Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 34(2): 2326-2343, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31907993

RESUMO

Polymorphonuclear neutrophils (PMNs) play a critical role in the innate immune response to invading pathogens. However, dysregulated mucosal trafficking of PMNs and associated epithelial tissue damage is a pathological hallmark of numerous inflammatory conditions including inflammatory bowel disease. The glycoprotein CD11b/CD18 plays a well-described role in regulating PMN transepithelial migration and PMN inflammatory functions. Previous studies have demonstrated that targeting of the N-linked glycan Lewis X on CD11b blocks PMN transepithelial migration (TEpM). Given evidence of glycosylation-dependent regulation of CD11b/CD18 function, we performed MALDI TOF Mass Spectrometry (MS) analyses on CD11b/CD18 purified from human PMNs. Unusual glycan epitopes identified on CD11b/CD18 included high Mannose oligosaccharides recognized by the Galanthus Nivalis lectin and biantennary galactosylated N-glycans recognized by the Phaseolus Vulgaris erythroagglutinin lectin. Importantly, we show that selective targeting of glycans on CD11b with such lectins results in altered intracellular signaling events that inhibit TEpM and differentially affect key PMN inflammatory functions including phagocytosis, superoxide release and apoptosis. Taken together, these data demonstrate that discrete glycan motifs expressed on CD11b/CD18 such as biantennary galactose could represent novel targets for selective manipulation of CD11b function and reduction of PMN-associated tissue damage in chronic inflammatory diseases.


Assuntos
Antígeno CD11b/imunologia , Antígenos CD18/imunologia , Epitopos/imunologia , Neutrófilos/imunologia , Antígeno CD11b/química , Antígenos CD18/química , Epitopos/química , Humanos , Lectinas de Ligação a Manose/química , Lectinas de Ligação a Manose/farmacologia , Neutrófilos/química , Fagocitose , Fito-Hemaglutininas/química , Fito-Hemaglutininas/farmacologia , Lectinas de Plantas/química , Lectinas de Plantas/farmacologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Superóxidos/química , Superóxidos/imunologia , Migração Transendotelial e Transepitelial
2.
Front Immunol ; 11: 619925, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33679708

RESUMO

Neutrophils are the most prevalent leukocytes in the human body. They have a pivotal role in the innate immune response against invading bacterial and fungal pathogens, while recent emerging evidence also demonstrates their role in cancer progression and anti-tumor responses. The efficient execution of many neutrophil effector responses requires the presence of ß2 integrins, in particular CD11a/CD18 or CD11b/CD18 heterodimers. Although extensively studied at the molecular level, the exact signaling cascades downstream of ß2 integrins still remain to be fully elucidated. In this review, we focus mainly on inside-out and outside-in signaling of these two ß2 integrin members expressed on neutrophils and describe differences between various neutrophil stimuli with respect to integrin activation, integrin ligand binding, and the pertinent differences between mouse and human studies. Last, we discuss how integrin signaling studies could be used to explore the therapeutic potential of targeting ß2 integrins and the intracellular signaling cascade in neutrophils in several, among other, inflammatory conditions in which neutrophil activity should be dampened to mitigate disease.


Assuntos
Antígenos CD18/fisiologia , Ativação de Neutrófilo/fisiologia , Neutrófilos/metabolismo , Transdução de Sinais , Animais , Anti-Inflamatórios/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/fisiologia , Antígeno CD11a/química , Antígeno CD11a/fisiologia , Antígeno CD11b/química , Antígeno CD11b/fisiologia , Antígenos CD18/química , Adesão Celular/fisiologia , Quimiocinas/farmacologia , Quimiocinas/fisiologia , Quimiotaxia de Leucócito/fisiologia , Proteínas do Citoesqueleto/metabolismo , Dimerização , Humanos , Inflamação , Camundongos , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Fagocitose/fisiologia , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Selectinas/fisiologia , Especificidade da Espécie , Talina/metabolismo , Migração Transendotelial e Transepitelial/fisiologia
3.
Angew Chem Int Ed Engl ; 59(6): 2235-2240, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31769915

RESUMO

Studying the dynamic interaction between host cells and pathogen is vital but remains technically challenging. We describe herein a time-resolved chemical proteomics strategy enabling host and pathogen temporal interaction profiling (HAPTIP) for tracking the entry of a pathogen into the host cell. A novel multifunctional chemical proteomics probe was introduced to label living bacteria followed by in vivo crosslinking of bacteria proteins to their interacting host-cell proteins at different time points initiated by UV for label-free quantitative proteomics analysis. We observed over 400 specific interacting proteins crosslinked with the probe during the formation of Salmonella-containing vacuole (SCV). This novel chemical proteomics approach provides a temporal interaction profile of host and pathogen in high throughput and would facilitate better understanding of the infection process at the molecular level.


