Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 459
Filtrar
1.
Circ Res ; 132(9): e151-e168, 2023 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-37021588

RESUMO

BACKGROUND: Neutrophil migration is critical to the initiation and resolution of inflammation. Macrophage-1 antigen (Mac-1; CD11b/CD18, αMß2) is a leukocyte integrin essential for firm adhesion to endothelial ICAM-1 (intercellular adhesion molecule 1) and migration of neutrophils in the shear forces of the circulation. PDI (protein disulfide isomerase) has been reported to influence neutrophil adhesion and migration. We aimed to elucidate the molecular mechanism of PDI control of Mac-1 affinity for ICAM-1 during neutrophil migration under fluid shear. METHODS: Neutrophils isolated from whole blood were perfused over microfluidic chips coated with ICAM-1. Colocalization of Mac-1 and PDI on neutrophils was visualized by fluorescently labeled antibodies and confocal microscopy. The redox state of Mac-1 disulfide bonds was mapped by differential cysteine alkylation and mass spectrometry. Wild-type or disulfide mutant Mac-1 was expressed recombinantly in Baby Hamster Kidney cells to measure ligand affinity. Mac-1 conformations were measured by conformation-specific antibodies and molecular dynamics simulations. Neutrophils crawling on immobilized ICAM-1 were measured in presence of oxidized or reduced PDI, and the effect of PDI inhibition using isoquercetin on neutrophil crawling on inflamed endothelial cells was examined. Migration indices in the X- and Y-direction were determined and the crawling speed was calculated. RESULTS: PDI colocalized with high-affinity Mac-1 at the trailing edge of stimulated neutrophils when crawling on ICAM-1 under fluid shear. PDI cleaved 2 allosteric disulfide bonds, C169-C176 and C224-C264, in the ßI domain of the ß2 subunit, and cleavage of the C224-C264 disulfide bond selectively controls Mac-1 disengagement from ICAM-1 under fluid shear. Molecular dynamics simulations and conformation-specific antibodies reveal that cleavage of the C224-C264 bond induces conformational change and mechanical stress in the ßI domain. This allosterically alters the exposure of an αI domain epitope associated with a shift of Mac-1 to a lower-affinity state. These molecular events promote neutrophil motility in the direction of flow at high shear stress. Inhibition of PDI by isoquercetin reduces neutrophil migration in the direction of flow on endothelial cells during inflammation. CONCLUSIONS: Shear-dependent PDI cleavage of the neutrophil Mac-1 C224-C264 disulfide bond triggers Mac-1 de-adherence from ICAM-1 at the trailing edge of the cell and enables directional movement of neutrophils during inflammation.


Assuntos
Molécula 1 de Adesão Intercelular , Antígeno de Macrófago 1 , Humanos , Antígeno de Macrófago 1/fisiologia , Adesão Celular/fisiologia , Células Endoteliais , Inflamação , Movimento Celular/fisiologia , Neutrófilos
2.
Front Immunol ; 12: 626798, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33796101

RESUMO

Mammalian cells do not produce chitin, an insoluble polymer of N-acetyl-D-glucosamine (GlcNAc), although chitin is a structural component of the cell wall of pathogenic microorganisms such as Candida albicans. Mammalian cells, including cells of the innate immune system elaborate chitinases, including chitotriosidase (Chit1), which may play a role in the anti-fungal immune response. In the current study, using knockout mice, we determined the role of Chit1 against systemic candidiasis. Chit1-deficient mice showed significant decrease in kidney fungal burden compared to mice expressing the functional enzyme. Using in vitro anti-candidal neutrophil functional assays, the introduction of the Chit1:chitin digestion end-product, chitobiose (N-acetyl-D-glucosamine dimer, GlcNAc2), decreased fungal-induced neutrophil swarming and Candida killing in vitro. Also, a role for the lectin-like binding site on the neutrophil integrin CR3 (Mac-1, CD11b/CD18) was found through physiological competitive interference by chitobiose. Furthermore, chitobiose treatment of wild type mice during systemic candidiasis resulted in the significant increase in fungal burden in the kidney. These data suggest a counterproductive role of Chit1 in mounting an efficient anti-fungal defense against systemic candidiasis.


