Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
PLoS One ; 15(5): e0233644, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32469940

RESUMO

Cytotoxic and pro-inflammatory histones are present in neutrophil extracellular traps (NETs) and are elevated in blood in several inflammatory conditions, sepsis being a major example. Compounds which can attenuate activities of histones are therefore of interest, with heparin being one such material that has previously been shown to bind to histones. Heparin, a successful anticoagulant for nearly a century, has been shown experimentally to bind to histones and exhibit a protective effect in inflammatory conditions. In the present study carried out in whole blood, heparin and selectively desulfated heparin reduced histone induced inflammatory markers such as interleukin 6 (IL 6), interleukin 8 (IL 8) and tissue factor and C3a, a complement component. The selectively desulfated heparins, with reduced anticoagulant activities, retained a high degree of effectiveness as an anti-histone agent, whereas fully desulfated heparin was found to be ineffective. The results from this study indicate that the presence of sulfate and other specific structural features are required for heparin to attenuate the inflammatory action of histones in whole blood.


Assuntos
Anti-Inflamatórios/farmacologia , Anticoagulantes/farmacologia , Heparina/farmacologia , Histonas/imunologia , Inflamação/tratamento farmacológico , Anti-Inflamatórios/química , Anticoagulantes/química , Complemento C3a/análise , Complemento C3a/imunologia , Heparina/análogos & derivados , Histonas/sangue , Humanos , Inflamação/sangue , Inflamação/imunologia , Interleucina-6/sangue , Interleucina-6/imunologia , Interleucina-8/sangue , Interleucina-8/imunologia
2.
FASEB J ; 34(6): 7540-7560, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32301538

RESUMO

The vascular endothelium has been discovered in the past several years to be important in shaping the cellular immune response. During the immune response the vascular endothelium is constantly perturbed by biologically potent molecules, including the complement activation peptides, C3a and C5a. Despite the importance of C3a and C5a in inflammation and immunity, their role in modulating lymphocyte function via activation of vascular endothelial cells is unknown. Accordingly, we investigated the regulated expression of the C3a and C5a receptors (complement anaphylatoxin C3a receptor [C3aR] and complement anaphylatoxin C5a receptor 1 [C5aR1]) on human umbilical vascular endothelial cells (HUVECs) and examined how C3a or C5a activation of HUVECs affects the activation and polarization of lymphatic cells. Our findings demonstrated that C3a and C5a increase C3aR and C5aR1 expression by HUVECs as well as directing their cellular transmigration and spreading through transwell filters. Moreover, C3a- or C5a-stimulated endothelial cells: (1) caused activation of B-lymphoblasts with significant increase in Fas Ligand (CD95L) (FasL), CD69, and IL-R1 expression, and (2) skewed T-lymphoblast cells toward a Th1 subtype, (CD4+ /CCR5+ ) that correlated with significant increase of IFN-γ. Collectively, these data indicate that C3a and C5a signaling is important in the activation and polarization of lymphocytes as they traffic through the vascular endothelium during the immune response.


Assuntos
Anafilatoxinas/imunologia , Linfócitos B/imunologia , Complemento C3a/imunologia , Complemento C5a/imunologia , Peptídeos/imunologia , Linfócitos T/imunologia , Células Cultivadas , Ativação do Complemento/imunologia , Endotélio Vascular/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/imunologia , Receptor da Anafilatoxina C5a/imunologia , Receptores de Complemento/imunologia , Transdução de Sinais/imunologia
3.
Oncogene ; 39(12): 2612-2623, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32020055

RESUMO

Neutrophils represent the immune system's first line of defense and are rapidly recruited into inflamed tissue. In cancer associated inflammation, phenotypic heterogeneity has been ascribed to this cell type, whereby neutrophils can manifest anti- or pro-metastatic functions depending on the cellular/micro-environmental context. Here, we demonstrate that pro-metastatic immature low-density neutrophils (iLDNs) more efficiently accumulate in the livers of mice bearing metastatic lesions compared with anti-metastatic mature high-density neutrophils (HDNs). Transcriptomic analyses reveal enrichment of a migration signature in iLDNs relative to HDNs. We find that conditioned media derived from liver-metastatic breast cancer cells, but not lung-metastatic variants, specifically induces chemotaxis of iLDNs and not HDNs. Chemotactic responses are due to increased surface expression of C3aR in iLDNs relative to HDNs. In addition, we detect elevated secretion of cancer-cell derived C3a from liver-metastatic versus lung-metastatic breast cancer cells. Perturbation of C3a/C3aR signaling axis with either a small molecule inhibitor, SB290157, or reducing the levels of secreted C3a from liver-metastatic breast cancer cells by short hairpin RNAs, can abrogate the chemotactic response of iLDNs both in vitro and in vivo, respectively. Together, these data reveal novel mechanisms through which iLDNs prefentially accumulate in liver tissue harboring metastases in response to tumor-derived C3a secreted from the liver-aggressive 4T1 breast cancer cells.


Assuntos
Complemento C3a/imunologia , Neoplasias Hepáticas/imunologia , Neutrófilos/imunologia , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Meios de Cultivo Condicionados , Feminino , Neoplasias Hepáticas/secundário , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Receptores de Complemento/agonistas , Receptores de Complemento/metabolismo
4.
Front Immunol ; 10: 954, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31134065

RESUMO

Lung cancer is the leading cause of cancer death in men and women. Lung adenocarcinoma (LUAD), represents approximately 40% of all lung cancer cases. Advances in recent years, such as the identification of oncogenes and the use of immunotherapies, have changed the treatment of LUAD. Yet survival rates still remain low. Additionally, there is still a gap in understanding the molecular and cellular interactions between cancer cells and the immune tumor microenvironment (TME). Defining how cancer cells with distinct oncogenic drivers interact with the TME and new strategies for enhancing anti-tumor immunity are greatly needed. The complement cascade, a central part of the innate immune system, plays an important role in regulation of adaptive immunity. Initially it was proposed that complement activation on the surface of cancer cells would inhibit cancer progression via membrane attack complex (MAC)-dependent killing. However, data from several groups have shown that complement activation promotes cancer progression, probably through the actions of anaphylatoxins (C3a and C5a) on the TME and engagement of immunoevasive pathways. While originally shown to be produced in the liver, recent studies show localized complement production in numerous cell types including immune cells and tumor cells. These results suggest that complement inhibitory drugs may represent a powerful new approach for treatment of NSCLC, and numerous new anti-complement drugs are in clinical development. However, the mechanisms by which complement is activated and affects tumor progression are not well understood. Furthermore, the role of local complement production vs. systemic activation has not been carefully examined. This review will focus on our current understanding of complement action in LUAD, and describe gaps in our knowledge critical for advancing complement therapy into the clinic.


Assuntos
Imunidade Adaptativa/imunologia , Adenocarcinoma de Pulmão/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Ativação do Complemento/imunologia , Neoplasias Pulmonares/imunologia , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Complemento C3a/imunologia , Complemento C5a/imunologia , Inativadores do Complemento/uso terapêutico , Humanos , Imunoterapia/métodos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética
5.
Mol Vis ; 25: 79-92, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30820144

RESUMO

Purpose: Smoking and the incidence of age-related macular degeneration (AMD) have been linked to an overactive complement system. Here, we examined in a retrospective cohort study whether AMD-associated single nucleotide polymorphisms (SNPs), smoking, ethnicity, and disease status are correlated with blood complement levels. Methods: Population: The study involved 91 AMD patients and 133 controls, which included 73% Americans of European descent (EUR) and 27% Americans of African descent (AFR) in South Carolina. Readouts: Participants were genotyped for 10 SNPs and systemic levels of complement factor H (CFH) activity, and the complement activation products C3a, C5a, and Bb were assessed. Main Outcome Measures: Univariate and multivariable logistic regression models were used to examine associations between AMD status and distinct readouts. Results: AMD affects EUR individuals more than AFRs. EUR but not AFR AMD subjects revealed higher levels of Factors C3a and Bb. In all subjects, a 10-unit increase in C3a levels was associated with an approximately 10% increase in the odds of being AMD-positive, and C3a and Bb were associated with smoking. While CFH activity levels were not correlated with AMD, a significant interaction was evident between patient age and CFH activity. Finally, EURs had lower odds of AMD with enhanced copies of rs1536304 (VEGFA) and higher odds with more copy numbers of rs3766404 (CFH). Conclusions: Our results support previous studies of systemic complement components being potential biomarkers for AMD, but they suggest that smoking and disease do not synergistically affect complement levels. We also suggest a novel susceptibility and protective haplotypes in the South Carolinian AMD population. Our studies indicate that augmented complement activation associated with advanced AMD could be attributed to a decrease in CFH activity in younger patients.


Assuntos
Ativação do Complemento/genética , Fator H do Complemento/genética , Degeneração Macular/genética , Polimorfismo de Nucleotídeo Único , Fumar/genética , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , População Negra , Estudos de Casos e Controles , Complemento C3a/genética , Complemento C3a/imunologia , Complemento C5a/genética , Complemento C5a/imunologia , Fator B do Complemento/genética , Fator B do Complemento/imunologia , Fator H do Complemento/imunologia , Feminino , Expressão Gênica , Humanos , Modelos Logísticos , Degeneração Macular/etnologia , Degeneração Macular/imunologia , Degeneração Macular/patologia , Masculino , Estudos Retrospectivos , Fumar/etnologia , Fumar/imunologia , Fumar/fisiopatologia , South Carolina , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , População Branca
6.
Am J Respir Cell Mol Biol ; 60(4): 454-464, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30422670

RESUMO

In the present study, we sought to elucidate the mechanisms by which monocytes migrate into the pleural space in the presence of anaphylatoxins in tuberculous pleural effusion (TPE). Monocytes in both pleural effusion and blood were counted, and their phenotypic characteristics were analyzed. Activation of the complement system was detected in TPE. The effects of Mpt64 and anaphylatoxins on the production of chemokines in pleural mesothelial cells (PMCs) were measured. The chemoattractant activity of chemokines produced by PMCs for monocytes was observed. Levels of CD14+CD16+ monocytes were significantly higher in TPE than in blood. Three pathways of the complement system were activated in TPE. C3a-C3aR1, C5a-C5aR1, CCL2-CCR2, CCL7-CCR2, and CX3CL1-CX3CR1 were coexpressed in PMCs and monocytes isolated from TPE. Moreover, we initially found that Mpt64 stimulated the expression of C3a and C5a in PMCs. C3a and C5a not only induced CCL2, CCL7, and CX3CL1 expression in PMCs but also stimulated production of IL-1ß, IL-17, and IL-27 in monocytes. C3a and C5a stimulated PMCs to secrete CCL2, CCL7, and CX3CL1, which recruited CD14+CD16+ monocytes to the pleural cavity. As a result, the infiltration of CD14+CD16+ monocytes engaged in the pathogenesis of TPE by excessive production of inflammatory cytokines.


Assuntos
Anafilatoxinas/metabolismo , Monócitos/metabolismo , Derrame Pleural/patologia , Tuberculose Pulmonar/patologia , Movimento Celular/fisiologia , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Quimiocina CX3CL1/metabolismo , Complemento C3a/imunologia , Complemento C5a/imunologia , Células Epiteliais , Epitélio/patologia , Humanos , Pleura/citologia , Pleura/patologia , Derrame Pleural/microbiologia
7.
Front Immunol ; 9: 2449, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30405635

RESUMO

Autocrine activation of the complement receptors C3aR and CD46 by complement activation components C3a and C3b produced through C3 cleavage by the protease cathepsin L (CTSL) during T cell stimulation is a requirement for IFN-γ production and Th1 induction in human CD4+ T cells. Thus, lack of autocrine CD46 activation, such as in CD46-deficient patients, is associated with defective Th1 responses and recurrent infections. We have identified LGMN [the gene coding for legumain, also known as asparaginyl endopeptidase (AEP)] as one of the key genes induced by CD46 co-stimulation during human CD4+ T cell activation. AEP processes and activates a range of proteins, among those α1-thymosin and CTSL, which both drive intrinsically Th1 activity-but has so far not been described to be functionally active in human T cells. Here we found that pharmacological inhibition of AEP during activation of human CD4+ T cells reduced CTSL activation and the CTSL-mediated generation of intracellular C3a. This translated into a specific reduction of IFN-γ production without affecting cell proliferation or survival. In line with these findings, CD4+ T cells isolated from Lgmn-/- mice also displayed a specific defect in IFN-γ secretion and Th1 induction. Furthermore, we did not observe a role for AEP-driven autocrine α1-thymosin activation in T cell-derived IFN-γ production. These data suggest that AEP is an "upstream" activator of the CTSL-C3-IFN-γ axis in human CD4+ T cells and hence an important supporter of human Th1 induction.


Assuntos
Catepsina L/metabolismo , Complemento C3a/imunologia , Complemento C3b/imunologia , Cisteína Endopeptidases/metabolismo , Interferon gama/metabolismo , Células Th1/imunologia , Animais , Proliferação de Células , Cisteína Endopeptidases/genética , Humanos , Interferon gama/biossíntese , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Proteína Cofatora de Membrana/metabolismo , Camundongos , Camundongos Knockout , Receptores de Complemento/metabolismo , Timalfasina/metabolismo
8.
Innate Immun ; 24(1): 66-78, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29297237

RESUMO

Complement is an important arm of the innate immune system. Recent studies have shown that products of complement pathway activation can interact directly with other innate immune signaling molecules, including TLRs and inflammasome family members, during some infectious and chronic inflammatory disorders. Activation of the complement system generates anaphylatoxins, such as C3a and C5a, which modulate inflammation. However, the biological effects of interactions between the anaphylatoxins with their receptors may vary across species. In this study, we demonstrate that human complement and rat complement differ in the way they modulate the inflammatory response to the human pathogen, Neisseria gonorrhoeae, as well as purified pathogen-associated ligands, such as LPS. While rat serum down-regulates MyD88-dependent pro-inflammatory cytokine responses in macrophages, human serum has no effect, or in some cases an enhancing effect. Further, the inhibitory effect of rat serum on otherwise pro-inflammatory stimuli is mediated by complement, specifically C3a-C3a receptor interactions, via an undefined signaling mechanism that down-regulates the transcription factor, NF-κB and NLRP3 inflammasome-mediated caspase-1 activation. This study highlights important functional differences between rodent and human complement that could explain some of the differences in immune responses between these two species. Understanding the crosstalk between complement and other arms of the innate immune system will facilitate the development of better anti-inflammatory therapeutics.


Assuntos
Complemento C3a/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Animais , Ativação do Complemento , Citocinas/biossíntese , Gonorreia/imunologia , Humanos , Lipopolissacarídeos/farmacologia , Fator 88 de Diferenciação Mieloide/imunologia , NF-kappa B/biossíntese , Proteína 3 que Contém Domínio de Pirina da Família NLR/biossíntese , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Neisseria gonorrhoeae/imunologia , Ratos Sprague-Dawley , Transdução de Sinais/imunologia , Especificidade da Espécie
9.
Rheumatology (Oxford) ; 57(1): 120-124, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29045759

RESUMO

Objectives: HCQ has been described as having a beneficial effect in patients with APS but its mechanism of action is unclear. We hypothesized that HCQ may have effects on subnormal angiogenesis, inflammation and haemostatic biomarkers seen in APS. The aim of our study was to assess laboratory markers [annexin A5 (AnxA5) anticoagulant activity, tissue factor (TF) levels, thromboelastography (TEG), CRP, Bb, C3a and VEGF] in HCQ-naïve patients with aPL at baseline and after commencing HCQ. Methods: Twenty-two patients with aPL [20 female, 2 male, median age 55 (range 18-70) years] had blood taken pre- and 3 months after starting HCQ 200 mg daily. Results: Soluble TF levels were significantly reduced comparing baseline and 3 months after HCQ commencement [401.8 (152.8) vs 300.9 (108) pg/ml (P = 0.010)]. No significant changes were found in the following [reported as pre- and post-HCQ commencement, mean (s.d.)]: AnxA5 anticoagulant ratio [187.1 (29.5) vs 193 (31) (P = 0.157)], anti-domain1 ß2 glycoprotein1 IgG activity [1.8 (2) vs 1.2 (1.4) µg/ml (P = 0.105)], complement C3a-des-Arg [147.8 (84.5) vs 154.4 (88.1) ng/ml (P = 0.905)], complement Bb [1.3 (0.7) vs 1.1 (0.7) µg/ml (P = 0.422)], VEGF [68.8 (40) vs 59.4 (19.6) pg/ml (P = 0.454)] and CRP [7 (3.5) vs 7 (3.9) µg/ml (P = 0.917)]. TEG results including TEG reaction time, achievement of clot firmness, TEG maximum amplitude and TEG percentage lysis 30 and 60 min after maximum amplitude showed no significant difference. Conclusion: HCQ significantly reduced soluble TF levels in patients with aPL. No significant change was observed in AnxA5 activity, anti-domain 1 IgG activity, TEG, CRP, complement Bb and C3a-des-Arg, and VEGF. Further studies of a larger patient cohort are needed.


Assuntos
Síndrome Antifosfolipídica/tratamento farmacológico , Antirreumáticos/uso terapêutico , Hidroxicloroquina/uso terapêutico , Adolescente , Adulto , Idoso , Anexina A5/metabolismo , Anticorpos Antinucleares/imunologia , Anticorpos Antifosfolipídeos/imunologia , Síndrome Antifosfolipídica/imunologia , Síndrome Antifosfolipídica/metabolismo , Proteína C-Reativa/metabolismo , Complemento C3a/imunologia , Proteínas do Sistema Complemento/imunologia , Hemostasia , Humanos , Imunoglobulina G/imunologia , Pessoa de Meia-Idade , Neovascularização Fisiológica , Estudos Prospectivos , Tromboelastografia , Tromboplastina/metabolismo , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem , beta 2-Glicoproteína I/imunologia
10.
Curr Mol Med ; 17(2): 140-148, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28429668

RESUMO

OBJECTIVE: This study aimed to investigate the mechanisms whereby Amyloidbeta (Aß) induces the production of angiogenic factors by a human retinal pigment epithelial cell line (ARPE-19) cells. METHODS: ARPE-19 cells obtained from the American Type Culture Collection (ATCC) were utilized in this study. The expression level of vascular endothelial growth factor (VEGF), Interleukin-8 (IL-8), monocyte chemoattractant protein-1 (MCP-1) and complement activation fragments C3a and C5a were measured by Real-time quantitative PCR (RT-PCR) and Enzyme-linked immunosorbent assay (ELISA). The production of mitochondria-associated reactive oxygen species (ROS) was measured by flow cytometry. RESULTS: The expression of VEGF, IL-8, MCP-1, C3a and C5a was significantly increased in Aß-treated ARPE-19 cells. Mitochondria-associated ROS production was also significantly increased when exposed to Aß. Inhibition of mitochondrial ROS with Diphenyleneiodonium chloride (DPI) markedly decreased the Aß induced production of VEGF, IL-8, MCP-1, C3a and C5a by ARPE-19 cells. Anti-C3a or anti-C5a neutralizing antibodies did not have a detectable influence on the secretion of VEGF, IL-8 and MCP-1 by ARPE-19 cells upon stimulation with Aß. CONCLUSION: Our results support the hypothesis that Aß is involved in the pathogenesis of choroidal neovascularization (CNV) formation by promoting the production of the angiogenic cytokines VEGF, IL-8 and MCP-1 by RPE cells. Mitochondrial ROS was shown to play a role in the regulation of Aß induced expression of these cytokines.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Indutores da Angiogênese/metabolismo , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Biomarcadores , Linhagem Celular , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Complemento C3a/imunologia , Complemento C5a/imunologia , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Mitocôndrias/genética , Epitélio Pigmentado da Retina/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
J Immunol ; 198(8): 3237-3244, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28275134

RESUMO

Listeria monocytogenes is an intracellular Gram-positive bacterium that induces expression of type I IFNs (IFN-α/IFN-ß) during infection. These cytokines are detrimental to the host during infection by priming leukocytes to undergo L. monocytogenes-mediated apoptosis. Our previous studies showed that C5aR1-/- and C3aR-/- mice are highly susceptible to L. monocytogenes infection as a result of increased IFN-ß-mediated apoptosis of major leukocyte cell populations, including CD4+ and CD8+ T cells. However, the mechanisms by which C3a and C5a modulate IFN-ß expression during L. monocytogenes infection were not examined in these initial investigations. Accordingly, we report in this article that C5a and C3a suppress IFN-ß production in response to L. monocytogenes via cyclic di-AMP (c-di-AMP), a secondary messenger molecule of L. monocytogenes, in J774A.1 macrophage-like cells and in bone marrow-derived dendritic cells (BMDCs). Moreover, C5a and C3a suppress IFN-ß production by acting through their respective receptors, because no inhibition was seen in C5aR1-/- or C3aR-/- BMDCs, respectively. C5a and C3a suppress IFN-ß production in a manner that is dependent on Bruton's tyrosine kinase, p38 MAPK, and TANK-binding kinase 1 (TBK1), as demonstrated by the individual use of Bruton's tyrosine kinase, p38 MAPK, and TBK1 inhibitors. Pretreatment of cells with C5a and C3a reduced the expression of the IFN-ß signaling molecules DDX41, STING, phosphorylated TBK1, and phosphorylated p38 MAPK in wild-type BMDCs following treatment with c-di-AMP. Collectively, these data demonstrate that C3a and C5a, via direct signaling through their specific receptors, suppress IFN-ß expression by modulation of a distinct innate cytosolic surveillance pathway involving DDX41, STING, and other downstream molecular targets of L. monocytogenes-generated c-di-AMP.


Assuntos
Complemento C3a/imunologia , Complemento C5a/imunologia , Imunidade Inata/imunologia , Interferon beta/imunologia , Listeriose/imunologia , Transdução de Sinais/imunologia , Animais , Western Blotting , Complemento C3a/metabolismo , Complemento C5a/metabolismo , AMP Cíclico , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Interferon beta/biossíntese , Listeria monocytogenes , Listeriose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
J Allergy Clin Immunol ; 139(5): 1548-1558.e4, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27702673

RESUMO

BACKGROUND: The mechanism of TH2/TH17-predominant and TH2/TH17-low asthma is unknown. OBJECTIVE: We sought to study the immune mechanism of TH2/TH17-predominant and TH2/TH17-low asthma. METHODS: In a previously reported cohort of 60 asthmatic patients, 16 patients were immunophenotyped with TH2/TH17-predominant asthma and 22 patients with TH2/TH17-low asthma. We examined bronchoalveolar lavage (BAL) fluid leukocytes, cytokines, mediators, and epithelial cell function for these asthma subgroups. RESULTS: Patients with TH2/TH17-predominant asthma had increased IL-1ß, IL-6, IL-23, C3a, and serum amyloid A levels in BAL fluid, and these correlated with IL-1ß and C3a levels. TH2/TH17 cells expressed higher levels of the IL-1 receptor and phospho-p38 mitogen-activated protein kinase. Anakinra, an IL-1 receptor antagonist protein, inhibited BAL TH2/TH17 cell counts. TH2/TH17-low asthma had 2 distinct subgroups: neutrophilic asthma (45%) and pauci-inflammatory asthma (55%). This contrasted with patients with TH2/TH17-predominant and TH2-predominant asthma, which included neutrophilic asthma in 6% and 0% of patients, respectively. BAL fluid neutrophils strongly correlated with BAL fluid myeloperoxidase, IL-8, IL-1α, IL-6, granulocyte colony-stimulating factor, and GM-CSF levels. Sixty percent of the patients with neutrophilic asthma had a pathogenic microorganism in BAL culture, which suggested a subclinical infection. CONCLUSION: We uncovered a critical role for the IL-1ß pathway in patients with TH2/TH17-predminant asthma. A subgroup of patients with TH2/TH17-low asthma had neutrophilic asthma and increased BAL fluid IL-1α, IL-6, IL-8, granulocyte colony-stimulating factor, and GM-CSF levels. IL-1α was directly involved in IL-8 production and likely contributed to neutrophilic asthma. Sixty percent of neutrophilic patients had a subclinical infection.


Assuntos
Asma/imunologia , Neutrófilos/imunologia , Células Th17/imunologia , Células Th2/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Linhagem Celular , Células Cultivadas , Complemento C3a/imunologia , Citocinas/imunologia , Células Epiteliais/imunologia , Humanos , Contagem de Leucócitos , Lipopolissacarídeos , Proteína Amiloide A Sérica/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
13.
J Pathol ; 240(4): 495-506, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27659908

RESUMO

The discovery that genetic abnormalities in complement factor H (FH) are associated with an increased risk for age-related macular degeneration (AMD), the most common cause of blindness among the elderly, raised hope of new treatments for this vision-threatening disease. Nonetheless, over a decade after the identification of this important association, how innate immunity contributes to AMD remains unresolved. Pentraxin 3 (PTX3), an essential component of the innate immunity system that plays a non-redundant role in controlling inflammation, regulates complement by interacting with complement components. Here, we show that PTX3 is induced by oxidative stress, a known cause of AMD, in the retinal pigmented epithelium (RPE). PTX3 deficiency in vitro and in vivo magnified complement activation induced by oxidative stress, leading to increased C3a, FB, and C3d, but not C5b-9 complex formation. Increased C3a levels, resulting from PTX3 deficiency, raised the levels of Il1b mRNA and secretion of activated interleukin (IL)-1ß by interacting with C3aR. Importantly, PTX3 deficiency augmented NLRP3 inflammasome activation, resulting in enhanced IL-1ß, but not IL-18, production by the RPE. Thus, in the presence of PTX3 deficiency, the complement and inflammasome pathways worked in concert to produce IL-1ß in sufficient abundance to, importantly, result in macrophages accumulating in the choroid. These results demonstrate that PTX3 acts as an essential brake for complement and inflammasome activation by regulating the abundance of FH in the RPE, and provide critical insights into the complex interplay between oxidative stress and innate immunity in the early stages of AMD development. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Proteína C-Reativa/imunologia , Fator H do Complemento/imunologia , Inflamassomos/imunologia , Degeneração Macular/imunologia , Proteínas do Tecido Nervoso/imunologia , Estresse Oxidativo/imunologia , Aldeídos/farmacologia , Animais , Proteína C-Reativa/deficiência , Células Cultivadas , Corioide/imunologia , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Complemento C3a/imunologia , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Imunidade Inata/imunologia , Interleucina-1beta/biossíntese , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/deficiência , Estresse Oxidativo/efeitos dos fármacos , Epitélio Pigmentado da Retina/imunologia
14.
J Cell Sci ; 129(9): 1928-39, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27006116

RESUMO

Complement activation has long been associated with inflammation, primarily due to the elaboration of the complement anaphylotoxins C5a and C3a. In this work, we demonstrate that the phagocytosis of complement-opsonized particles promotes host inflammatory responses by a new mechanism that depends on the terminal complement components (C5b-C9). We demonstrate that during the phagocytosis of complement-opsonized particles, the membrane attack complex (MAC) of complement can be transferred from the activating particle to the macrophage plasma membrane by a 'bystander' mechanism. This MAC-mediated bystander damage initiates NLRP3 inflammasome activation, resulting in caspase-1 activation and IL-1ß and IL-18 secretion. Inflammasome activation is not induced when macrophages phagocytize unopsonized particles or particles opsonized with serum deficient in one of the terminal complement components. The secretion of IL-1ß and IL-18 by macrophages depends on NLRP3, ASC (also known as PYCARD) and caspase-1, as macrophages deficient in any one of these components fail to secrete these cytokines following phagocytosis. The phagocytosis of complement-opsonized particles increases leukocyte recruitment and promotes T helper 17 cell (TH17) biasing. These findings reveal a new mechanism by which complement promotes inflammation and regulates innate and adaptive immunity.


Assuntos
Efeito Espectador/imunologia , Complemento C3a/imunologia , Complemento C5a/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Macrófagos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Células Th17/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Efeito Espectador/genética , Proteínas Adaptadoras de Sinalização CARD , Complemento C3a/genética , Complemento C5a/genética , Complexo de Ataque à Membrana do Sistema Complemento/genética , Células HEK293 , Humanos , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Fagocitose/genética
15.
Ann Am Thorac Soc ; 12 Suppl 2: S164-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26595734

RESUMO

Heterogeneity in the development and progression of cigarette smoke-induced lung diseases strongly argues for a need to improve the clinical and phenotypic characterization of patients with chronic obstructive lung disease and emphysema. Smokers with emphysema are at a much higher risk for accelerated loss of lung function, increased cardiovascular morbidity, and development of lung cancer. Recent evidence in human translational studies and animal models suggests that emphysema is associated with activation of specialized antigen-presenting cells and that cigarette smoke can disrupt the induction of immune tolerance in the lungs. Quantitative assessment of cytokines expressed by autoreactive T lymphocytes in response to human lung elastin fragments has shown a strong positive correlation between T helper Type 1 (Th1) and Th17 cells' immune responses and emphysema. In search of factors that could reduce the threshold for induction of autoimmune inflammation, we have discovered that cleavage of complement protein 3 (C3) generates bioactive molecules (e.g., C3a) and activates lung antigen-presenting cells. The autocrine and paracrine function of C3a and its receptor are required in T cell-mediated inflammatory responses to cigarette smoke in both human and preclinical models of emphysema. Targeting upstream molecules that reduce the potential for generation of autoreactive T cells could lead to the development of novel therapeutics to prevent progression of emphysema in smokers.


Assuntos
Tolerância Periférica/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Enfisema Pulmonar/imunologia , Linfócitos T/imunologia , Animais , Autoimunidade , Complemento C3a/imunologia , Citocinas/imunologia , Modelos Animais de Doenças , Progressão da Doença , Humanos , Camundongos , Fenótipo , Fumar/efeitos adversos
16.
Nat Commun ; 6: 8483, 2015 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-26404464

RESUMO

The complement system is important for the host defence against infection as well as for the development of inflammatory diseases. Here we show that C1q/TNF-related protein 6 (CTRP6; gene symbol C1qtnf6) expression is elevated in mouse rheumatoid arthritis (RA) models. C1qtnf6(-/-) mice are highly susceptible to induced arthritis due to enhanced complement activation, whereas C1qtnf6-transgenic mice are refractory. The Arthus reaction and the development of experimental autoimmune encephalomyelitis are also enhanced in C1qtnf6(-/-) mice and C1qtnf6(-/-) embryos are semi-lethal. We find that CTRP6 specifically suppresses the alternative pathway of the complement system by competing with factor B for C3(H2O) binding. Furthermore, treatment of arthritis-induced mice with intra-articular injection of recombinant human CTRP6 cures the arthritis. CTRP6 is expressed in human synoviocytes, and CTRP6 levels are increased in RA patients. These results indicate that CTRP6 is an endogenous complement regulator and could be used for the treatment of complement-mediated diseases.


Assuntos
Adipocinas/imunologia , Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Via Alternativa do Complemento/imunologia , Adipocinas/genética , Adulto , Animais , Artrite Experimental/genética , Artrite Experimental/patologia , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Reação de Arthus/genética , Reação de Arthus/imunologia , Reação de Arthus/metabolismo , Western Blotting , Colágeno/imunologia , Colágeno/metabolismo , Convertases de Complemento C3-C5/imunologia , Complemento C3a/imunologia , Complemento C5a/imunologia , Via Alternativa do Complemento/genética , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Citometria de Fluxo , Humanos , Imunoprecipitação , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Membrana Sinovial/citologia , Membrana Sinovial/metabolismo
17.
Immunity ; 42(4): 767-77, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25888260

RESUMO

Radiotherapy induces DNA damage and cell death, but recent data suggest that concomitant immune stimulation is an integral part of the therapeutic action of ionizing radiation. It is poorly understood how radiotherapy supports tumor-specific immunity. Here we report that radiotherapy induced tumor cell death and transiently activated complement both in murine and human tumors. The local production of pro-inflammatory anaphylatoxins C3a and C5a was crucial to the tumor response to radiotherapy and concomitant stimulation of tumor-specific immunity. Dexamethasone, a drug frequently given during radiotherapy, limited complement activation and the anti-tumor effects of the immune system. Overall, our findings indicate that anaphylatoxins are key players in radiotherapy-induced tumor-specific immunity and the ensuing clinical responses.


Assuntos
Complemento C3a/imunologia , Complemento C5a/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Imunidade Inata/efeitos da radiação , Melanoma Experimental/imunologia , Neoplasias Cutâneas/imunologia , Animais , Antineoplásicos Hormonais/farmacologia , Ativação do Complemento , Complemento C3a/genética , Complemento C5a/genética , Dexametasona/farmacologia , Raios gama , Humanos , Imunidade Inata/efeitos dos fármacos , Melanoma Experimental/genética , Melanoma Experimental/patologia , Melanoma Experimental/radioterapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Complemento/genética , Receptores de Complemento/imunologia , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/radioterapia , Carga Tumoral/efeitos da radiação
18.
Biomed Res Int ; 2015: 479140, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25793196

RESUMO

The use of beneficial microorganisms, the so-called probiotics, to improve human health is gaining popularity. However, not all of the probiotic strains trigger the same responses and they differ in their interaction with the host. In spite of the limited knowledge on mechanisms of action some of the probiotic effects seem to be exerted through maintenance of the gastrointestinal barrier function and modulation of the immune system. In the present work, we have addressed in vitro the response of the intestinal epithelial cell line HT29 to the strain Bifidobacterium breve IPLA20004. In the array of 84 genes involved in inflammation tested, the expression of 12 was modified by the bifidobacteria. The genes of chemokine CXCL6, the chemokine receptor CCR7, and, specially, the complement component C3 were upregulated. Indeed, HT29 cells cocultivated with B. breve produced significantly higher levels of protein C3a. The proteome of HT29 cells showed increased levels of cytokeratin-8 in the presence of B. breve. Altogether, it seems that B. breve IPLA20004 could favor the recruitment of innate immune cells to the mucosa reinforcing, as well as the physical barrier of the intestinal epithelium.


Assuntos
Bifidobacterium/imunologia , Fatores Imunológicos/genética , Fatores Imunológicos/imunologia , Mucosa Intestinal/microbiologia , Proteoma/genética , Proteoma/imunologia , Linhagem Celular Tumoral , Quimiocina CXCL6/genética , Quimiocina CXCL6/imunologia , Complemento C3a/genética , Complemento C3a/imunologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Células HT29 , Humanos , Fatores Imunológicos/metabolismo , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Mucosa Intestinal/imunologia , Queratina-8/genética , Queratina-8/imunologia , Probióticos/metabolismo , Receptores CCR7/genética , Receptores CCR7/imunologia
19.
Exp Hematol ; 43(1): 14-22, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25308956

RESUMO

Mixed chimerism, a phenomenon involved in the development of specific alloantigen tolerance, could be achieved through the transplantation of hematopoietic stem cells into properly prepared recipients. Because the C3a complement component modulates hematopoietic cell trafficking after transplantation, in the present study, we investigated the influence of the C3a deficiency on mixed chimerism and alloantigen tolerance induction. To induce mixed chimerism, C57BL/6J (wild-type strain; H-2K(b); I-E(-)) and B6.129S4-C3(tm1Crr)/J (C3a-deficient) mice were exposed to 3 G total body irradiation (day -1). Subsequently, these mice were treated with CD8-blocking (day -2) and CD40L-blocking (days 0 and 4) antibodies, followed by transplantation with 20 × 10(6) Balb/c (H-2K(d); I-E(+)) bone marrow cells (day 0). The degree of mixed chimerism in peripheral blood leukocytes was measured several times during the 20-week experiment. The tolerance to Balb/c mouse antigens was assessed based on the number of lymphocytes expressing Vß5 and Vß11 T-cell receptor and on skin-graft (day 0) acceptance. Applying our experimental model, mixed chimerism and alloantigen tolerance were effectively induced in C57BL/6J (wild-type) mice, but not in C3a(-/-) animals. The present study is, to our knowledge, the first to demonstrate that C3a is vital for achieving stable mixed chimerism and related to this induction of transplant tolerance.


Assuntos
Quimerismo , Complemento C3a/deficiência , Tolerância ao Transplante/imunologia , Animais , Transplante de Medula Óssea , Complemento C3a/genética , Complemento C3a/imunologia , Feminino , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Sobrevivência de Enxerto , Antígenos H-2/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Isoantígenos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Quimera por Radiação , Receptores de Antígenos de Linfócitos T alfa-beta , Transplante de Pele , Organismos Livres de Patógenos Específicos , Quimeras de Transplante , Tolerância ao Transplante/genética
20.
Mucosal Immunol ; 8(4): 874-85, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25465103

RESUMO

Exposure to cigarette smoke can initiate sterile inflammatory responses in the lung and activate myeloid dendritic cells (mDCs) that induce differentiation of T helper type 1 (Th1) and Th17 cells in the emphysematous lungs. Consumption of complement proteins increases in acute inflammation, but the contribution of complement protein 3 (C3) to chronic cigarette smoke-induced immune responses in the lung is not clear. Here, we show that following chronic exposure to cigarette smoke, C3-deficient (C3(-/-)) mice develop less emphysema and have fewer CD11b(+)CD11c(+) mDCs infiltrating the lungs as compared with wild-type mice. Proteolytic cleavage of C3 by neutrophil elastase releases C3a, which in turn increases the expression of its receptor (C3aR) on lung mDCs. Mice deficient in the C3aR (C3ar(-/-)) partially phenocopy the attenuated responses to chronic smoke observed in C3(-/-) mice. Consistent with a role for C3 in emphysema, C3 and its active fragments are deposited on the lung tissue of smokers with emphysema, and smoke-exposed mice. Together, these findings suggest a critical role for C3a through autocrine/paracrine induction of C3aR in the pathogenesis of cigarette smoke-induced sterile inflammation and provide new therapeutic targets for the treatment of emphysema.


Assuntos
Enfisema/etiologia , Enfisema/metabolismo , Receptores de Complemento/metabolismo , Fumar/efeitos adversos , Animais , Comunicação Autócrina , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Ativação do Complemento , Complemento C3/genética , Complemento C3/imunologia , Complemento C3/metabolismo , Complemento C3a/imunologia , Complemento C3a/metabolismo , Modelos Animais de Doenças , Enfisema/diagnóstico , Regulação da Expressão Gênica , Humanos , Elastase de Leucócito/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Metaloproteinase 12 da Matriz/metabolismo , Camundongos Knockout , Comunicação Parácrina , Proteólise , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA