposition was determined in mice. Ethanol enhanced liver damage caused by LPS, as determined by plasma parameters and histological indices of inflammation and damage. This effect was concomitant with a significant increase in PAI-1 expression. Extracellular fibrin accumulation caused by LPS was also robustly increased by ethanol preexposure. Coadministration of the thrombin inhibitor hirudin or the MEK (mitogen-activated protein kinase) inhibitor U0126 significantly attenuated the enhanced liver damage caused by ethanol preexposure; this protection correlated with a significant blunting of the induction of PAI-1 caused by ethanol/LPS. Furthermore, thrombin/MEK inhibition prevented the synergistic effect of ethanol on the extracellular accumulation of fibrin caused by LPS. Similar protective effects on fibrin accumulation were observed in tumor necrosis factor receptor 1 (TNFR-1)(-/-) mice or in wild-type injected with PAI-1-inactivating antibody. CONCLUSION: These results suggest that enhanced LPS-induced liver injury caused by ethanol is mediated, at least in part, by fibrin accumulation in livers, mediated by an inhibition of fibrinolysis by PAI-1. These results also support the hypothesis that fibrin accumulation may play a critical role in the development of early alcohol-induced liver injury.


Asunto(s)
Etanol/toxicidad , Fibrina/metabolismo , Lipopolisacáridos/toxicidad , Hepatopatías Alcohólicas/metabolismo , Hígado/metabolismo , Consumo de Bebidas Alcohólicas/efectos adversos , Animales , Antitrombina III , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos , Hígado/patología , Hepatopatías Alcohólicas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptido Hidrolasas/sangre , Inhibidor 1 de Activador Plasminogénico/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Triglicéridos/metabolismo
3.
J Immunoassay Immunochem ; 29(1): 42-57, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18080879

RESUMEN

Functions and binding properties of four CD11c-specific mAbs are described here. The mAb 496B stimulated, while 496K inhibited ligand binding of CD11c. The stimulatory mAb, 496B, as well as the inhibitory mAbs BU15 and 496 K appear to act allosterically, as they do not bind the CD11c I domain. The mAb 3.9 bound preferentially to activated forms of CD11c and the binding was divalent cation dependent. CD11c binding to 3.9 recapitulates many of the integrin-ligand interactions. Our data suggest that 3.9 is a competitive antagonist, BU15 and 496K are allosteric antagonists, and 496B is an allosteric agonist of CD11c. These mAbs provide a set of tools to study the functions of the dendritic cell marker, CD11c.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígeno CD11c/análisis , Antígeno CD11c/inmunología , Anticuerpos Bloqueadores/inmunología , Cationes Bivalentes/química , Humanos
4.
Biochem Biophys Res Commun ; 358(3): 938-41, 2007 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-17512497

RESUMEN

Natural Killer (NK) cells kill certain tumor cells and virus infected cells in an antigen-independent manner. Members of CD18 integrins such as CD11a, CD11b, and CD11c are expressed in all NK cells. CD18-blocking mAbs inhibit the killing activity of NK cells implying an essential role of these integrins in NK cell cytotoxicity. In this report we show that the pan CD18-activating mAb, 240Q, augments cytotoxicity of resting NK cells. Since activation of either CD11a or CD11c alone fails to augment the NK cell activity, we postulate that a functional synergy of the individual CD18 integrins is responsible for the observed stimulatory effect of pan CD18 activation on NK cell cytotoxicity.


