Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mBio ; 12(5): e0240821, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34634939

RESUMO

Complement-opsonized HIV-1 triggers efficient antiviral type I interferon (IFN) responses in dendritic cells (DCs), which play an important role in protective responses at the earliest stages in retroviral infection. In contrast, HIV-1 suppresses or escapes sensing by STING- and MAVS-associated sensors. Here, we identified a complement receptor-mediated sensing pathway, where DCs are activated in CCR5/RLR (RIG-I/MDA5)/MAVS/TBK1-dependent fashion. Increased fusion of complement-opsonized HIV-1 via complement receptor 4 and CCR5 leads to increased incoming HIV-1 RNA in the cytoplasm, sensed by a nonredundant cooperative effect of RIG-I and MDA5. Moreover, complement-opsonized HIV-1 down-modulated the MAVS-suppressive Raf-1/PLK1 pathway, thereby opening the antiviral recognition pathway via MAVS. This in turn was followed by MAVS aggregation and subsequent TBK1/IRF3/NF-κB activation in DCs exposed to complement- but not non-opsonized HIV-1. Our data strongly suggest that complement is important in the induction of efficient antiviral immune responses by preventing HIV-1 suppressive mechanisms as well as inducing specific cytosolic sensors. IMPORTANCE Importantly, our study highlights an unusual target on DCs-the α chain of complement receptor 4 (CR4) (CD11c)-for therapeutic interventions in HIV-1 treatment. Targeting CD11c on DCs mediated a potent antiviral immune response via clustering of CR4 and CCR5 and subsequent opening of an antiviral recognition pathway in DCs via MAVS. This novel finding might provide novel tools for specifically boosting endogenous antiviral immunity via CR4, abundantly expressed on multiple DC subsets.


Assuntos
Proteínas do Sistema Complemento/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Interferon Tipo I/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/genética , Humanos , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/imunologia , Interferon Tipo I/genética , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/imunologia , Receptores CCR5/genética , Receptores CCR5/imunologia
2.
Nat Commun ; 12(1): 4230, 2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-34244494

RESUMO

Extracellular matrix protein-1 (ECM1) promotes tumorigenesis in multiple organs but the mechanisms associated to ECM1 isoform subtypes have yet to be clarified. We report in this study that the secretory ECM1a isoform induces tumorigenesis through the GPR motif binding to integrin αXß2 and the activation of AKT/FAK/Rho/cytoskeleton signaling. The ATP binding cassette subfamily G member 1 (ABCG1) transduces the ECM1a-integrin αXß2 interactive signaling to facilitate the phosphorylation of AKT/FAK/Rho/cytoskeletal molecules and to confer cancer cell cisplatin resistance through up-regulation of the CD326-mediated cell stemness. On the contrary, the non-secretory ECM1b isoform binds myosin and blocks its phosphorylation, impairing cytoskeleton-mediated signaling and tumorigenesis. Moreover, ECM1a induces the expression of the heterogeneous nuclear ribonucleoprotein L like (hnRNPLL) protein to favor the alternative mRNA splicing generating ECM1a. ECM1a, αXß2, ABCG1 and hnRNPLL higher expression associates with poor survival, while ECM1b higher expression associates with good survival. These results highlight ECM1a, integrin αXß2, hnRNPLL and ABCG1 as potential targets for treating cancers associated with ECM1-activated signaling.


Assuntos
Processamento Alternativo , Carcinoma Epitelial do Ovário/genética , Proteínas da Matriz Extracelular/metabolismo , Recidiva Local de Neoplasia/epidemiologia , Neoplasias Ovarianas/genética , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Animais , Carcinoma Epitelial do Ovário/mortalidade , Carcinoma Epitelial do Ovário/patologia , Carcinoma Epitelial do Ovário/terapia , Linhagem Celular Tumoral , Quimioterapia Adjuvante , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Intervalo Livre de Doença , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas da Matriz Extracelular/genética , Feminino , Seguimentos , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/metabolismo , Estimativa de Kaplan-Meier , Camundongos , Pessoa de Meia-Idade , Mutação , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/patologia , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Ovário/patologia , Ovário/cirurgia , Fosforilação/genética , Prognóstico , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA-Seq , Transdução de Sinais/genética , Ensaios Antitumorais Modelo de Xenoenxerto
3.
BJOG ; 128(8): 1282-1291, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33539617