Assuntos
Candidíase/imunologia , Hexosaminidases/fisiologia , Animais , Candidíase/enzimologia , Dissacarídeos/farmacologia , Modelos Animais de Doenças , Feminino , Antígeno de Macrófago 1/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Neutrófilos/imunologia , Índice de Gravidade de Doença
3.
Exp Parasitol ; 220: 107968, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32781093

RESUMO

The parasitic protozoan Leishmania infantum resides primarily in macrophages throughout mammalian infection. Infection is initiated by deposition of the metacyclic promastigote into the dermis of a mammalian host by the sand fly vector. Promastigotes enter macrophages by ligating surface receptors such as complement receptor 3 (CR3), inducing phagocytosis of the parasite. At the binding site of metacyclic promastigotes, we observed large asymmetrical aggregates of macrophage membrane with underlying actin, resembling membrane ruffles. Actin accumulation was observed at the point of initial contact, before phagosome formation and accumulation of peri-phagosomal actin. Ruffle-like structures did not form during phagocytosis of attenuated promastigotes or during phagocytosis of the intracellular amastigote form of L. infantum. Entry of promastigotes through massive actin accumulation was associated with a subsequent delay in fusion of the parasitophorous vacuole (PV) with the lysosomal markers LAMP-1 and Cathepsin D. Actin accumulation was also associated with entry through CR3, since macrophages from CD11b knockout (KO) mice did not form massive aggregates of actin during phagocytosis of metacyclic promastigotes. Furthermore, intracellular survival of L. infantum was significantly decreased in CD11b KO compared to wild type macrophages, although entry rates were similar. We conclude that both promastigote virulence and host cell CR3 are needed for the formation of ruffle-like membrane structures at the site of metacyclic promastigote phagocytosis, and that formation of actin-rich aggregates during entry correlates with the intracellular survival of virulent promastigotes.


Assuntos
Actinas/metabolismo , Leishmania infantum/fisiologia , Leishmaniose Visceral/parasitologia , Antígeno de Macrófago 1/fisiologia , Fagocitose/fisiologia , Animais , Catepsina D/metabolismo , Membrana Celular/ultraestrutura , Cricetinae , Humanos , Leishmania infantum/patogenicidade , Leishmania infantum/ultraestrutura , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Macrófagos/parasitologia , Masculino , Mesocricetus , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Confocal , Vacúolos/parasitologia , Virulência
4.
Front Immunol ; 11: 1097, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32595635

RESUMO

Phagocytosis is a specialized process that enables cellular ingestion and clearance of microbes, dead cells and tissue debris that are too large for other endocytic routes. As such, it is an essential component of tissue homeostasis and the innate immune response, and also provides a link to the adaptive immune response. However, ingestion of large particulate materials represents a monumental task for phagocytic cells. It requires profound reorganization of the cell morphology around the target in a controlled manner, which is limited by biophysical constraints. Experimental and theoretical studies have identified critical aspects associated with the interconnected biophysical properties of the receptors, the membrane, and the actin cytoskeleton that can determine the success of large particle internalization. In this review, we will discuss the major physical constraints involved in the formation of a phagosome. Focusing on two of the most-studied types of phagocytic receptors, the Fcγ receptors and the complement receptor 3 (αMß2 integrin), we will describe the complex molecular mechanisms employed by phagocytes to overcome these physical constraints.


Assuntos
Fagocitose/imunologia , Fagocitose/fisiologia , Citoesqueleto de Actina/metabolismo , Animais , Fenômenos Biofísicos , Movimento Celular/imunologia , Movimento Celular/fisiologia , Extensões da Superfície Celular/imunologia , Extensões da Superfície Celular/fisiologia , Humanos , Ligantes , Antígeno de Macrófago 1/química , Antígeno de Macrófago 1/imunologia , Antígeno de Macrófago 1/fisiologia , Modelos Imunológicos , Miosina Tipo II/imunologia , Miosina Tipo II/fisiologia , Fagossomos/imunologia , Fagossomos/fisiologia , Conformação Proteica , Pseudópodes/imunologia , Pseudópodes/fisiologia , Receptores de IgG/química , Receptores de IgG/imunologia , Receptores de IgG/fisiologia
5.
Front Immunol ; 11: 738, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32425937

RESUMO

Phagocytic integrins are endowed with the ability to engulf and dispose of particles of different natures. Evolutionarily conserved from worms to humans, they are involved in pathogen elimination and apoptotic and tumoral cell clearance. Research in the field of integrin-mediated phagocytosis has shed light on the molecular events controlling integrin activation and their effector functions. However, there are still some aspects of the regulation of the phagocytic process that need to be clarified. Here, we have revised the molecular events controlling phagocytic integrin activation and the downstream signaling driving particle engulfment, and we have focused particularly on αMß2/CR3, αXß2/CR4, and a brief mention of αVß5/αVß3integrins.


Assuntos
Integrinas/fisiologia , Fagocitose/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Apoptose , Humanos , Integrina alfaXbeta2/fisiologia , Integrinas/química , Antígeno de Macrófago 1/fisiologia , Proteínas de Membrana/fisiologia , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Talina/fisiologia , Proteínas rap1 de Ligação ao GTP/fisiologia
6.
Eur Surg Res ; 60(1-2): 53-62, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30909276

RESUMO

BACKGROUND: Polyphosphates (PolyPs) have been reported to exert pro-inflammatory effects. However, the molecular mechanisms regulating PolyP-provoked tissue accumulation of leukocytes are not known. The aim of the present investigation was to determine the role of specific adhesion molecules in PolyP-mediated leukocyte recruitment. METHODS: PolyPs and TNF-α were intrascrotally administered, and anti-P-selectin, anti-E-selectin, anti-P-selectin glycoprotein ligand-1 (PSGL-1), anti-membrane-activated complex-1 (Mac-1), anti-lymphocyte function antigen-1 (LFA-1), and neutrophil depletion antibodies were injected intravenously or intraperitoneally. Intravital microscopy of the mouse cremaster microcirculation was used to examine leukocyte-endothelium interactions and recruitment in vivo. RESULTS: Intrascrotal injection of PolyPs increased leukocyte accumulation. Depletion of neutrophils abolished PolyP-induced leukocyte-endothelium interactions, indicating that neutrophils were the main leukocyte subtype responding to PolyP challenge. Immunoneutralization of P-selectin and PSGL-1 abolished PolyP-provoked neutrophil rolling, adhesion, and emigration. Moreover, immunoneutralization of Mac-1 and LFA-1 had no impact on neutrophil rolling but markedly reduced neutrophil adhesion and emigration evoked by PolyPs. CONCLUSION: These results suggest that P-selectin and PSGL-1 exert important roles in PolyP-induced inflammatory cell recruitment by mediating neutrophil rolling. In addition, our data show that Mac-1 and LFA-1 are necessary for supporting PolyP-triggered firm adhesion of neutrophils to microvascular endothelium. These novel findings define specific molecules as potential targets for pharmacological intervention in PolyP-dependent inflammatory diseases.


Assuntos
Comunicação Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Microcirculação/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Polifosfatos/farmacologia , Animais , Células Endoteliais/fisiologia , Antígeno-1 Associado à Função Linfocitária/fisiologia , Antígeno de Macrófago 1/fisiologia , Masculino , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia , Selectina-P/fisiologia
7.
Int Arch Allergy Immunol ; 178(4): 295-306, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30630188

RESUMO

BACKGROUND: Even in subjects who are not sensitized to house dust mite (HDM), allergic symptoms can be aggravated by exposure to dust, suggesting that innate immune responses may be involved in these processes. Since eosinophils express pattern recognition receptors, HDM may directly upregulate eosinophil functions through these re ceptors. The objective of this study was to examine whether Dermatophagoides farinae (Df), a representative HDM, or Der f 1, a major allergen of Df, modifies the effector functions of eosinophils. METHODS: Eosinophils isolated from the blood of healthy donors or allergic patients were stimulated with Df extract or Der f 1, and their adhesion to recombinant human intercellular adhesion molecule (ICAM)-1 was measured using eosinophil peroxidase assays. Generation of the eosinophil superoxide anion (O2-) was examined based on the superoxide dismutase-inhibitable reduction of cytochrome C. Eosinophil-derived neurotoxin (EDN) concentrations in cell media were measured by ELISA as a marker of degranulation. RESULTS: Df extract or Der f 1 directly induced eosinophil adhesion to ICAM-1, O2- generation, and EDN release. Anti-αM- or anti-ß2-integrin antibodies or protease-activated receptor (PAR)-2 antagonists suppressed the eosinophil adhesion, O2- generation, and EDN release induced by Df extract or Der f 1. Eosinophils from allergic patients showed higher adhesion to ICAM-1 than those from healthy donors. CONCLUSIONS: These findings suggested that Df extract and Der f 1 directly activate eosinophil functions through αMß2-integrin and PAR-2. Eosinophil activation by HDM may play roles in the aggravation of allergic symptoms, not only in HDM-sensitized patients, but also in nonsensitized patients.


Assuntos
Dermatophagoides farinae/imunologia , Eosinófilos/fisiologia , Antígeno de Macrófago 1/fisiologia , Receptor PAR-2/fisiologia , Animais , Adesão Celular , Humanos , Superóxidos/metabolismo , Regulação para Cima
8.
J Leukoc Biol ; 104(6): 1159-1171, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30088666

RESUMO

Sepsis is a severe inflammatory condition associated with high mortality. Transmigration of neutrophils into tissues increases their lifespan to promote deleterious function. Junctional adhesion molecule-C (JAM-C) plays a pivotal role in neutrophil transmigration into tissues. We aim to study the role of JAM-C on the aging of neutrophils to cause sepsis-induced acute lung injury (ALI). Sepsis was induced in C57BL/6J mice by cecal ligation and puncture (CLP) and JAM-C expression in serum was assessed. Bone marrow-derived neutrophils (BMDN) were treated with recombinant mouse JAM-C (rmJAM-C) ex vivo and their viability was assessed. CLP-operated animals were administrated with either isotype IgG or anti-JAM-C Ab at a concentration of 3 mg/kg and after 20 h, aged neutrophils (CXCR4+ ) were assessed in blood and lungs and correlated with systemic injury and inflammatory markers. Soluble JAM-C level in serum was up-regulated during sepsis. Treatment with rmJAM-C inhibited BMDN apoptosis, thereby increasing their lifespan. CLP increased the frequencies of CXCR4+ neutrophils in blood and lungs, while treatment with anti-JAM-C Ab significantly reduced the frequencies of CXCR4+ aged neutrophils. Treatment with anti-JAM-C Ab significantly reduced systemic injury markers (alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase) as well as systemic and lung inflammatory cytokines (IL-6 and IL-1ß) and chemokine (macrophage inflammatory protein-2). The blockade of JAM-C improved lung histology and reduced neutrophil contents in lungs of septic mice. Thus, reduction of the pro-inflammatory aged neutrophils by blockade of JAM-C has a novel therapeutic potential in sepsis-induced ALI.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Moléculas de Adesão Celular/antagonistas & inibidores , Terapia de Alvo Molecular , Neutrófilos/imunologia , Sepse/complicações , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/imunologia , Animais , Anticorpos/uso terapêutico , Especificidade de Anticorpos , Apoptose , Moléculas de Adesão Celular/sangue , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/fisiologia , Senescência Celular , Citocinas/sangue , Imunoglobulina G/farmacologia , Imunoglobulinas/sangue , Imunoglobulinas/imunologia , Imunoglobulinas/fisiologia , Pulmão/imunologia , Pulmão/patologia , Antígeno de Macrófago 1/efeitos dos fármacos , Antígeno de Macrófago 1/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/química , Reação em Cadeia da Polimerase em Tempo Real , Receptores CXCR4/análise , Proteínas Recombinantes/farmacologia , Migração Transendotelial e Transepitelial/efeitos dos fármacos
9.
J Biol Chem ; 293(18): 6869-6882, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29540475

RESUMO

Platelet factor 4 (PF4) is one of the most abundant cationic proteins secreted from α-granules of activated platelets. Based on its structure, PF4 was assigned to the CXC family of chemokines and has been shown to have numerous effects on myeloid leukocytes. However, the receptor for PF4 remains unknown. Here, we demonstrate that PF4 induces leukocyte responses through the integrin Mac-1 (αMß2, CD11b/CD18). Human neutrophils, monocytes, U937 monocytic and HEK293 cells expressing Mac-1 strongly adhered to immobilized PF4 in a concentration-dependent manner. The cell adhesion was partially blocked by anti-Mac-1 mAb and inhibition was enhanced when anti-Mac-1 antibodies were combined with glycosaminoglycans, suggesting that cell-surface proteoglycans act cooperatively with Mac-1. PF4 also induced Mac-1-dependent migration of human neutrophils and murine WT, but not Mac-1-deficient macrophages. Coating of Escherichia coli bacteria or latex beads with PF4 enhanced their phagocytosis by macrophages by ∼4-fold, and this process was blocked by different Mac-1 antagonists. Furthermore, PF4 potentiated phagocytosis by WT, but not Mac-1-deficient macrophages. As determined by biolayer interferometry, PF4 directly bound the αMI-domain, the major ligand-binding region of Mac-1, and this interaction was governed by a Kd of 1.3 ± 0.2 µm Using the PF4-derived peptide library, synthetic peptides duplicating the αMI-domain recognition sequences and recombinant mutant PF4 fragments, the binding sites for αMI-domain were identified in the PF4 segments Cys12-Ser26 and Ala57-Ser70 These results identify PF4 as a ligand for the integrin Mac-1 and suggest that many immune-modulating effects previously ascribed to PF4 are mediated through its interaction with Mac-1.


Assuntos
Leucócitos/metabolismo , Antígeno de Macrófago 1/metabolismo , Fator Plaquetário 4/metabolismo , Animais , Sítios de Ligação , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Escherichia coli/imunologia , Células HEK293 , Humanos , Leucócitos/citologia , Leucócitos/imunologia , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Fator Plaquetário 4/química , Fator Plaquetário 4/fisiologia
10.
J Exp Med ; 214(4): 1081-1092, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28298456

RESUMO

Recent genetic evidence supports a link between microglia and the complement system in Alzheimer's disease (AD). In this study, we uncovered a novel role for the microglial complement receptor 3 (CR3) in the regulation of soluble ß-amyloid (Aß) clearance independent of phagocytosis. Unexpectedly, ablation of CR3 in human amyloid precursor protein-transgenic mice results in decreased, rather than increased, Aß accumulation. In line with these findings, cultured microglia lacking CR3 are more efficient than wild-type cells at degrading extracellular Aß by secreting enzymatic factors, including tissue plasminogen activator. Furthermore, a small molecule modulator of CR3 reduces soluble Aß levels and Aß half-life in brain interstitial fluid (ISF), as measured by in vivo microdialysis. These results suggest that CR3 limits Aß clearance from the ISF, illustrating a novel role for CR3 and microglia in brain Aß metabolism and defining a potential new therapeutic target in AD.


Assuntos
Peptídeos beta-Amiloides/análise , Encéfalo/metabolismo , Antígeno de Macrófago 1/fisiologia , Microglia/fisiologia , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Benzoatos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteólise , Tioidantoínas/farmacologia
11.
J Exp Med ; 214(3): 851-874, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28183734

RESUMO

Integrin activation is required for neutrophil functions. Impaired integrin activation on neutrophils is the hallmark of leukocyte adhesion deficiency (LAD) syndrome in humans, characterized by impaired leukocyte recruitment and recurrent infections. The Src kinase-associated phosphoprotein 2 (Skap2) is involved in integrin functions in different leukocyte subtypes. However, the role of Skap2 in ß2 integrin activation and neutrophil recruitment is unknown. In this study, we demonstrate the crucial role of Skap2 in regulating actin polymerization and binding of talin-1 and kindlin-3 to the ß2 integrin cytoplasmic domain, thereby being indispensable for ß2 integrin activation and neutrophil recruitment. The direct interaction of Skap2 with the Wiskott-Aldrich syndrome protein via its SH3 domain is critical for integrin activation and neutrophil recruitment in vivo. Furthermore, Skap2 regulates integrin-mediated outside-in signaling events and neutrophil functions. Thus, Skap2 is essential to activate the ß2 integrins, and loss of Skap2 function is sufficient to cause a LAD-like phenotype in mice.


Assuntos
Antígenos CD18/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Infiltração de Neutrófilos , Neutrófilos/fisiologia , Animais , Adesão Celular , Quimiotaxia de Leucócito , Proteínas do Citoesqueleto/metabolismo , Selectina E/fisiologia , Antígeno de Macrófago 1/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Multimerização Proteica , Talina/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/fisiologia , Domínios de Homologia de src
13.
Eur Surg Res ; 56(1-2): 19-31, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26575178

RESUMO

BACKGROUND: Extracellular histones released during cell damage have the capacity to cause tissue injury associated with increased leukocyte accumulation. However, the molecular mechanisms regulating histone-induced leukocyte recruitment remain elusive. The objective of this study was to examine the role of adhesion molecules in histone-dependent leukocyte accumulation by use of intravital microscopy of the mouse cremaster microcirculation. METHODS: Histone 3 and TNF-α were intrascrotally administered, and anti-P-selectin, anti-P-selectin glycoprotein ligand-1 (PSGL-1), anti-membrane-activated complex-1 (Mac-1), anti-lymphocyte function antigen-1 (LFA-1) antibody and neutrophil depletion antibody were injected intravenously or intraperitoneally. RESULTS: Intrascrotal injection of histone 3 dose-dependently increased leukocyte recruitment. Neutrophil depletion abolished intravascular and extravascular leukocytes after histone 3 challenge, suggesting that neutrophils were the dominating leukocyte subtype responding to histone stimulation. Pretreatment with an anti-P-selectin and an anti-PSGL-1 antibody abolished histone-stimulated neutrophil rolling, adhesion and emigration. When the anti-P-selectin or the anti-PSGL-1 antibody was administrated after histone 3 stimulation, neutrophil rolling was reduced, whereas the number of firmly adherent and emigrated neutrophils were unchanged, suggesting that the inhibitory effect of blocking P-selectin and PSGL-1 on neutrophil adhesion and recruitment was due to the reduction in neutrophil rolling. Moreover, pretreatment with antibodies against Mac-1 and LFA-1 had no effect of neutrophil rolling but abolished adhesion and emigration evoked by histone 3. Thus, our data demonstrate that P-selectin and PSGL-1 play an important role in histone-induced inflammatory cell recruitment by mediating neutrophil rolling as a precondition for histone-provoked firm adhesion and emigration in vivo. Moreover, we conclude that both Mac-1 and LFA-1 are critical in supporting histone-provoked firm adhesion of neutrophils to endothelial cells. CONCLUSION: These novel findings define specific selectins and integrins as potential targets for pharmacological intervention in histone-dependent inflammatory diseases.


Assuntos
Endotélio Vascular/fisiologia , Histonas/farmacologia , Músculo Esquelético/irrigação sanguínea , Neutrófilos/fisiologia , Animais , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Antígeno-1 Associado à Função Linfocitária/fisiologia , Antígeno de Macrófago 1/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação , Selectina-P/fisiologia , Fator de Necrose Tumoral alfa/farmacologia
14.
Biochemistry ; 54(6): 1408-20, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25613106

RESUMO

The broad recognition specificity exhibited by integrin α(M)ß2 (Mac-1, CD11b/CD18) has allowed this adhesion receptor to play innumerable roles in leukocyte biology, yet we know little about how and why α(M)ß2 binds its multiple ligands. Within α(M)ß2, the α(M)I-domain is responsible for integrin's multiligand binding properties. To identify its recognition motif, we screened peptide libraries spanning sequences of many known protein ligands for α(M)I-domain binding and also selected the α(M)I-domain recognition sequences by phage display. Analyses of >1400 binding and nonbinding peptides derived from peptide libraries showed that a key feature of the α(M)I-domain recognition motif is a small core consisting of basic amino acids flanked by hydrophobic residues. Furthermore, the peptides selected by phage display conformed to a similar pattern. Identification of the recognition motif allowed the construction of an algorithm that reliably predicts the α(M)I-domain binding sites in the α(M)ß2 ligands. The recognition specificity of the α(M)I-domain resembles that of some chaperones, which allows it to bind segments exposed in unfolded proteins. The disclosure of the α(M)ß2 binding preferences allowed the prediction that cationic host defense peptides, which are strikingly enriched in the α(M)I-domain recognition motifs, represent a new class of α(M)ß2 ligands. This prediction has been tested by examining the interaction of α(M)ß2 with the human cathelicidin peptide LL-37. LL-37 induced a potent α(M)ß2-dependent cell migratory response and caused activation of α(M)ß2 on neutrophils. The newly revealed recognition specificity of α(M)ß2 toward unfolded protein segments and cationic proteins and peptides suggests that α(M)ß2 may serve as a previously proposed "alarmin" receptor with important roles in innate host defense.


Assuntos
Antígeno de Macrófago 1/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Ligantes , Antígeno de Macrófago 1/química , Antígeno de Macrófago 1/fisiologia , Dados de Sequência Molecular , Conformação Proteica
15.
Shock ; 42(6): 532-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25075642

RESUMO

PURPOSE: Macrophage 1 antigen (Mac-1, CD11bCD18) is a leukocyte adhesion molecule that is involved in many functions including leukocyte recruitment, phagocytosis, and neutrophil apoptosis. The previous report of mild polymicrobial, abdominal sepsis showed that the administration of anti-CD11b-blocking antibody administration attenuated lung injury without any survival benefit. Here we tested the impact of Mac-1 deficiency in severe polymicrobial abdominal sepsis model. METHODS: Polymicrobial sepsis was studied using cecal ligation and puncture model in wild-type (WT) or Mac-1-deficient (CD11b knockout [KO]) mice, and their outcomes were examined. Bacterial tissue load and the recruitment of neutrophils to the abdominal cavity were assessed. In vitro bacterial killing assay was performed. Serum cytokine levels were measured using multiarray. Apoptosis of spleen tissues was assessed using Western blot analysis and immunohistochemistry (cleaved caspase 3 and TUNEL staining). In addition, in vitro apoptosis assay was performed using primary splenocytes from both WT and KO mice. The recruitment of neutrophils to lung was assessed by measuring myeloperoxidase activity. RESULTS: Macrophage 1 antigen deficiency significantly decreased survival (survival percentage WT 43.5% vs. KO 13.0%; P = 0.0038) with higher bacterial load in blood and more severe systemic inflammation. Knockout mice demonstrated higher apoptosis both in vivo and in vitro. The recruitment of neutrophils to lung was not different between WT and KO mice. CONCLUSIONS: Macrophage 1 antigen deficiency was associated with poorer outcomes, more bacterial load, systemic inflammation, and splenic apoptosis. However, Mac-1 deficiency did not attenuate neutrophil recruitment to lung.


Assuntos
Antígeno de Macrófago 1/fisiologia , Sepse/microbiologia , Animais , Apoptose , Antígeno CD11b/metabolismo , Caspase 3/metabolismo , Adesão Celular , Citocinas/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Leucócitos/citologia , Ligantes , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Peroxidase/metabolismo , Baço/metabolismo
16.
J Leukoc Biol ; 96(3): 411-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24799603

RESUMO

The histamine H4 receptor regulates the inflammatory response. However, it is not known whether this receptor has a functional role in human neutrophils. We found that fMLP (1 µM), but not histamine (0.1-1 µM), induced Mac-1-dependent adhesion, polarization, and degranulation (release of lactoferrin). A pretreatment of neutrophils with histamine (0.001-1 µM) or JNJ 28610244 (0.1-10 µM), a specific H4 receptor agonist, led to inhibition of degranulation. Total inhibition of degranulation was obtained with 0.1 µM histamine and 10 µM JNJ 28610244. Furthermore, such inhibition by histamine of degranulation was reversed by JNJ 7777120 and JNJ 28307474, two selective H4 receptor antagonists. However, neither histamine nor the H4 receptor agonist JNJ 28610244 prevented fMLP-induced, Mac-1-dependent adhesion, indicating that the H4 receptor may block signals emanating from Mac-1-controlling degranulation. Likewise, engagement of the H4 receptor by the selective agonist JNJ 28610244 blocked Mac-1-dependent activation of p38 MAPK, the kinase that controls neutrophil degranulation. We also show expression of the H4 receptor at the mRNA level in ultrapure human neutrophils and myeloid leukemia PLB-985 cells. We concluded that engagement of this receptor by selective H4 receptor agonists may represent a good, therapeutic approach to accelerate resolution of inflammation.


Assuntos
Degranulação Celular , Neutrófilos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores Histamínicos/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Degranulação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Células Cultivadas , Citocalasina B/farmacologia , Fibrinogênio , Histamina/farmacologia , Humanos , Indóis/farmacologia , Leucemia Promielocítica Aguda/patologia , Antígeno-1 Associado à Função Linfocitária/química , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Antígeno de Macrófago 1/fisiologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/efeitos dos fármacos , Oximas/farmacologia , Piperazinas/farmacologia , Piperidinas/farmacologia , Conformação Proteica/efeitos dos fármacos , Piridinas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Histamínicos H4 , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
17.
J Surg Res ; 189(2): 304-12, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24726062

RESUMO

BACKGROUND: Fungal and bacterial coinfections are common in surgical settings; however, little is known about the effects of polymicrobial interactions on the cellular mechanisms involved in innate immune recognition and phagocytosis. MATERIALS AND METHODS: Zymosan particles, cell wall derivatives of the yeast Saccharomyces cerevisiae, are used to model fungal interactions with host immune cells since they display carbohydrates, including beta-glucan, that are characteristic of fungal pathogens. Using in vitro cell culture, RAW 264.7 macrophages were challenged with zymosan, and phagocytosis determined via light microscopy. The effects of different concentrations of lipopolysaccharide (LPS) on zymosan phagocytosis were assessed. In addition, the transfer of supernatant from LPS-treated cells to naïve cells, the effects of soluble carbohydrates laminarin, mannan, or galactomannan, and the impact of complement receptor 3 (CR3) inhibition on phagocytosis were also determined. RESULTS: LPS enhanced phagocytosis of zymosan in a dose-dependent manner. Transfer of supernatants from LPS-primed cells to naïve cells had no effect on phagocytosis. Laminarin inhibited zymosan phagocytosis in naïve cells but not in LPS-primed cells. Neither mannan, galactomannan, nor CR3 inhibition had a significant effect on ingestion of unopsonized zymosan in naïve or LPS-treated cells. CONCLUSIONS: Zymosan recognition by naïve cells is inhibited by laminarin, but not mannan, galactomannan, or CR3 inhibition. LPS enhancement of phagocytosis is laminarin insensitive and not mediated by supernatant factors or zymosan engagement by the mannose or CR3 receptors. Our data suggest alternative mechanisms of zymosan recognition in the presence and absence of LPS.


Assuntos
Lipopolissacarídeos/fisiologia , Antígeno de Macrófago 1/fisiologia , Macrófagos/fisiologia , Proteínas Opsonizantes/fisiologia , Fagocitose , Polissacarídeos/fisiologia , Zimosan/fisiologia , Animais , Anticorpos Bloqueadores/farmacologia , Linhagem Celular , Lectinas Tipo C/fisiologia , Antígeno de Macrófago 1/imunologia , Receptor de Manose , Lectinas de Ligação a Manose/fisiologia , Camundongos , Receptores de Superfície Celular/fisiologia
18.
J Clin Invest ; 124(4): 1483-96, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24642468

RESUMO

Interactions between platelets, leukocytes, and activated endothelial cells are important during microvascular occlusion; however, the regulatory mechanisms of these heterotypic cell-cell interactions remain unclear. Here, using intravital microscopy to evaluate mice lacking specific isoforms of the serine/threonine kinase AKT and bone marrow chimeras, we found that hematopoietic cell-associated AKT2 is important for neutrophil adhesion and crawling and neutrophil-platelet interactions on activated endothelial cells during TNF-α-induced venular inflammation. Studies with an AKT2-specific inhibitor and cells isolated from WT and Akt KO mice revealed that platelet- and neutrophil-associated AKT2 regulates heterotypic neutrophil-platelet aggregation under shear conditions. In particular, neutrophil AKT2 was critical for membrane translocation of αMß2 integrin, ß2-talin1 interaction, and intracellular Ca2+ mobilization. We found that the basal phosphorylation levels of AKT isoforms were markedly increased in neutrophils and platelets isolated from patients with sickle cell disease (SCD), an inherited hematological disorder associated with vascular inflammation and occlusion. AKT2 inhibition reduced heterotypic aggregation of neutrophils and platelets isolated from SCD patients and diminished neutrophil adhesion and neutrophil-platelet aggregation in SCD mice, thereby improving blood flow rates. Our results provide evidence that neutrophil AKT2 regulates αMß2 integrin function and suggest that AKT2 is important for neutrophil recruitment and neutrophil-platelet interactions under thromboinflammatory conditions such as SCD.


Assuntos
Neutrófilos/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Vasculite/fisiopatologia , Anemia Falciforme/sangue , Anemia Falciforme/patologia , Anemia Falciforme/fisiopatologia , Animais , Sinalização do Cálcio , Comunicação Celular/fisiologia , Modelos Animais de Doenças , Humanos , Antígeno de Macrófago 1/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/patologia , Agregação Plaquetária/fisiologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/genética , Talina/fisiologia , Vasculite/patologia
19.
J Pharmacol Sci ; 123(1): 1-8, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-23978960

RESUMO

Pantetheinase is an enzyme hydrolyzing pantetheine, an intermediate of the coenzyme A degradation pathway. Pantetheinase has long been considered as the enzyme that recycles pantothenic acid (vitamin B5) generated during coenzyme A breakdown. Genetic analyses showed that mammals have multiple genes known as vanin family genes. Recent studies using mice lacking the vanin-1 gene (pantetheinase gene) suggest that pantetheinase is actively involved in the progression of inflammatory reactions by generating cysteamine. Additional studies using human leukocytes demonstrate that human neutrophils have abundant pantetheinase proteins on the surface and inside the cells. The second pantetheinase protein, GPI-80/VNN2, is suggested to work as a modulator of the function of Mac-1 (CD11b/CD18), an adhesion molecule important to neutrophil functions. This review delineates the characteristics of the pantetheinase/vanin gene family and how they affect inflammation.


Assuntos
Amidoidrolases/fisiologia , Coenzima A/metabolismo , Inflamação/genética , Inflamação/metabolismo , Neutrófilos/fisiologia , Animais , Moléculas de Adesão Celular/fisiologia , Cisteamina/metabolismo , Progressão da Doença , Proteínas Ligadas por GPI/fisiologia , Humanos , Hidrólise , Antígeno de Macrófago 1/fisiologia , Família Multigênica , Neutrófilos/enzimologia , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Panteteína/metabolismo , Ácido Pantotênico/metabolismo , Proteólise
20.
Blood ; 122(20): 3473-81, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-23878142

RESUMO

Interleukin-1ß (IL-1ß) is a proinflammatory cytokine and a therapeutic target in several chronic autoimmune states. Monocytes and macrophages are the major sources of IL-1ß. IL-1ß production by these cells requires Toll-like receptor (TLR) and adenosine triphosphate (ATP)-mediated P2X purinoceptor 7 (P2X7) signals, which together activate the inflammasome. However, how TLR signals and ATP availability are regulated during monocyte activation is unclear and the involvement of another danger signal system has been proposed. Here, we demonstrate that both lipopolysaccharide (LPS) and the anaphylatoxin C3a are needed for IL-1ß production in human macrophages and dendritic cells, while in monocytes, C3a enhanced the secretion of LPS-induced IL-1ß. C3a and LPS-stimulated monocytes increased T helper 17 (Th17) cell induction in vitro, and human rejecting, but not nonrejecting, kidney transplant biopsies were characterized by local generation of C3a and monocyte and Th17 cell infiltration. Mechanistically, C3a drives IL-1ß production in monocytes by controlling the release of intracellular ATP into the extracellular space via regulation of as-yet unidentified ATP-releasing channels in an extracellular signal-regulated kinase 1/2-dependent fashion. These data define a novel function for complement in inflammasome activation in monocytes and suggest that C3aR-mediated signaling is a vital component of the IL-1ß-Th17 axis.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Transporte/fisiologia , Complemento C3/fisiologia , Inflamassomos/fisiologia , Interleucina-1beta/metabolismo , Monócitos/metabolismo , Células Cultivadas , Complemento C3/agonistas , Citocinas/biossíntese , Citocinas/genética , Células Dendríticas/metabolismo , Ativação Enzimática , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Humanos , Transplante de Rim , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases , Antígeno de Macrófago 1/efeitos dos fármacos , Antígeno de Macrófago 1/fisiologia , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptores Purinérgicos P2X7/fisiologia , Proteínas Recombinantes/farmacologia , Células Th17/metabolismo , Receptores Toll-Like/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...