Asunto(s)
Anticuerpos/química , Antígenos CD18/biosíntesis , Integrinas/metabolismo , Células Asesinas Naturales/citología , Anticuerpos Monoclonales/química , Antígeno CD11a/metabolismo , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Adhesión Celular , Citometría de Flujo , Humanos , Hibridomas/metabolismo , Células K562 , Células Asesinas Naturales/metabolismo
5.
J Leukoc Biol ; 81(6): 1395-403, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17389580

RESUMEN

CD11c, a member of the leukointegrin family, is expressed prominently on tissue macrophages and dendritic cells and binds to complement fragment (iC3b), provisional matrix molecules (fibrinogen), and the Ig superfamily cell adhesion molecule, ICAM-1. CD11c has been proposed to function in phagocytosis, cell migration, and cytokine production by monocytes/macrophages as well as induction of T cell proliferation by Langerhans cells. Using assays to quantify CD11c-mediated cell adhesion, we demonstrate that CD11c recognizes ICAM-2 and VCAM-1. The CD11c-binding site on VCAM-1 appears to be different from that used by the integrin alpha4. CD11c and alpha4beta1 contributed to monocyte capture and transmigration on inflamed human aortic endothelial cells. We discovered that the anti-mouse CD11c mAb N418 blocks CD11c binding to iC3b, ICAM-1, and VCAM-1. Treatment of mice with N418 reduced SRBC-induced delayed-type hypersensitivity significantly. CD11c appeared to contribute predominantly to the sensitization phase and somewhat less to the response to SRBC challenge. This suggests a novel role for CD11c during leukocyte recruitment, antigen uptake, and the survival of APC.


Asunto(s)
Antígeno CD11c/inmunología , Antígenos CD18/inmunología , Hipersensibilidad Tardía/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Antígenos CD/metabolismo , Aorta/citología , Sitios de Unión , Antígeno CD11c/metabolismo , Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Complemento C3b/inmunología , Células Endoteliales/inmunología , Células Endoteliales/fisiología , Eritrocitos/inmunología , Humanos , Integrina alfa4/metabolismo , Ligandos , Ratones , Monocitos/inmunología , Monocitos/fisiología , Unión Proteica , Ovinos , Molécula 1 de Adhesión Celular Vascular/metabolismo
6.
Adv Immunol ; 91: 111-57, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16938539

RESUMEN

Initially linked to the pathogenesis of inflammatory and hematologic diseases, integrins have become validated drug targets with the approval of five drugs. Moreover, there are several promising drug candidates in preclinical and clinical stages of development for multiple clinical indications. Integrins are attractive drug targets as their antagonism can block several steps in disease progression or maintenance. Integrin inhibitors can block the proliferation, migration, or tissue localization of inflammatory, angiogenic, and tumor cells, as well as signaling and gene expression contributing to disease. There has been a rapid increase in the elucidation of integrin structure, their allosteric mechanisms of bidirectional signaling, and the structure of complexes with drugs. This information brings greater focus to how integrins support various cellular functions and how they have been and may be targeted to develop novel drugs. Here we review conformational switches, including an internal ligand, which allosterically regulate the transition from low- to high-affinity ligand binding. We address some of the successes, disappointments, and challenges in targeting competitive or allosteric sites to develop therapeutics. We also discuss new opportunities, including a structure-based approach to discover novel drugs to treat inflammatory and other diseases. This approach targets structural relatives of the von Willebrand factor A-domain present in integrins and many functionally diverse proteins.


Asunto(s)
Integrinas/química , Integrinas/metabolismo , Animales , Enfermedades Hematológicas/inmunología , Enfermedades Hematológicas/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo
7.
Blood ; 107(5): 2101-11, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16269618

RESUMEN

Polymorphonuclear leukocyte (PMN) recruitment to vascular endothelium during acute inflammation involves cooperation between selectins, G-proteins, and beta2-integrins. LFA-1 (CD11a/CD18) affinity correlates with specific adhesion functions because a shift from low to intermediate affinity supports rolling on ICAM-1, whereas high affinity is associated with shear-resistant leukocyte arrest. We imaged PMN adhesion on cytokine-inflamed endothelium in a parallel-plate flow chamber to define the dynamics of beta2-integrin function during recruitment and transmigration. After arrest on inflamed endothelium, high-affinity LFA-1 aligned along the uropod-pseudopod major axis, which was essential for efficient neutrophil polarization and subsequent transmigration. An allosteric small molecule inhibitor targeted to the I-domain stabilized LFA-1 in an intermediate-affinity conformation, which supported neutrophil rolling but inhibited cell polarization and abrogated transmigration. We conclude that a shift in LFA-1 from intermediate to high affinity during the transition from rolling to arrest provides the contact-mediated signaling and guidance necessary for PMN transmigration on inflamed endothelium.