RESUMO

OBJECTIVE: To study genetic variants and their function within genes coding for complement receptors in pre-eclampsia. DESIGN: A case-control study. SETTING: Pre-eclampsia is a common vascular disease of pregnancy. The clearance of placenta-derived material is one of the functions of the complement system in pregnancy. POPULATION: We genotyped 500 women with pre-eclamptic pregnancies and 190 pregnant women without pre-eclampsia, as controls, from the FINNPEC cohort, and 122 women with pre-eclamptic pregnancies and 1905 controls from the national FINRISK cohort. METHODS: The functional consequences of genotypes discovered by targeted exomic sequencing were explored by analysing the binding of the main ligand iC3b to mutated CR3 or CR4, which were transiently expressed on the surface of COS-1 cells. MAIN OUTCOME MEASURES: Allele frequencies were compared between pre-eclamptic pregnancies and controls in genetic studies. The functional consequences of selected variants were measured by binding assays. RESULTS: The most significantly pre-eclampsia-linked CR3 variant M441K (P = 4.27E-4, OR = 1.401, 95% CI = 1.167-1.682) displayed a trend of increased adhesion to iC3b (P = 0.051). The CR4 variant A251T was found to enhance the adhesion of CR4 to iC3b, whereas W48R resulted in a decrease of the binding of CR4 to iC3b. CONCLUSIONS: Results suggest that changes in complement-facilitated phagocytosis are associated with pre-eclampsia. Further studies are needed to ascertain whether aberrant CR3 and CR4 activity leads to altered pro- and anti-inflammatory cytokine responses in individuals carrying the associated variants, and the role of these receptors in pre-eclampsia pathogenesis. TWEETABLE ABSTRACT: Genetic variants of complement receptors CR3 and CR4 have functional consequences that are associated with pre-eclampsia.


Assuntos
Antígeno CD11b/genética , Integrina alfaXbeta2/genética , Antígeno de Macrófago 1/genética , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/imunologia , Antígenos CD18/metabolismo , Citocinas/biossíntese , Feminino , Genótipo , Humanos , Integrina alfaXbeta2/metabolismo , Antígeno de Macrófago 1/metabolismo , Mutação , Fagocitose , Gravidez
4.
Front Immunol ; 11: 2010, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32922405

RESUMO

Dendritic cells (DCs) possess intrinsic cellular defense mechanisms to specifically inhibit HIV-1 replication. In turn, HIV-1 has evolved strategies to evade innate immune sensing by DCs resulting in suboptimal maturation and poor antiviral immune responses. We previously showed that complement-opsonized HIV-1 (HIV-C) was able to efficiently infect various DC subsets significantly higher than non-opsonized HIV-1 (HIV) and therefore also mediate a higher antiviral immunity. Thus, complement coating of HIV-1 might play a role with respect to viral control occurring early during infection via modulation of DCs. To determine in detail which complement receptors (CRs) expressed on DCs was responsible for infection and superior pro-inflammatory and antiviral effects, we generated stable deletion mutants for the α-chains of CR3, CD11b, and CR4, CD11c using CRISPR/Cas9 in THP1-derived DCs. We found that CD11c deletion resulted in impaired DC infection as well as antiviral and pro-inflammatory immunity upon exposure to complement-coated HIV-1. In contrast, sole expression of CD11b on DCs shifted the cells to an anti-inflammatory, regulatory DC type. We here illustrated that CR4 comprised of CD11c and CD18 is the major player with respect to DC infection associated with a potent early pro-inflammatory immune response. A more detailed characterization of CR3 and CR4 functions using our powerful tool might open novel avenues for early therapeutic intervention during HIV-1 infection.


Assuntos
Células Dendríticas/imunologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Integrina alfaXbeta2/metabolismo , Antígeno de Macrófago 1/metabolismo , Antígeno CD11b/genética , Antígeno CD11c/genética , Antígenos CD18/genética , Sistemas CRISPR-Cas , Proteínas do Sistema Complemento/metabolismo , Humanos , Imunidade , Integrina alfaXbeta2/genética , Antígeno de Macrófago 1/genética , Deleção de Sequência/genética , Transdução de Sinais , Células THP-1
5.
Infect Immun ; 88(9)2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32571987

RESUMO

Even though both cellular and humoral immunities contribute to host defense, the role played by humoral immunity against the airborne opportunistic fungal pathogen Aspergillus fumigatus has been underexplored. In this study, we aimed at deciphering the role of the complement system, the major humoral immune component, against A. fumigatus Mass spectrometry analysis of the proteins extracted from A. fumigatus conidial (asexual spores and infective propagules) surfaces opsonized with human serum indicated that C3 is the major complement protein involved. Flow cytometry and immunolabeling assays further confirmed C3b (activated C3) deposition on the conidial surfaces. Assays using cell wall components of conidia indicated that the hydrophobin RodAp, ß-(1,3)-glucan (BG) and galactomannan (GM) could efficiently activate C3. Using complement component-depleted sera, we showed that while RodAp activates C3 by the alternative pathway, BG and GM partially follow the classical and lectin pathways, respectively. Opsonization facilitated conidial aggregation and phagocytosis, and complement receptor (CR3 and CR4) blockage on phagocytes significantly inhibited phagocytosis, indicating that the complement system exerts a protective role against conidia by opsonizing them and facilitating their phagocytosis mainly through complement receptors. Conidial opsonization with human bronchoalveolar lavage fluid (BALF) confirmed C3 to be the major complement protein interacting with conidia. Nevertheless, complement C2 and mannose-binding lectin (MBL), the classical and lectin pathway components, respectively, were not identified, indicating that BALF activates the alternative pathway on the conidial surface. Moreover, the cytokine profiles were different upon stimulation of phagocytes with serum- and BALF-opsonized conidia, highlighting the importance of studying interaction of conidia with complement proteins in their biological niche.