Assuntos
Proteínas de Bactérias/química , Sondas Moleculares/química , Proteômica/métodos , Salmonella typhimurium/metabolismo , Proteínas de Bactérias/metabolismo , Antígeno CD11b/química , Antígeno CD11b/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Lipopolissacarídeos/química , Lipopolissacarídeos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Maleimidas/química , Análise de Componente Principal , Infecções por Salmonella/diagnóstico , Infecções por Salmonella/metabolismo , Infecções por Salmonella/microbiologia , Succinimidas/química , Fatores de Tempo , Raios Ultravioleta
4.
J Am Chem Soc ; 141(35): 13829-13840, 2019 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-31382746

RESUMO

Nanoparticles have been extensively used to deliver therapeutic drugs to tumor tissues through the extravasation of a leaky vessel via enhanced permeation and retention effect (EPR, passive targeting) or targeted interaction of tumor-specific ligands (active targeting). However, the therapeutic efficacy of drug-loaded nanoparticles is hampered by its heterogeneous distribution owing to limited penetration in tumor tissue. Inspired by the fact that cancer cells can recruit inflammatory immune cells to support their survival, we developed a click reaction-assisted immune cell targeting (CRAIT) strategy to deliver drug-loaded nanoparticles deep into the avascular regions of the tumor. Immune cell-targeting CD11b antibodies are modified with trans-cyclooctene to enable bioorthogonal click chemistry with mesoporous silica nanoparticles functionalized with tetrazines (MSNs-Tz). Sequential injection of modified antibodies and MSNs-Tz at intervals of 24 h results in targeted conjugation of the nanoparticles onto CD11b+ myeloid cells, which serve as active vectors into tumor interiors. We show that the CRAIT strategy allows the deep tumor penetration of drug-loaded nanoparticles, resulting in enhanced therapeutic efficacy in an orthotopic 4T1 breast tumor model. The CRAIT strategy does not require ex vivo manipulation of cells and can be applied to various types of cells and nanovehicles.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Nanopartículas/química , Dióxido de Silício/química , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antígeno CD11b/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Química Click , Ciclo-Octanos/química , Modelos Animais de Doenças , Doxorrubicina/administração & dosagem , Portadores de Fármacos/química , Ensaios de Seleção de Medicamentos Antitumorais , Camundongos , Imagem Óptica , Tamanho da Partícula , Porosidade , Propriedades de Superfície
5.
J Nucl Med ; 60(9): 1317-1324, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30796172

RESUMO

Current noninvasive imaging methods for monitoring immune response were largely developed for interrogation of the local reaction. This study developed the radiotracer 64Cu-labeled anti-CD11b (64Cu-αCD11b) for longitudinal assessment of local and systemic immune response involving mobilization of CD11b+ myeloid cells by small-animal PET/CT. Methods: Acute or chronic inflammation in the ears of BALB/c mice was induced by 12-o-tetradecanoylphorbol-13-acetate. Acute lung inflammation was induced by intratracheal lipopolysaccharide inoculation. αCD11b was conjugated with p-SCN-Bn-DOTA followed by labeling with 64Cu. PET/CT and biodistribution were evaluated at different times after intravenous injection of 64Cu-αCD11b. Cell populations from bone marrow (BM) and spleen were analyzed by flow cytometry. Results:64Cu-αCD11b was primarily taken up by BM and spleen in control mice. In comparison, 64Cu-αCD11b uptake was significantly reduced in the BM and spleen of CD11b-knockout mice, indicating that 64Cu-αCD11b selectively homed to CD11b+ myeloid cells in vivo. In mice with ear inflammation, for the local inflammatory response, 64Cu-αCD11b PET/CT revealed significantly higher 64Cu-αCD11b uptake in the inflamed ears in the acute inflammation phase than the chronic phase, consistent with markedly increased infiltration of CD11b+ cells into the inflammatory lesions at the acute phase. Moreover, imaging of 64Cu-αCD11b also showed the difference in mouse systemic response for different inflammatory stages. Compared with uptake in control mice, BM 64Cu-αCD11b uptake in mice with ear inflammation was significantly lower in the acute phase and higher in the chronic phase, reflecting an initial mobilization of CD11b+ cells from the BM to the inflammatory foci followed by a compensatory regeneration of CD11b+ myeloid cells in the BM. Similarly, in mice with lung inflammation, 64Cu-αCD11b PET/CT readily detected acute lung inflammation and recruitment of CD11b+ myeloid cells from the BM. Immunohistochemistry staining and flow cytometry results confirmed the noninvasive imaging of PET/CT. Conclusion:64Cu-αCD11b PET/CT successfully tracked ear and pulmonary inflammation in mice and differentiated acute from chronic inflammation at the local and systemic levels. 64Cu-αCD11b PET/CT is a robust quantitative method for imaging of local and systemic immune responses.