Asunto(s)
Endotelio Vascular/metabolismo , Rodamiento de Leucocito , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Neutrófilos/metabolismo , Transducción de Señal , Adhesión Celular , Células Cultivadas , Técnicas de Cocultivo , Endotelio Vascular/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Molécula 1 de Adhesión Intercelular/metabolismo , Microscopía Confocal , Neutrófilos/patología , Seudópodos/metabolismo , Seudópodos/patología
8.
J Biol Chem ; 280(31): 28290-8, 2005 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-15955822

RESUMEN

Neutrophil rolling and transition to arrest on inflamed endothelium are dynamically regulated by the affinity of the beta(2) integrin CD11a/CD18 (leukocyte function associated antigen 1 (LFA-1)) for binding intercellular adhesion molecule (ICAM)-1. Conformational shifts are thought to regulate molecular affinity and adhesion stability. Also critical to adhesion efficiency is membrane redistribution of active LFA-1 into dense submicron clusters where multimeric interactions occur. We examined the influences of affinity and dimerization of LFA-1 on LFA-1/ICAM-1 binding by engineering a cell-free model in which two recombinant LFA-1 heterodimers are bound to respective Fab domains of an antibody attached to latex microspheres. Binding of monomeric and dimeric ICAM-1 to dimeric LFA-1 was measured in real time by fluorescence flow cytometry. ICAM-1 dissociation kinetics were measured while LFA-1 affinity was dynamically shifted by the addition of allosteric small molecules. High affinity LFA-1 dissociated 10-fold faster when bound to monomeric compared with dimeric ICAM-1, corresponding to bond lifetimes of 25 and 330 s, respectively. Downshifting LFA-1 into an intermediate affinity state with the small molecule I domain allosteric inhibitor IC487475 decreased the difference in dissociation rates between monomeric and dimeric ICAM-1 to 4-fold. When LFA-1 was shifted into the low affinity state by lovastatin, both monomeric and dimeric ICAM-1 dissociated in less than 1 s, and the dissociation rates were within 50% of each other. These data reveal the respective importance of LFA-1 affinity and proximity in tuning bond lifetime with ICAM-1 and demonstrate a nonlinear increase in the bond lifetime of the dimer versus the monomer at higher affinity.


Asunto(s)
Adhesión Celular/fisiología , Molécula 1 de Adhesión Intercelular/fisiología , Antígeno-1 Asociado a Función de Linfocito/fisiología , Neutrófilos/fisiología , Regulación Alostérica , Sistema Libre de Células , Dimerización , Endotelio Vascular/fisiología , Citometría de Flujo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Cinética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Modelos Biológicos , Conformación Proteica
9.
Nat Immunol ; 6(5): 497-506, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15834409

RESUMEN

It is widely believed that rolling lymphocytes require successive chemokine-induced signaling for lymphocyte function-associated antigen 1 (LFA-1) to achieve a threshold avidity that will mediate lymphocyte arrest. Using an in vivo model of lymphocyte arrest, we show here that LFA-1-mediated arrest of lymphocytes rolling on high endothelial venules bearing LFA-1 ligands and chemokines was abrupt. In vitro flow chamber models showed that endothelium-presented but not soluble chemokines triggered instantaneous extension of bent LFA-1 in the absence of LFA-1 ligand engagement. To support lymphocyte adhesion, this extended LFA-1 conformation required immediate activation by its ligand, intercellular adhesion molecule 1. These data show that chemokine-triggered lymphocyte adhesiveness involves a previously unrecognized extension step that primes LFA-1 for ligand binding and firm adhesion.