Assuntos
Aspergillus fumigatus/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Complemento C3/imunologia , Polissacarídeos Fúngicos/farmacologia , Macrófagos/efeitos dos fármacos , Soro/imunologia , Esporos Fúngicos/imunologia , Aspergilose/genética , Aspergilose/imunologia , Aspergilose/microbiologia , Aspergillus fumigatus/química , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/microbiologia , Parede Celular/química , Parede Celular/imunologia , Ativação do Complemento/efeitos dos fármacos , Complemento C3/genética , Citocinas/biossíntese , Citocinas/imunologia , Polissacarídeos Fúngicos/imunologia , Polissacarídeos Fúngicos/isolamento & purificação , Galactose/análogos & derivados , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Imunidade Celular , Imunidade Humoral , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/imunologia , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Mananas/imunologia , Mananas/isolamento & purificação , Mananas/farmacologia , Proteínas Opsonizantes/farmacologia , Fagocitose/efeitos dos fármacos , Cultura Primária de Células , Ligação Proteica , Espécies Reativas de Oxigênio , Soro/química , Soro/microbiologia , Esporos Fúngicos/química , beta-Glucanas/imunologia , beta-Glucanas/isolamento & purificação , beta-Glucanas/farmacologia
6.
J Immunol ; 204(5): 1345-1361, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31969389

RESUMO

Aggregation of α-synuclein (αSN) is an important histological feature of Parkinson disease. Recent studies showed that the release of misfolded αSN from human and rodent neurons is relevant to the progression and spread of αSN pathology. Little is known, however, about the mechanisms responsible for clearance of extracellular αSN. This study found that human complement receptor (CR) 4 selectively bound fibrillar αSN, but not monomeric species. αSN is an abundant protein in the CNS, which potentially could overwhelm clearance of cytotoxic αSN species. The selectivity of CR4 toward binding fibrillar αSN consequently adds an important αSN receptor function for maintenance of brain homeostasis. Based on the recently solved structures of αSN fibrils and the known ligand preference of CR4, we hypothesize that the parallel monomer stacking in fibrillar αSN creates a known danger-associated molecular pattern of stretches of anionic side chains strongly bound by CR4. Conformational change in the receptor regulated tightly clearance of fibrillar αSN by human monocytes. The induced change coupled concomitantly with phagolysosome formation. Data mining of the brain transcriptome in Parkinson disease patients supported CR4 as an active αSN clearance mechanism in this disease. Our results associate an important part of the innate immune system, namely complement receptors, with the central molecular mechanisms of CNS protein aggregation in neurodegenerative disorders.


Assuntos
Integrina alfaXbeta2 , Macrófagos , Doença de Parkinson , Fagossomos , Agregação Patológica de Proteínas , alfa-Sinucleína , Humanos , Integrina alfaXbeta2/química , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Doença de Parkinson/genética , Doença de Parkinson/imunologia , Doença de Parkinson/patologia , Fagossomos/química , Fagossomos/genética , Fagossomos/imunologia , Fagossomos/patologia , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/imunologia , Agregação Patológica de Proteínas/patologia , Estrutura Quaternária de Proteína , alfa-Sinucleína/química , alfa-Sinucleína/genética , alfa-Sinucleína/imunologia
7.
Sci Rep ; 7(1): 4050, 2017 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-28642550

RESUMO

Complement Receptor Immunoglobulin (CRIg), selectively expressed by macrophages, plays an important role in innate immunity by promoting phagocytosis of bacteria. Thus modulation of CRIg on macrophages by cytokines can be an important mechanism by which cytokines regulate anti-microbial immunity. The effects of the cytokines, tumor necrosis factor, transforming growth factor-ß1, interferon-γ, interleukin (IL)-4, IL-13, IL-10, IL-1ß, IL-6, lymphotoxin-α, macrophage-colony stimulating factor (M-CSF) and GM-CSF on CRIg expression were examined in human macrophages. We demonstrated that cytokines regulated the CRIg expression on macrophages during their development from monocytes in culture at the transcriptional level using qPCR and protein by Western blotting. Both CRIg spliced forms (Long and Short), were similarly regulated by cytokines. Direct addition of cytokines to matured CRIg+ macrophages also changed CRIg mRNA expression, suggesting that cytokines control macrophage function via CRIg, at two checkpoints. Interestingly the classical complement receptors, CR3 and CR4 were differentially regulated by cytokines. The changes in CRIg but not CR3/CR4 mRNA expression correlated with ability to phagocytose Candida albicans by macrophages. These findings suggest that CRIg is likely to be a control point in infection and immunity through which cytokines can mediate their effects, and is differentially regulated from CR3 and CR4 by cytokines.


Assuntos
Candida albicans/imunologia , Citocinas/metabolismo , Expressão Gênica , Macrófagos/imunologia , Macrófagos/microbiologia , Fagocitose/imunologia , Receptores de Complemento/genética , Receptores de Complemento/imunologia , Candidíase/etiologia , Candidíase/metabolismo , Citocinas/farmacologia , Humanos , Mediadores da Inflamação , Integrina alfaXbeta2/genética , Antígeno de Macrófago 1/genética , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Fagocitose/efeitos dos fármacos
8.
Mol Cells ; 40(5): 355-362, 2017 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-28535664

RESUMO

The ß2 integrins are cell surface transmembrane proteins regulating leukocyte functions, such as adhesion and migration. Two members of ß2 integrin, αMß2 and αXß2, share the leukocyte distribution profile and integrin αXß2 is involved in antigen presentation in dendritic cells and transendothelial migration of monocytes and macrophages to atherosclerotic lesions. Receptor for advanced glycation end products (RAGE), a member of cell adhesion molecules, plays an important role in chronic inflammation and atherosclerosis. Although RAGE and αXß2 play an important role in inflammatory response and the pathogenesis of atherosclerosis, the nature of their interaction and structure involved in the binding remain poorly defined. In this study, using I-domain as a ligand binding motif of αXß2, we characterize the binding nature and the interacting moieties of αX I-domain and RAGE. Their binding requires divalent cations (Mg2+ and Mn2+) and shows an affinity on the sub-micro molar level: the dissociation constant of αX I-domains binding to RAGE being 0.49 µM. Furthermore, the αX I-domains recognize the V-domain, but not the C1 and C2-domains of RAGE. The acidic amino acid substitutions on the ligand binding site of αX I-domain significantly reduce the I-domain binding activity to soluble RAGE and the alanine substitutions of basic amino acids on the flat surface of the V-domain prevent the V-domain binding to αX I-domain. In conclusion, the main mechanism of αX I-domain binding to RAGE is a charge interaction, in which the acidic moieties of αX I-domains, including E244, and D249, recognize the basic residues on the RAGE V-domain encompassing K39, K43, K44, R104, and K107.


Assuntos
Integrina alfaXbeta2/química , Integrina alfaXbeta2/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Alanina/genética , Alanina/metabolismo , Substituição de Aminoácidos , Sítios de Ligação , Humanos , Integrina alfaXbeta2/genética , Cinética , Mutação , Domínios Proteicos , Receptor para Produtos Finais de Glicação Avançada/química , Receptor para Produtos Finais de Glicação Avançada/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ressonância de Plasmônio de Superfície
9.
Proc Natl Acad Sci U S A ; 114(13): 3403-3408, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28292891

RESUMO

Recognition by the leukocyte integrins αXß2 and αMß2 of complement iC3b-opsonized targets is essential for effector functions including phagocytosis. The integrin-binding sites on iC3b remain incompletely characterized. Here, we describe negative-stain electron microscopy and biochemical studies of αXß2 and αMß2 in complex with iC3b. Despite high homology, the two integrins bind iC3b at multiple distinct sites. αXß2 uses the αX αI domain to bind iC3b on its C3c moiety at one of two sites: a major site at the interface between macroglobulin (MG) 3 and MG4 domains, and a less frequently used site near the C345C domain. In contrast, αMß2 uses its αI domain to bind iC3b at the thioester domain and simultaneously interacts through a region near the αM ß-propeller and ß2 ßI domain with a region of the C3c moiety near the C345C domain. Remarkably, there is no overlap between the primary binding site of αXß2 and the binding site of αMß2 on iC3b. Distinctive binding sites on iC3b by integrins αXß2 and αMß2 may be biologically beneficial for leukocytes to more efficiently capture opsonized pathogens and to avoid subversion by pathogen factors.


Assuntos
Complemento C3b/metabolismo , Integrina alfaXbeta2/metabolismo , Antígeno de Macrófago 1/metabolismo , Sítios de Ligação , Complemento C3b/química , Complemento C3b/genética , Humanos , Integrina alfaXbeta2/química , Integrina alfaXbeta2/genética , Leucócitos/química , Leucócitos/metabolismo , Antígeno de Macrófago 1/química , Antígeno de Macrófago 1/genética , Ligação Proteica , Domínios Proteicos , Estrutura Terciária de Proteína
10.
Sci Rep ; 6: 26966, 2016 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-27231021

RESUMO

The Gram-positive bacterium Streptococcus suis serotype 2 (S. suis 2), an important zoonotic pathogen, induces strong systemic infections in humans; sepsis and meningitis are the most common clinical manifestations and are often accompanied by bacteremia. However, the mechanisms of S. suis 2 survival in human blood are not well understood. In our previous study, we identified muramidase-released protein (MRP), a novel human fibrinogen (hFg)-binding protein (FBP) in S. suis 2 that is an important epidemic infection marker with an unknown mechanism in pathogenesis. The present study demonstrates that the N-terminus of MRP (a.a. 283-721) binds to both the Aα and Bß chains of the D fragment of hFg. Strikingly, the hFg-MRP interaction improved the survival of S. suis 2 in human blood and led to the aggregation and exhaustion of polymorphonuclear neutrophils (PMNs) via an αXß2 integrin-dependent mechanism. Other Fg-binding proteins, such as M1 (GAS) and FOG (GGS), also induced PMNs aggregation; however, the mechanisms of these FBP-hFg complexes in the evasion of PMN-mediated innate immunity remain unclear. MRP is conserved across highly virulent strains in Europe and Asia, and these data shed new light on the function of MRP in S. suis pathogenesis.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Fibrinogênio/metabolismo , Integrina alfaXbeta2/genética , Streptococcus suis/genética , Fatores de Virulência/metabolismo , Animais , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Antígenos de Bactérias/toxicidade , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/toxicidade , Sítios de Ligação , Agregação Celular/efeitos dos fármacos , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Fibrinogênio/química , Fibrinogênio/genética , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Integrina alfaXbeta2/imunologia , Viabilidade Microbiana , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/microbiologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Streptococcus suis/isolamento & purificação , Streptococcus suis/metabolismo , Streptococcus suis/patogenicidade , Suínos , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/toxicidade
11.
Mol Biol (Mosk) ; 49(5): 811-6, 2015.
Artigo em Russo | MEDLINE | ID: mdl-26510599

RESUMO

The third component of complement, C3, plays a central role in human innate immunity. The subsequent proteolysis product of native C3, iC3b, is the primary ligand of complement receptors (CRs) CR3 and CR4. CR3 and CR4 are ß2-family integrins, and their binding to iC3b contributes to phagocytosis. How iC3b binds to its receptors and transmits signals into the cells is not clear. To perform structural and functional studies on the interaction between iC3b and its receptors CR3/CR4, we isolated the integrin-binding fragment of iC3b, MG3-4. Low temperature is required for its soluble expression in Escherichia coli. Purified MG3-4 existed as a dimer in solution and was easy to aggregate. We tried different agents and found glycerol could efficiently stabilize the MG3-4 fragment to avoid aggregation. Using surface plasmon resonance (SPR) analysis, we confirmed MG3-4 could bind I domain, the iC3b-binding domain of CR3. Here, we report the successful production of a soluble, stable, and biologically active integrin-binding moiety of human iC3b for further studies.


Assuntos
Complemento C3b/química , Integrina alfaXbeta2/química , Antígeno de Macrófago 1/química , Clonagem Molecular , Complemento C3b/genética , Complemento C3b/imunologia , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Glicerol/química , Humanos , Imunidade Inata , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/imunologia , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Modelos Moleculares , Agregados Proteicos , Ligação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Soluções , Ressonância de Plasmônio de Superfície
12.
Immunol Lett ; 168(1): 13-21, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26306739

RESUMO

The opportunistic pathogenic yeast Candida albicans employs several mechanisms to interfere with the human complement system. This includes the acquisition of host complement regulators, the release of molecules that scavenge complement proteins or block cellular receptors, and the secretion of proteases that inactivate complement components. Secreted aspartic protease 2 (Sap2) was previously shown to cleave C3b, C4b and C5. C. albicans also recruits the complement inhibitor factor H (FH), but yeast-bound FH can enhance the antifungal activity of human neutrophils via binding to complement receptor type 3 (CR3). In this study, we characterized FH binding to human monocyte-derived macrophages. Inhibition studies with antibodies and siRNA targeting CR3 (CD11b/CD18) and CR4 (CD11c/CD18), as well as analysis of colocalization of FH with these integrins indicated that both function as FH receptors on macrophages. Preincubation of C. albicans yeast cells with FH induced increased production of IL-1ß and IL-6 in macrophages. Furthermore, FH enhanced zymosan-induced production of these cytokines. C. albicans Sap2 cleaved FH, diminishing its complement regulatory activity, and Sap2-treatment resulted in less detectable CR3 and CR4 on macrophages. These data show that FH enhances the activation of human macrophages when bound on C. albicans. However, the fungus can inactivate both FH and its receptors on macrophages by secreting Sap2, which may represent an additional means for C. albicans to evade the host innate immune system.


Assuntos
Ácido Aspártico Endopeptidases/imunologia , Candida albicans/imunologia , Fator H do Complemento/imunologia , Proteínas Fúngicas/imunologia , Integrina alfaXbeta2/imunologia , Antígeno de Macrófago 1/imunologia , Ácido Aspártico Endopeptidases/metabolismo , Western Blotting , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Antígeno CD11b/metabolismo , Antígeno CD11c/genética , Antígeno CD11c/imunologia , Antígeno CD11c/metabolismo , Antígenos CD18/genética , Antígenos CD18/imunologia , Antígenos CD18/metabolismo , Candida albicans/enzimologia , Candida albicans/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Fator H do Complemento/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Proteínas Fúngicas/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/metabolismo , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/metabolismo , Interferência de RNA
13.
PLoS One ; 9(7): e102014, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25061945

RESUMO

In dengue virus (DENV) infection, complement system (CS) activation appears to have protective and pathogenic effects. In severe dengue fever (DF), the levels of DENV non-structural-1 protein and of the products of complement activation, including C3a, C5a and SC5b-9, are higher before vascular leakage occurs, supporting the hypothesis that complement activation contributes to unfavourable outcomes. The clinical manifestations of DF range from asymptomatic to severe and even fatal. Here, we aimed to characterise CS by their receptors or activation product, in vivo in DF patients and in vitro by DENV-2 stimulation on monocytes. In comparison with healthy controls, DF patients showed lower expression of CR3 (CD11b), CR4 (CD11c) and, CD59 on monocytes. The DF patients who were high producers of SC5b-9 were also those that showed more pronounced bleeding or vascular leakage. Those findings encouraged us to investigate the role of CS in vitro, using monocytes isolated from healthy subjects. Prior blocking with CR3 alone (CD11b) or CR3 (CD11b/CD18) reduced viral infection, as quantified by the levels of intracellular viral antigen expression and soluble DENV non-structural viral protein. However, we found that CR3 alone (CD11b) or CR3 (CD11b/CD18) blocking did not influence major histocompatibility complex presentation neither active caspase-1 on monocytes, thus probably ruling out inflammasome-related mechanisms. Although it did impair the secretion of tumour necrosis factor alpha and interferon alpha. Our data provide strategies of blocking CR3 (CD11b) pathways could have implications for the treatment of viral infection by antiviral-related mechanisms.


Assuntos
Vírus da Dengue/imunologia , Integrina alfaXbeta2/imunologia , Antígeno de Macrófago 1/imunologia , Dengue Grave/imunologia , Adulto , Caspase 1/imunologia , Ativação do Complemento/imunologia , Complemento C3a/biossíntese , Complemento C3a/imunologia , Complemento C5a/biossíntese , Complemento C5a/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/biossíntese , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Vírus da Dengue/patogenicidade , Feminino , Regulação Viral da Expressão Gênica , Humanos , Integrina alfaXbeta2/genética , Antígeno de Macrófago 1/genética , Masculino , Pessoa de Meia-Idade , Monócitos , Dengue Grave/genética , Dengue Grave/patologia , Dengue Grave/virologia , Proteínas não Estruturais Virais/imunologia
14.
J Exp Med ; 211(7): 1485-97, 2014 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-24889201

RESUMO

Rodent-borne hantaviruses are emerging human pathogens that cause severe human disease. The underlying mechanisms are not well understood, as hantaviruses replicate in endothelial and epithelial cells without causing any cytopathic effect. We demonstrate that hantaviruses strongly stimulated neutrophils to release neutrophil extracellular traps (NETs). Hantavirus infection induced high systemic levels of circulating NETs in patients and this systemic NET overflow was accompanied by production of autoantibodies to nuclear antigens. Analysis of the responsible mechanism using neutrophils from ß2 null mice identified ß2 integrin receptors as a master switch for NET induction. Further experiments suggested that ß2 integrin receptors such as complement receptor 3 (CR3) and 4 (CR4) may act as novel hantavirus entry receptors. Using adenoviruses, we confirmed that viral interaction with ß2 integrin induced strong NET formation. Collectively, ß2 integrin-mediated systemic NET overflow is a novel viral mechanism of immunopathology that may be responsible for characteristic aspects of hantavirus-associated disease such as kidney and lung damage.


Assuntos
Antígenos CD18/imunologia , Infecções por Hantavirus/imunologia , Neutrófilos/imunologia , Orthohantavírus/imunologia , Adenoviridae , Animais , Autoanticorpos/genética , Autoanticorpos/imunologia , Antígenos CD18/genética , Células CHO , Cricetinae , Cricetulus , Feminino , Infecções por Hantavirus/genética , Infecções por Hantavirus/patologia , Humanos , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/imunologia , Nefropatias/genética , Nefropatias/imunologia , Nefropatias/patologia , Nefropatias/virologia , Pneumopatias/genética , Pneumopatias/imunologia , Pneumopatias/patologia , Pneumopatias/virologia , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Masculino , Camundongos , Camundongos Mutantes , Neutrófilos/patologia
15.
Int J Biochem Cell Biol ; 45(7): 1204-11, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23542015

RESUMO

Integrins αLß2, αMß2 and αXß2 are expressed on leukocytes. Their primary ligands are counter transmembrane receptors or plasma proteins, such as intercellular cell adhesion molecule-1 (ICAM-1) or components of complement system (iC3b, iC4b), respectively. Function blocking antibodies for these integrins may also reduce cell adhesion to collagens. To make the first systematical comparison of human α(L)ß2, α(M)ß2 and α(X)ß2 as collagen receptors, we produced the corresponding integrin αI domains both in wild-type and activated form and measured their binding to collagens I-VI. In the "closed" (wild-type) conformation, the α(L)I and α(M)I domains bound with low avidity to their primary ligands, and the interaction with collagens was also very weak. Gain-of-function mutations α(L) I306G, α(L) K287C/K294C and α(M) I316G are considered to mimic "open", activated αI domains. The binding of these activated αI domains to the primary ligands was clearly stronger and they also recognized collagens with moderate avidity (K(d)400 nM). After activation, the αLI domain favored collagen I (K(d )≈ 80 nM) when compared to collagen IV. The integrin αXI domain acted in a very different manner since already in native, wild-type form it bound to collagen IV and iC3b (K(d) ≈ 200-400 nM). Antibodies against αXß2 and αMß2 blocked promyelocytic leukemia cell adhesion to the collagenous GFOGER motif, a binding site for the ß1 integrin containing collagen receptors. In brief, leukocyte ß2 integrins may act as collagen receptors in a heterodimer specific manner.


Assuntos
Colágeno/metabolismo , Integrina alfaXbeta2/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Antígeno de Macrófago 1/metabolismo , Motivos de Aminoácidos , Anticorpos Bloqueadores/imunologia , Sítios de Ligação , Adesão Celular , Linhagem Celular Tumoral , Células HL-60 , Humanos , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/imunologia , Leucemia Promielocítica Aguda/metabolismo , Antígeno-1 Associado à Função Linfocitária/genética , Antígeno-1 Associado à Função Linfocitária/imunologia , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Colágeno
16.
J Cell Biol ; 203(4): 629-42, 2013 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-24385486

RESUMO

How is massive conformational change in integrins achieved on a rapid timescale? We report crystal structures of a metastable, putative transition state of integrin αXß2. The αXß2 ectodomain is bent; however, a lattice contact stabilizes its ligand-binding αI domain in a high affinity, open conformation. Much of the αI α7 helix unwinds, loses contact with the αI domain, and reshapes to form an internal ligand that binds to the interface between the ß propeller and ßI domains. Lift-off of the αI domain above this platform enables a range of extensional and rotational motions without precedent in allosteric machines. Movements of secondary structure elements in the ß2 ßI domain occur in an order different than in ß3 integrins, showing that integrin ß subunits can be specialized to assume different intermediate states between closed and open. Mutations demonstrate that the structure trapped here is metastable and can enable rapid equilibration between bent and extended-open integrin conformations and up-regulation of leukocyte adhesiveness.


Assuntos
Integrina alfaXbeta2/química , Integrina alfaXbeta2/metabolismo , Leucócitos/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Complemento C3b/metabolismo , Cristalografia por Raios X , Epitopos/química , Eritrócitos/metabolismo , Células HEK293 , Humanos , Integrina alfaXbeta2/genética , Ligantes , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Ligação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Ovinos
17.
Immunobiology ; 218(4): 652-63, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22906751

RESUMO

Dendritic cells (DCs) play a decisive role in immunity; they interact with various pathogens via several pattern recognition and different opsonophagocytotic receptors, including Fc- and complement-receptors. ß2-integrins, including complement receptors CR3 (CD11b/CD18) and CR4 (CD11c/CD18) participate in many immunological processes, especially those involving cell migration, adherence, and phagocytosis. Human monocyte derived dendritic cells (MDCs) are known to express CR3 as well as CR4, however possible differences regarding the role of these receptors has not been addressed so far. Our aim was to explore whether there is a difference between the binding and uptake of various complement-opsonized microorganisms, mediated by CR3 and CR4. Studying the expression of receptors during differentiation of MDCs we found that the appearance of CD11b decreased, whereas that of CD11c increased. Interestingly, both receptors were present in the cell membrane in an active conformation. Here we demonstrate that ligation of CD11b directs MDCs to enhanced phagocytosis, while the maturation of the cells and their inflammatory cytokine production are not affected. Blocking CD11c alone did not change the uptake of opsonized yeast or bacteria by MDCs. We confirmed these results using siRNA; namely downregulation of CD11b blocked the phagocytosis of microbes while silencing CD11c had no effect on their uptake. Our data clearly demonstrate that complement C3-dependent phagocytosis of MDCs is mediated mainly by CR3.


Assuntos
Movimento Celular/fisiologia , Células Dendríticas/imunologia , Antígeno de Macrófago 1/imunologia , Monócitos/imunologia , Fagocitose/fisiologia , Adesão Celular/fisiologia , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Inativação Gênica , Humanos , Integrina alfaXbeta2/biossíntese , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/imunologia , Antígeno de Macrófago 1/biossíntese , Antígeno de Macrófago 1/genética , Masculino , Monócitos/citologia , Monócitos/metabolismo
18.
Parasite Immunol ; 34(11): 547-50, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22882618

RESUMO

Complement receptors for C3-derived fragments (CR1-4) play critical roles in innate and adaptive immune responses. Of these receptors, CR3 and CR4 are important in binding and phagocytosis of complement-opsonized pathogens including parasites. The role of CR3 and CR4 in malaria or in cerebral malaria (CM) has received little attention and remains poorly understood in both human disease and rodent models of malaria. CR3 and CR4 are members of the ß(2) -integrin family of adhesion molecules and are expressed on all leucocytes that participate in the development of CM, most importantly as it relates to parasite phagocytosis (monocytes/macrophages) and antigen processing and presentation (dendritic cells). Thus, it is possible that these receptors might play an important role in disease development. To address this question, we examined the role of CR3(-/-) and CR4(-/-) in experimental cerebral malaria (ECM). We found that both CR3(-/-) and CR4(-/-) mice were fully susceptible to ECM and developed disease comparable to wild-type mice. Our results indicate that CR3 and CR4 are not critical to the pathogenesis of ECM despite their role in elimination of complement-opsonized pathogens. These findings support recent studies indicating the importance of the terminal complement pathway and the membrane attack complex in ECM pathogenesis.


Assuntos
Suscetibilidade a Doenças , Integrina alfaXbeta2/genética , Antígeno de Macrófago 1/genética , Malária Cerebral/genética , Malária Cerebral/imunologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Deleção de Sequência
19.
J Immunol ; 189(5): 2468-77, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22844116

RESUMO

The opportunistic fungus Candida albicans is one of the leading causes of infections in immunocompromised patients, and innate immunity provides a principal mechanism for protection from the pathogen. In the present work, the role of integrin α(X)ß2 in the pathogenesis of fungal infection was assessed. Both purified α(X)ß2 and α(X)ß2-expressing human epithelial kidney 293 cells recognized and bound to the fungal hyphae of SC5314 strain of C. albicans but not to the yeast form or to hyphae of a strain deficient in the fungal mannoprotein, Pra1. The binding of the integrin to the fungus was inhibited by ß-glucans but not by mannans, implicating a lectin-like activity in recognition but distinct in specificity from that of α(M)ß2. Mice deficient in α(X)ß2 were more prone to systemic infection with the LD50 fungal inoculum decreasing 3-fold in α(X)ß2-deficient mice compared with wild-type mice. After challenging i.v. with 1.5 × 104 cell/g, 60% of control C57BL/6 mice died within 14 d compared with 100% mortality of α(X)ß2-deficient mice within 9 d. Organs taken from α(X)ß2-deficient mice 16 h postinfection revealed a 10-fold increase in fungal invasion into the brain and a 2-fold increase into the liver. These data indicate that α(X)ß2 is important for protection against systemic C. albicans infections and macrophage subsets in the liver, Kupffer cells, and in the brain, microglial cells use α(X)ß2 to control fungal invasion.


Assuntos
Candida albicans/patogenicidade , Candidíase/imunologia , Candidíase/prevenção & controle , Integrina alfaXbeta2/metabolismo , Leucócitos/metabolismo , Receptores Imunológicos/metabolismo , Animais , Candidíase/microbiologia , Adesão Celular/imunologia , Linhagem Celular , Movimento Celular/imunologia , Citofagocitose/imunologia , Células HEK293 , Humanos , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/fisiologia , Leucócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores Imunológicos/fisiologia
20.
Vet Immunol Immunopathol ; 135(3-4): 266-74, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20060597

RESUMO

Pneumonia caused by Mannheimia haemolytica is an important disease of domestic sheep (DS, Ovis aries) and cattle (BO). M. haemolytica is a normal commensal of the upper respiratory tract in ruminants, but during stress and viral infection it breaches the host innate mucosal defense and descents into lungs causing fibrinous pleuropneumonia. Leukotoxin (Lkt) produced by M. haemolytica is cytolytic to all subsets of ruminant leukocytes. Earlier, we and others have shown that DS and BO LFA-1 (CD11a/CD18) and Mac-1 (CD11b/CD18) can mediate Lkt-induced cytolysis. It is not clear whether CR4 (CD11c/CD18), which is involved in chemotaxis, phagocytosis and regulates host immune response can also mediate Lkt-induced cytolysis in ruminants. The host innate immune response to M. haemolytica is poorly understood and the involvement of CR4 in M. haemolytica pathogenesis is one of the most understudied. This problem is further compounded by the lack of cd11c genes from any ruminant species. Therefore, the objectives of this study were to clone cd11c and determine whether CR4 can serve as a receptor for Lkt. In this direction we cloned two alleles of cd11c gene from leukocytes isolated from DS blood by RT-PCR. Transfectants developed expressing functional DS CR4 were found to be cytotoxic to Lkt from four different isolates of M. haemolytica. This is the first report confirming the ability of a recombinant ovine CR4 to bind to M. haemolytica Lkt and mediate concentration-dependent lysis of host cells, thus, confirming their role in M. haemolytica pathogenesis. This is a critical step in understanding host innate immunity and the management of pneumonia in sheep.


Assuntos
Toxinas Bacterianas/toxicidade , Exotoxinas/toxicidade , Integrina alfaXbeta2/metabolismo , Mannheimia haemolytica/patogenicidade , Ovinos/imunologia , Sequência de Aminoácidos , Animais , Toxinas Bacterianas/imunologia , Sequência de Bases , Bovinos , Linhagem Celular , Clonagem Molecular , Citotoxinas/imunologia , Citotoxinas/toxicidade , Primers do DNA/genética , Exotoxinas/imunologia , Humanos , Integrina alfaXbeta2/química , Integrina alfaXbeta2/genética , Mannheimia haemolytica/imunologia , Dados de Sequência Molecular , Infecções por Pasteurellaceae/imunologia , Infecções por Pasteurellaceae/microbiologia , Infecções por Pasteurellaceae/veterinária , Pasteurelose Pneumônica/imunologia , Pasteurelose Pneumônica/microbiologia , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Ovinos/genética , Doenças dos Ovinos/imunologia , Doenças dos Ovinos/microbiologia , Transfecção , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...