Assuntos
Antígeno CD11b/química , Radioisótopos de Cobre/química , Sistema Imunitário/diagnóstico por imagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Animais , Medula Óssea/diagnóstico por imagem , Medula Óssea/metabolismo , Feminino , Citometria de Fluxo , Compostos Heterocíclicos/química , Imuno-Histoquímica , Isotiocianatos/química , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Pneumonia/diagnóstico por imagem , Compostos Radiofarmacêuticos , Ratos , Baço/diagnóstico por imagem , Baço/metabolismo , Distribuição Tecidual
6.
Pharm Res ; 35(1): 6, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29294201

RESUMO

PURPOSE: To synthesize and assess the in vitro biological activity of nanoparticles containing leukemia inhibitory factor (LIF). These NanoLIF particles are designed to prolong the neuroprotective and anti-inflammatory actions of LIF in future preclinical studies of ischemic stroke. METHODS: LIF was packaged in nanoparticles made of poly(ethylene glycol)-poly(lactic acid) (PEG-PLA) polymer to form LIF-loaded nanoparticles (NanoLIF). The surface of NanoLIF was also modified with the CD11b antibody (CD11b-NanoLIF) targeting activated peripheral macrophages to increase cytokine delivery to inflammatory macrophages. ELISA was used to quantify bioactive cytokine inside and releasing from NanoLIF. NanoLIF biological activity was measured using the M1 murine leukemia cell proliferation assay. RESULTS: NanoLIF and CD11b-NanoLIF had diameters of approximately 30 nm, neutral surface charge, and physicochemical stability retaining biological activity of the cytokine during incubation at 25°C for 12 h. NanoLIF particles released LIF relatively fast from 0 to 6 h after incubation at 37°C followed by slow release from 24 to 72 h according to a two-phase exponential decay model. NanoLIF and CD11b-NanoLIF significantly decreased M1 cell proliferation over 72 h compared to free LIF. CONCLUSIONS: NanoLIF and CD11b-NanoLIF preserved the metabolic stability and biological activity of LIF in vitro. These results are promising to improve the therapeutic potential of LIF in treating neurodegenerative and inflammatory diseases.


Assuntos
Anti-Inflamatórios/farmacologia , Citocinas/metabolismo , Fator Inibidor de Leucemia/farmacologia , Nanopartículas/química , Polietilenoglicóis/química , Animais , Isquemia Encefálica/tratamento farmacológico , Antígeno CD11b/química , Antígeno CD11b/imunologia , Linhagem Celular Tumoral , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Humanos , Fator Inibidor de Leucemia/química , Fator Inibidor de Leucemia/imunologia , Fator Inibidor de Leucemia/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Doenças Neurodegenerativas , Tamanho da Partícula , Acidente Vascular Cerebral/tratamento farmacológico , Propriedades de Superfície
7.
J Biol Chem ; 292(50): 20756-20768, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29079572

RESUMO

The ligand-binding ßI and αI domains of integrin are the best-studied von Willebrand factor A domains undergoing significant conformational changes for affinity regulation. In both ßI and αI domains, the α1- and α7-helixes work in concert to shift the metal-ion-dependent adhesion site between the resting and active states. An absolutely conserved Gly in the middle of the α1-helix of ßI helps maintain the resting ßI conformation, whereas the homologous position in the αI α1-helix contains a conserved Phe. A functional role of this Phe is structurally unpredictable. Using αLß2 integrin as a model, we found that the residue volume at the Phe position in the α1-helix is critical for αLß2 activation because trimming the Phe by small amino acid substitutions abolished αLß2 binding with soluble and immobilized intercellular cell adhesion molecule 1. Similar results were obtained for αMß2 integrin. Our experimental and molecular dynamics simulation data suggested that the bulky Phe acts as a pawl that stabilizes the downward ratchet-like movement of ß6-α7 loop and α7-helix, required for high-affinity ligand binding. This mechanism may apply to other von Willebrand factor A domains undergoing large conformational changes. We further demonstrated that the conformational cross-talk between αL αI and ß2 ßI could be uncoupled because the ß2 extension and headpiece opening could occur independently of the αI activation. Reciprocally, the αI activation does not inevitably lead to the conformational changes of the ß2 subunit. Such loose linkage between the αI and ßI is attributed to the αI flexibility and could accommodate the αLß2-mediated rolling adhesion of leukocytes.


Assuntos
Antígenos CD18/metabolismo , Integrina alfa1/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Modelos Moleculares , Fenilalanina/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Antígeno CD11b/química , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Antígenos CD18/química , Antígenos CD18/genética , Sequência Conservada , Humanos , Proteínas Imobilizadas , Integrina alfa1/química , Integrina alfa1/genética , Molécula 1 de Adesão Intercelular/química , Cinética , Ligantes , Simulação de Dinâmica Molecular , Mutação , Conformação Proteica , Conformação Proteica em alfa-Hélice , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Solubilidade
8.
J Vis Exp ; (122)2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28447995

RESUMO

Microglia are the primary responders to central nervous system insults; however, much remains unknown about their role in regulating neuroinflammation. Microglia are mesodermal cells that function similarly to macrophages in surveying inflammatory stress. The classical (M1-type) and alternative (M2-type) activations of macrophages have also been extended to microglia in an effort to better understand the underlying interplay these phenotypes have in neuroinflammatory conditions such as Parkinson's, Alzheimer's, and Huntington's Diseases. In vitro experimentation utilizing primary microglia offers rapid and reliable results that may be extended to the in vivo environment. Although this is a clear advantage over in vivo experimentation, isolating microglia while achieving adequate yields of optimal purity has been a challenge. Common methods currently in use either suffer from low recovery, low purity, or both. Herein, we demonstrate a refinement of the column-free CD11b magnetic separation method that achieves a high cell recovery and enhanced purity in half the amount of time. We propose this optimized method as a highly useful model of primary microglial isolation for the purposes of studying neuroinflammation and neurodegeneration.


Assuntos
Antígeno CD11b/química , Separação Celular/métodos , Microglia/fisiologia , Contagem de Células , Humanos , Magnetismo
9.
Transfusion ; 55(12): 2939-48, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26361072

RESUMO

BACKGROUND: HNA-3a antibodies induce severe transfusion-related acute lung injury (TRALI) in which neutrophils play a major role. As neutrophil passage through the pulmonary microvasculature is a critical step in the pathogenesis of TRALI, we investigated the impact of HNA-3a antibodies on two important factors that could impair granulocyte passage through lung capillaries: the elasticity of neutrophils and the expression and activation of adhesion molecules. STUDY DESIGN AND METHODS: The impact of HNA-3a antibodies on the elasticity of neutrophils was investigated using atomic force microscopy (AFM). Neutrophils were settled on poly-2-hydroxyethyl-methacrylate-coated glass slides before treatment with anti-HNA-3a plasma samples, control plasma, or control plasma containing formyl-methionyl-leucyl-phenylalanine (fMLP). Elasticity measurements were carried out in a temperature-controlled perfusion chamber using an atomic force microscopy (AFM) device. The impact of HNA-3a antibodies on the surface expression of total CD11b, activation of CD11b, and L-selectin (CD62L) shedding was investigated by flow cytometry. The functional impact of HNA-3a antibodies on neutrophil adhesion was assessed using fibrinogen-coated plates. RESULTS: HNA-3a antibodies induced stiffening of neutrophils (+24%-40%; p < 0.05) to a similar extent as fMLP. This effect was blocked by treatment of neutrophils with cytochalasin D. While total surface expression of CD11b and L-selectin on neutrophils was largely unaffected, HNA-3a antibodies induced alloantigen-specific activation of CD11b (+72%-107%; p < 0.05) and increased adhesion of neutrophils to fibrinogen. CONCLUSION: Accumulation of neutrophils in the pulmonary microvasculature during severe TRALI is likely mediated by increased rigidity and CD11b-mediated adhesion of neutrophils leading to retention of neutrophils.


Assuntos
Antígeno CD11b/fisiologia , Isoanticorpos/fisiologia , Isoantígenos/imunologia , Selectina L/fisiologia , Neutrófilos/fisiologia , Lesão Pulmonar Aguda/etiologia , Antígeno CD11b/química , Adesão Celular , Humanos , Microscopia de Força Atômica , Conformação Proteica , Reação Transfusional
10.
FEBS Lett ; 589(3): 374-9, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25554420

RESUMO

The interaction of Bordetella pertussis adenylate cyclase toxin (CyaA) with complement receptor 3 (CR3, CD11b/CD18) involves N-linked oligosaccharide chains. To investigate the relative importance of the individual N-glycans of CR3 for toxin activity, the asparagine residues of the consensus N-glycosylation sites of CR3 were substituted with glutamine residues that cannot be glycosylated. Examination of CR3 mutant variants and mass spectrometry analysis of the N-glycosylation pattern of CR3 revealed that N-glycans located in the C-terminal part of the CD11b subunit are involved in binding and cytotoxic activity of CyaA. We suggest that these N-glycans form a defined clustered saccharide patch that enables multivalent contact of CR3 with CyaA, enhancing both affinity and specificity of the integrin-toxin interaction.


Assuntos
Toxina Adenilato Ciclase/metabolismo , Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Antígeno de Macrófago 1/metabolismo , Polissacarídeos/metabolismo , Toxina Adenilato Ciclase/genética , Substituição de Aminoácidos , Asparagina/genética , Bordetella pertussis/metabolismo , Bordetella pertussis/patogenicidade , Antígeno CD11b/química , Antígenos CD18/química , Glutamina/genética , Glicosilação , Humanos , Antígeno de Macrófago 1/genética , Estrutura Terciária de Proteína
11.
J Biol Chem ; 290(1): 142-56, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25371205

RESUMO

The bi-component leukocidins of Staphylococcus aureus are important virulence factors that lyse human phagocytic cells and contribute to immune evasion. The γ-hemolysins (HlgAB and HlgCB) and Panton-Valentine leukocidin (PVL or LukSF) were shown to assemble from soluble subunits into membrane-bound oligomers on the surface of target cells, creating barrel-like pore structures that lead to cell lysis. LukGH is the most distantly related member of this toxin family, sharing only 30-40% amino acid sequence identity with the others. We observed that, unlike other leukocidin subunits, recombinant LukH and LukG had low solubility and were unable to bind to target cells, unless both components were present. Using biolayer interferometry and intrinsic tryptophan fluorescence we detected binding of LukH to LukG in solution with an affinity in the low nanomolar range and dynamic light scattering measurements confirmed formation of a heterodimer. We elucidated the structure of LukGH by x-ray crystallography at 2.8-Šresolution. This revealed an octameric structure that strongly resembles that reported for HlgAB, but with important structural differences. Structure guided mutagenesis studies demonstrated that three salt bridges, not found in other bi-component leukocidins, are essential for dimer formation in solution and receptor binding. We detected weak binding of LukH, but not LukG, to the cellular receptor CD11b by biolayer interferometry, suggesting that in common with other members of this toxin family, the S-component has the primary contact role with the receptor. These new insights provide the basis for novel strategies to counteract this powerful toxin and Staphylococcus aureus pathogenesis.


Assuntos
Proteínas de Bactérias/química , Antígeno CD11b/química , Proteínas Hemolisinas/química , Leucocidinas/química , Staphylococcus aureus/química , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Antígeno CD11b/metabolismo , Cristalografia por Raios X , Células HL-60 , Proteínas Hemolisinas/metabolismo , Humanos , Leucocidinas/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Staphylococcus aureus/metabolismo , Relação Estrutura-Atividade
12.
PLoS One ; 9(1): e85349, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465541

RESUMO

The urokinase plasminogen activator receptor (uPAR) plays a role in tumor progression and has been proposed as a target for the treatment of cancer. We recently described the development of a novel humanized monoclonal antibody that targets uPAR and has anti-tumor activity in multiple xenograft animal tumor models. This antibody, ATN-658, does not inhibit ligand binding (i.e. uPA and vitronectin) to uPAR and its mechanism of action remains unclear. As a first step in understanding the anti-tumor activity of ATN-658, we set out to identify the epitope on uPAR to which ATN-658 binds. Guided by comparisons between primate and human uPAR, epitope mapping studies were performed using several orthogonal techniques. Systematic site directed and alanine scanning mutagenesis identified the region of aa 268-275 of uPAR as the epitope for ATN-658. No known function has previously been attributed to this epitope Structural insights into epitope recognition were obtained from structural studies of the Fab fragment of ATN-658 bound to uPAR. The structure shows that the ATN-658 binds to the DIII domain of uPAR, close to the C-terminus of the receptor, corroborating the epitope mapping results. Intriguingly, when bound to uPAR, the complementarity determining region (CDR) regions of ATN-658 closely mimic the binding regions of the integrin CD11b (αM), a previously identified uPAR ligand thought to be involved in leukocyte rolling, migration and complement fixation with no known role in tumor progression of solid tumors. These studies reveal a new functional epitope on uPAR involved in tumor progression and demonstrate a previously unrecognized strategy for the therapeutic targeting of uPAR.


Assuntos
Anticorpos Monoclonais/química , Antígeno CD11b/química , Epitopos/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Antígeno CD11b/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Cães , Drosophila melanogaster , Mapeamento de Epitopos , Epitopos/imunologia , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica/efeitos dos fármacos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Receptores de Ativador de Plasminogênio Tipo Uroquinase/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Homologia de Sequência de Aminoácidos , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/imunologia , Vitronectina/química , Vitronectina/imunologia
13.
Proc Natl Acad Sci U S A ; 110(26): 10794-9, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23754403

RESUMO

Staphylococcus aureus causes diseases ranging from superficial wound infections to more invasive manifestations like osteomyelitis and endocarditis. The evasion of host phagocytes recruited to the site of infection is essential to the success of S. aureus as a pathogen. A single S. aureus strain can produce up to five different bicomponent pore-forming leukotoxins that lyse immune cells by forming pores in the cellular plasma membrane. Although these leukotoxins have been considered redundant due to their cytotoxic activity toward human neutrophils, each toxin displays varied species and cell-type specificities. This suggests that cellular factors may influence which cells each toxin targets. Here we describe the identification of CD11b, the α subunit of the αM/ß2 integrin (CD11b/CD18), macrophage-1 antigen, or complement receptor 3, as a cellular receptor for leukocidin A/B (LukAB), an important toxin that contributes to S. aureus killing of human neutrophils. We demonstrate that CD11b renders human neutrophils susceptible to LukAB-mediated killing by purified LukAB as well as during S. aureus infection ex vivo. LukAB directly interacts with human CD11b by binding to the I domain, a property that determines the species specificity exhibited by this toxin. Identification of a LukAB cellular target has broad implications for the use of animal models to study the role of LukAB in S. aureus pathogenesis, explains the toxin's tropism toward human neutrophils and other phagocytes, and provides a cellular therapeutic target to block the effect of LukAB toward human neutrophils.


Assuntos
Toxinas Bacterianas/imunologia , Toxinas Bacterianas/toxicidade , Antígeno CD11b/metabolismo , Leucocidinas/imunologia , Leucocidinas/toxicidade , Neutrófilos/imunologia , Neutrófilos/microbiologia , Staphylococcus aureus/patogenicidade , Antígeno CD11b/química , Células HEK293 , Células HL-60 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Antígeno de Macrófago 1/metabolismo , Neutrófilos/patologia , Fagocitose/imunologia , Estrutura Terciária de Proteína , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Virulência/imunologia
14.
PLoS One ; 8(2): e57951, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469114

RESUMO

One-half of the integrin α-subunit Propeller domains contain and extra vWFA domain (αA domain), which mediates integrin binding to extracellular physiologic ligands via its metal-ion-dependent adhesion site (MIDAS). We used electron microscopy to determine the 3D structure of the αA-containing ectodomain of the leukocyte integrin CD11b/CD18 (αMß2) in its inactive state. A well defined density for αA was observed within a bent ectodomain conformation, while the structure of the ectodomain in complex with the Fab fragment of mAb107, which binds at the MIDAS face of CD11b and stabilizes the inactive state, further revealed that αA is restricted to a relatively small range of orientations relative to the Propeller domain. Using Fab 107 as probe in fluorescent lifetime imaging microscopy (FLIM) revealed that αA is positioned relatively far from the membrane surface in the inactive state, and a systematic orientation search revealed that the MIDAS face would be accessible to extracellular ligand in the inactive state of the full-length cellular integrin. These studies are the first to define the 3D EM structure of an αA-containing integrin ectodomain and to position the ligand-binding face of αA domain in relation to the plasma membrane, providing new insights into current models of integrin activation.


Assuntos
Antígeno CD11b/química , Antígeno CD11b/metabolismo , Antígenos CD18/química , Antígenos CD18/metabolismo , Membrana Celular/metabolismo , Microscopia Eletrônica , Anticorpos Monoclonais/química , Sítios de Ligação , Humanos , Imageamento Tridimensional , Fragmentos Fab das Imunoglobulinas/metabolismo , Ligantes , Metais/metabolismo , Modelos Moleculares , Estrutura Terciária de Proteína
15.
J Vis Exp ; (63): e4040, 2012 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-22644046

RESUMO

Within the intestine reside unique populations of innate and adaptive immune cells that are involved in promoting tolerance towards commensal flora and food antigens while concomitantly remaining poised to mount inflammatory responses toward invasive pathogens. Antigen presenting cells, particularly DCs and macrophages, play critical roles in maintaining intestinal immune homeostasis via their ability to sense and appropriately respond to the microbiota. Efficient isolation of intestinal DCs and macrophages is a critical step in characterizing the phenotype and function of these cells. While many effective methods of isolating intestinal immune cells, including DCs and macrophages, have been described, many rely upon long digestions times that may negatively influence cell surface antigen expression, cell viability, and/or cell yield. Here, we detail a methodology for the rapid isolation of large numbers of viable, intestinal DCs and macrophages. Phenotypic characterization of intestinal DCs and macrophages is carried out by directly staining isolated intestinal cells with specific fluorescence-labeled monoclonal antibodies for multi-color flow cytometric analysis. Furthermore, highly pure DC and macrophage populations are isolated for functional studies utilizing CD11c and CD11b magnetic-activated cell sorting beads followed by cell sorting.


Assuntos
Separação Celular/métodos , Células Dendríticas/citologia , Intestino Delgado/citologia , Macrófagos/citologia , Animais , Antígeno CD11b/química , Antígeno CD11c/química , Citometria de Fluxo , Magnetismo , Camundongos
16.
Ann Rheum Dis ; 71(12): 2028-34, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22586164

RESUMO

OBJECTIVES: The rs1143679 variant of ITGAM, encoding the R77H variant of CD11b (part of complement receptor 3; CR3), is among the strongest genetic susceptibility effects in human systemic lupus erythematosus (SLE). The authors aimed to demonstrate R77H function in ex-vivo human cells. METHODS: Monocytes/monocyte-derived macrophages from healthy volunteers homozygous for either wild type (WT) or 77H CD11b were studied. The genotype-specific expression of CD11b, and CD11b activation using conformation-specific antibodies were measured. Genotype-specific differences in iC3b-mediated phagocytosis, adhesion to a range of ligands and the secretion of cytokines following CR3 ligation were studied. The functionality of R77H was confirmed by replicating findings in COS7 cells expressing variant-specific CD11b. RESULTS: No genotype-specific difference in CD11b expression or in the expression of CD11b activation epitopes was observed. A 31% reduction was observed in the phagocytosis of iC3b opsonised sheep erythrocytes (sRBC(iC3b)) by 77H cells (p=0.003) and reduced adhesion to a range of ligands: notably a 24% reduction in adhesion to iC3b (p=0.014). In transfected COS7 cells, a 42% reduction was observed in phagocytosis by CD11b (77H)-expressing cells (p=0.004). A significant inhibition was seen in the release of Toll-like receptor 7/8-induced pro-inflammatory cytokines from WT monocytes when CR3 was pre-engaged using sRBC(iC3b), but no inhibition in 77H monocytes resulting in a significant difference between genotypes (interleukin (IL)-1ß p=0.030; IL-6 p=0.029; tumour necrosis factor alpha p=0.027). CONCLUSIONS: The R77H variant impairs a broad range of CR3 effector functions in human monocytes. This study discusses how perturbation of this pathway may predispose to SLE.


Assuntos
Antígeno CD11b/genética , Antígeno CD11b/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Antígeno de Macrófago 1/imunologia , Monócitos/imunologia , Adulto , Animais , Antígeno CD11b/química , Células COS , Adesão Celular/imunologia , Chlorocebus aethiops , Citocinas/metabolismo , Fibroblastos/citologia , Expressão Gênica/imunologia , Predisposição Genética para Doença/genética , Variação Genética , Genótipo , Homozigoto , Humanos , Antígeno de Macrófago 1/química , Macrófagos/citologia , Macrófagos/imunologia , Monócitos/citologia , Fagocitose/imunologia , Conformação Proteica , Estrutura Terciária de Proteína
17.
Macromol Biosci ; 12(5): 591-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22411735

RESUMO

The efficiency of the treatments involving CNS disorders is commonly diminished by the toxicity, reduced stability and lack of targeting of the administered neuroactive compounds. In this study, we have successfully multifunctionalized CMCht/PAMAM dendrimer nanoparticles by coupling the CD11b antibody and loading MP into the nanoparticles. The modification of the new antibody-conjugated nanoparticles was confirmed by S-TEM observation and (1) H NMR and FTIR spectroscopy. Cytotoxicity assays revealed that the conjugates did not affect the viability of both primary cultures of glial and microglial cells. Trace analyses of FITC-labelled nanoparticles revealed that the uptake of antibody-conjugated nanoparticles was conserved in microglial cells but significantly decreased in astrocytes and oligodendrocytes. Thus, this study demonstrates that antibody conjugation contributes to a modulation of the internalization of these nanocarriers by different cell types, which might be of relevance for specific targeting of CNS cell populations.


Assuntos
Astrócitos/efeitos dos fármacos , Quitosana/análogos & derivados , Dendrímeros/química , Portadores de Fármacos/farmacologia , Nanopartículas/química , Oligodendroglia/efeitos dos fármacos , Animais , Anticorpos/química , Anticorpos/imunologia , Astrócitos/citologia , Astrócitos/metabolismo , Antígeno CD11b/química , Antígeno CD11b/imunologia , Sobrevivência Celular/efeitos dos fármacos , Quitosana/química , Portadores de Fármacos/síntese química , Composição de Medicamentos , Endocitose/efeitos dos fármacos , Fluoresceína-5-Isotiocianato , Espectroscopia de Ressonância Magnética , Metilprednisolona/química , Metilprednisolona/farmacologia , Microscopia Eletrônica de Transmissão , Nanopartículas/uso terapêutico , Nanopartículas/ultraestrutura , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Especificidade de Órgãos , Cultura Primária de Células , Ratos , Espectroscopia de Infravermelho com Transformada de Fourier
18.
J Immunol ; 187(12): 6393-401, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22095715

RESUMO

A central feature of integrin interaction with physiologic ligands is the monodentate binding of a ligand carboxylate to a Mg(2+) ion hexacoordinated at the metal ion-dependent adhesion site (MIDAS) in the integrin A domain. This interaction stabilizes the A domain in the high-affinity state, which is distinguished from the default low-affinity state by tertiary changes in the domain that culminate in cell adhesion. Small molecule ligand-mimetic integrin antagonists act as partial agonists, eliciting similar activating conformational changes in the A domain, which has contributed to paradoxical adhesion and increased patient mortality in large clinical trials. As with other ligand-mimetic integrin antagonists, the function-blocking mAb 107 binds MIDAS of integrin CD11b/CD18 A domain (CD11bA), but in contrast, it favors the inhibitory Ca(2+) ion over the Mg(2+) ion at MIDAS. We determined the crystal structures of the Fab fragment of mAb 107 complexed to the low- and high-affinity states of CD11bA. Favored binding of the Ca(2+) ion at MIDAS is caused by the unusual symmetric bidentate ligation of a Fab-derived ligand Asp to a heptacoordinated MIDAS Ca(2+) ion. Binding of the Fab fragment of mAb 107 to CD11bA did not trigger the activating tertiary changes in the domain or in the full-length integrin. These data show that the denticity of the ligand Asp/Glu can modify the divalent cation selectivity at MIDAS and hence integrin function. Stabilizing the Ca(2+) ion at MIDAS by bidentate ligation to a ligand Asp/Glu may provide one approach for designing pure integrin antagonists.


Assuntos
Anticorpos Bloqueadores/metabolismo , Ácido Aspártico/imunologia , Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Cálcio/metabolismo , Animais , Anticorpos Bloqueadores/química , Ácido Aspártico/metabolismo , Sítios de Ligação de Anticorpos/imunologia , Antígeno CD11b/química , Antígenos CD18/química , Cátions Bivalentes/metabolismo , Adesão Celular/imunologia , Cristalografia por Raios X , Relação Dose-Resposta Imunológica , Desenho de Fármacos , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/metabolismo , Células K562 , Ligantes , Neutrófilos/imunologia , Neutrófilos/metabolismo , Ratos
19.
Blood ; 117(18): 4885-94, 2011 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-21403131

RESUMO

The ß(2)-integrin CD11b/CD18 mediates the firm adhesion of neutrophils (PMNs) to epithelial monolayers, a key step in PMN transepithelial migration. To complete the transmigration process, adherent PMNs must detach from epithelial monolayer surfaces to move forward. The mechanism that governs the detachment of adherent PMNs, however, is not clear. Here, we present evidence that cleavage of the CD11b extracellular domain containing the ligand-binding I-domain by 3 structural and functional related serine proteases (elastase, proteinase-3 and cathepsin G) serves as a novel mechanism for PMN detachment after the initial cell adhesion. Kinetic studies showed that the cleavage of CD11b is positively correlated with PMN detachment and subsequent transmigration. Moreover, the results demonstrated that elastase, proteinase-3 and cathepsin G all cleaved the purified, functionally active form of CD11b in a pattern similar to the CD11b shedding that occurs during PMN transmigration. Their cleavage sites on purified CD11b were located at (761)Thr-Ala(762) (elastase/proteinase-3) and (760)Phe-Thr(761) (cathepsin G), respectively. CD11b cleavage and PMN detachment and chemotaxis, were impaired in elastase/cathepsin G-deficient Beige mice; this defect could be restored by the addition of extracellular elastase. By illustrating CD11b shedding by elastase, proteinase-3 and cathepsin G as a novel mechanism for PMN detachment, our study provides novel therapeutic targets for controlling inflammation.


Assuntos
Antígeno CD11b/metabolismo , Quimiotaxia de Leucócito/fisiologia , Neutrófilos/fisiologia , Serina Proteases/metabolismo , Animais , Sítios de Ligação , Antígeno CD11b/química , Antígenos CD18/metabolismo , Catepsina G/metabolismo , Adesão Celular/fisiologia , Humanos , Técnicas In Vitro , Elastase de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mieloblastina/metabolismo , Estrutura Terciária de Proteína
20.
Contrast Media Mol Imaging ; 5(5): 268-75, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20973112

RESUMO

Imaging of macrophages with superparamagnetic iron oxide particles (SPIO) has been performed to improve detection of atherosclerotic plaque inflammation in human and mouse studies by molecular magnetic resonance imaging (MRI). Since affinity of the monocyte/macrophage integrin MAC-1 (CD11b/CD18) is upregulated in inflammation, we generated a contrast agent targeting CD11b (CD11b-SPIOs) for improved macrophage detection in plaques. CD11b-SPIOs and non-targeted SPIOs (control-SPIOs) were incubated in vitro with human monocytes/macrophages. As quantified by SPIO-induced MRI signal extinction, intracellular iron-content was significantly higher in monoytes/macrophages incubated with CD11b-SPIO than with control-SPIO in vitro (p < 0.05), suggesting an improved uptake of CD11b-SPIOs into monocytes. Therefore, the aortic arch (AA) and vessel branches of ApoE(-/-)-knockout mice on a Western-type diet were imaged before and 48 h after contrast agent injection of either CD11b-SPIOs or control-SPIOs, using a 9.4 T animal MRI system. The SPIO-induced change in the MRI signal was quantified, as well as the macrophage-content by anti-CD68 immunhistochemistry and the iron-content by Prussian-blue staining. However, SPIO-induced signal extinction in in vivo-MRI was similar in CD11b-SPIO and control-SPIO-injected animals, with a non-significant trend towards an improved uptake of CD11b-SPIOs in the subclavian artery and subsections of the AA. These data correlated well with the results obtained by histology. Although in vitro MRI-data indicated an increased uptake of targeted CD11b-SPIOs in monocytes/macrophages, in vivo mouse data do not allow improved atherosclerotic plaque detection compared WITH non-targeted SPIOs. Therefore, CD11b-targeted MRI contrast labelling of monocytes/macrophages does not seem to be a successful strategy in stable atherosclerotic plaques such as found in the ApoE(-/-)-knockout-model. However, the impressive correlation between MRI and histology data encourages further development of inflammation- and plaque-specific contrast agents for vulnerable plaque imaging.


Assuntos
Antígeno CD11b/química , Compostos Férricos/química , Imageamento por Ressonância Magnética/métodos , Monócitos/citologia , Monócitos/metabolismo , Nanopartículas/química , Placa Aterosclerótica/diagnóstico , Animais , Apolipoproteínas E/genética , Células Cultivadas , Meios de Contraste/química , Humanos , Camundongos , Camundongos Knockout , Placa Aterosclerótica/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...