Asunto(s)
Quimiocinas/metabolismo , Endotelio/metabolismo , Antígeno-1 Asociado a Función de Linfocito/química , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Linfocitos/citología , Linfocitos/metabolismo , Regulación Alostérica , Adhesión Celular , Células Cultivadas , Quimiocinas/farmacología , Citoesqueleto/metabolismo , Epítopos/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/inmunología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Conformación Proteica/efectos de los fármacos , Subunidades de Proteína/inmunología , Subunidades de Proteína/metabolismo , Solubilidad , Talina/metabolismo
10.
J Immunol ; 172(12): 7780-90, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15187162

RESUMEN

Two adhesive events critical to efficient recruitment of neutrophils at vascular sites of inflammation are up-regulation of endothelial selectins that bind sialyl Lewis(x) ligands and activation of beta(2)-integrins that support neutrophil arrest by binding ICAM-1. We have previously reported that neutrophils rolling on E-selectin are sufficient for signaling cell arrest through beta(2)-integrin binding of ICAM-1 in a process dependent upon ligation of L-selectin and P-selectin glycoprotein ligand 1 (PSGL-1). Unresolved are the spatial and temporal events that occur as E-selectin binds to human neutrophils and dynamically signals the transition from neutrophil rolling to arrest. Here we show that binding of E-selectin to sialyl Lewis(x) on L-selectin and PSGL-1 drives their colocalization into membrane caps at the trailing edge of neutrophils rolling on HUVECs and on an L-cell monolayer coexpressing E-selectin and ICAM-1. Likewise, binding of recombinant E-selectin to PMNs in suspension also elicited coclustering of L-selectin and PSGL-1 that was signaled via mitogen-activated protein kinase. Binding of recombinant E-selectin signaled activation of beta(2)-integrin to high-avidity clusters and elicited efficient neutrophil capture of beta(2)-integrin ligands in shear flow. Inhibition of p38 and p42/44 mitogen-activated protein kinase blocked the cocapping of L-selectin and PSGL-1 and the subsequent clustering of high-affinity beta(2)-integrin. Taken together, the data suggest that E-selectin is unique among selectins in its capacity for clustering sialylated ligands and transducing signals leading to neutrophil arrest in shear flow.


Asunto(s)
Antígenos CD18/metabolismo , Selectina E/fisiología , Selectina L/metabolismo , Glicoproteínas de Membrana/metabolismo , Neutrófilos/metabolismo , Agregación de Receptores , Sitios de Unión , Células Sanguíneas , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Selectina E/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neutrófilos/citología , Transporte de Proteínas , Transducción de Señal , Estrés Mecánico , Transfección
11.
Structure ; 12(3): 371-8, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15016353

RESUMEN

Complement factor B is a 90 kDa protein consisting of three domains: a three-module complement control protein, a von Willebrand factor A domain, and a C-terminal serine protease (SP) domain that adopts a default inactive (zymogen) conformation. The interaction between factor B and pathogen-bound C3b is mediated by its A domain, triggering a conformational change in factor B that ultimately creates the "C3 convertase" of the alternative complement pathway. We report the crystal structure of the A domain from factor B and show that it contains an integrin-like MIDAS motif that adopts the "open" conformation typical of integrin-ligand complexes, with an acidic residue (provided by a fortuitous crystal contact) completing the coordination of the metal ion. Modeling studies indicate that the factor B A domain can also adopt the closed conformation, supporting the hypothesis that an "integrin-like switch" is conserved in complement proteins and perhaps in 60 other A domains found within the human proteome.


Asunto(s)
Factor B del Complemento/química , Secuencia de Aminoácidos , Sitios de Unión , Complemento C2/genética , Factor B del Complemento/genética , Cristalografía por Rayos X , Integrinas/genética , Ligandos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
12.
Biochem Biophys Res Commun ; 308(4): 764-9, 2003 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-12927784

RESUMEN

Although members of the class I phosphoinositide 3-kinases (PI3Ks) have been implicated in neutrophil inflammatory responses, the contribution of the individual PI3K isoforms in neutrophil activation has not been tractable with the non-selective inhibitors, LY294002 and wortmannin. We have developed a novel series of PI3K inhibitors that is selective for PI3K delta, an isoform expressed predominantly in hematopoietic cells. In addition to being selective between members of class I PI3Ks, representatives of these inhibitors such as IC980033 and IC87114 did not inhibit any protein kinases tested. Utilizing these inhibitors we report here a novel role for PI3K delta in neutrophil activation. Inhibition of PI3K delta with IC980033 and IC87114 blocked both fMLP- and TNF1 alpha-induced neutrophil superoxide generation and elastase exocytosis. The PI3K delta inhibitor IC87114 also blocked TNF1 alpha-stimulated elastase exocytosis from neutrophils in a mouse model of inflammation. To our knowledge, this is the first in vivo efficacy demonstration of a PI3K delta inhibitor in an animal model. Inhibition of PI3K delta, however, had no effect on in vitro neutrophil bactericidal activity and Fc gamma R-stimulated superoxide generation. Thus, PI3K delta plays an essential role in certain signaling pathways of neutrophil activation and appears to be an attractive target for the development of an anti-inflammatory therapeutic.


Asunto(s)
Neutrófilos/enzimología , Neutrófilos/inmunología , Fosfatidilinositol 3-Quinasas/fisiología , Adenina/análogos & derivados , Adenina/farmacología , Androstadienos/farmacología , Animales , Antiinflamatorios/farmacología , Cromonas/farmacología , Fosfatidilinositol 3-Quinasa Clase I , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Exocitosis , Inflamación , Elastasa de Leucocito/química , Ratones , Ratones Endogámicos BALB C , Morfolinas/farmacología , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinasas/química , Isoformas de Proteínas , Quinazolinas/farmacología , Transducción de Señal , Superóxidos , Factores de Tiempo , Wortmanina
13.
Blood ; 101(11): 4437-45, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12595312

RESUMEN

Leukocyte arrest on vascular endothelium under disruptive shear flow is a multistep process that requires in situ integrin activation on the leukocyte surface by endothelium-displayed chemoattractants, primarily chemokines. A genetic deficiency of leukocyte adhesion to endothelium associated with defective beta2 integrin expression or function (LAD-1) has been described. We now report a novel severe genetic disorder in this multistep process associated with functional defects in multiple leukocyte integrins, reflected in recurrent infections, profound leukocytosis, and a bleeding tendency. This syndrome is associated with an impaired ability of neutrophil and lymphocyte beta1 and beta2 integrins to generate high avidity to their endothelial ligands and arrest cells on vascular endothelium in response to endothelial chemoattractant signals. Patient leukocytes roll normally on endothelial selectins, express intact integrins and G protein-coupled chemokine receptors (GPCR), spread on integrin ligands, and migrate normally along a chemotactic gradient. Activation of beta2 integrins in response to GPCR signals and intrinsic soluble ligand binding properties of the very late activation antigen-4 (VLA-4) integrin are also retained in patient leukocytes. Nevertheless, all integrins fail to generate firm adhesion to immobilized ligands in response to in situ GPCR-mediated activation by chemokines or chemoattractants, a result of a primary defect in integrin rearrangement at ligand-bearing contacts. This syndrome is the first example of a human integrin-activation deficiency associated with defective GPCR stimulation of integrin avidity at subsecond contacts, a key step in leukocyte arrest on vascular endothelium under shear flow.


Asunto(s)
Quimiocinas/fisiología , Endotelio Vascular/citología , Integrinas/metabolismo , Síndrome de Deficiencia de Adhesión del Leucocito/patología , Adhesión Celular , Quimiotaxis de Leucocito , Niño , Endotelio Vascular/química , Endotelio Vascular/fisiología , Humanos , Rodamiento de Leucocito , Síndrome de Deficiencia de Adhesión del Leucocito/sangre , Síndrome de Deficiencia de Adhesión del Leucocito/etiología , Leucocitos/química , Leucocitos/patología , Masculino , Perfusión , Receptores de Quimiocina/metabolismo , Estrés Mecánico , Venas Umbilicales/citología
14.
J Immunol ; 170(5): 2647-54, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12594293

RESUMEN

Neutrophil chemotaxis is a critical component of the innate immune response. Neutrophils can sense an extremely shallow gradient of chemoattractants and produce relatively robust chemotactic behavior. This directional migration requires cell polarization with actin polymerization occurring predominantly in the leading edge. Synthesis of phosphatidylinositol (3,4,5) trisphosphate (PIP3) by phosphoinositide 3-kinase (PI3K) contributes to asymmetric F-actin synthesis and cell polarization during neutrophil chemotaxis. To determine the contribution of the hemopoietic cell-restricted PI3K delta in neutrophil chemotaxis, we have developed a potent and selective PI3K delta inhibitor, IC87114. IC87114 inhibited polarized morphology of neutrophils, fMLP-stimulated PIP3 production and chemotaxis. Tracking analysis of IC87114-treated neutrophils indicated that PI3K delta activity was required for the directional component of chemotaxis, but not for random movement. Inhibition of PI3K delta, however, did not block F-actin synthesis or neutrophil adhesion. These results demonstrate that PI3K delta can play a selective role in the amplification of PIP3 levels that lead to neutrophil polarization and directional migration.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Neutrófilos/citología , Neutrófilos/enzimología , Fosfatidilinositol 3-Quinasas/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Polaridad Celular/efectos de los fármacos , Polaridad Celular/fisiología , Quimiotaxis de Leucocito/efectos de los fármacos , Cromonas/farmacología , Fosfatidilinositol 3-Quinasa Clase I , Inhibidores Enzimáticos/farmacología , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/fisiología , Morfolinas/farmacología , N-Formilmetionina Leucil-Fenilalanina/farmacología , Activación Neutrófila/efectos de los fármacos , Activación Neutrófila/fisiología , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fosfatos de Fosfatidilinositol/biosíntesis , Inhibidores de las Quinasa Fosfoinosítidos-3
15.
J Biol Chem ; 277(23): 20660-70, 2002 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-11929876

RESUMEN

Neutrophil recruitment during acute inflammation is triggered by G-protein-linked chemotactic receptors that in turn activate beta(2) integrin (CD18), deemed a critical step in facilitating cell capture and arrest under the shear force of blood flow. A conformational switch in the I domain allosteric site (IDAS) and in CD18 regulates LFA-1 affinity for endothelial ligands including intercellular adhesion molecule 1 (ICAM-1). We examined the dynamics of CD18 activation in terms of the efficiency of neutrophil capture of ICAM-1, and we correlated this with the membrane topography of 327C, an antibody that recognizes the active conformation of CD18 I-like domain. Adhesion increased in direct proportion to chemotactic stimulus rising 7-fold over a log range of interleukin-8 (IL-8). A threshold dose of approximately 75 pm IL-8, corresponding to ligation of only approximately 10-100 receptors, was sufficient to activate approximately 20,000 CD18 and a rapid boost in the capture efficiency on ICAM-1. This was accompanied by a rapid redistribution of active LFA-1, but not Mac-1, into membrane patches, a necessary component for optimum adhesion efficiency. Shear-resistant arrest on a monolayer of ICAM-1 was reversed within minutes of chemotactic stimulation correlating with a shift from high to low affinity CD18 and dispersal of patches of active CD18. Mobility of active CD18 into high avidity patches was dependent on phosphatidylinositol 3-kinase activity and not F-actin polymerization. The data reveal that the number of chemotactic receptors bound and the topography and lifetime of high affinity LFA-1 tightly regulate the efficiency of neutrophil capture on ICAM-1.


Asunto(s)
Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Neutrófilos/citología , Adulto , Regulación Alostérica , Anticuerpos Monoclonales/inmunología , Antígenos CD18/inmunología , Calcio/metabolismo , Quimiotaxis de Leucocito , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-8/farmacología , Transporte Iónico , Cinética , Activación Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA