Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 172(1-2): 162-175.e14, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29328911

RESUMO

Long-term epigenetic reprogramming of innate immune cells in response to microbes, also termed "trained immunity," causes prolonged altered cellular functionality to protect from secondary infections. Here, we investigated whether sterile triggers of inflammation induce trained immunity and thereby influence innate immune responses. Western diet (WD) feeding of Ldlr-/- mice induced systemic inflammation, which was undetectable in serum soon after mice were shifted back to a chow diet (CD). In contrast, myeloid cell responses toward innate stimuli remained broadly augmented. WD-induced transcriptomic and epigenomic reprogramming of myeloid progenitor cells led to increased proliferation and enhanced innate immune responses. Quantitative trait locus (QTL) analysis in human monocytes trained with oxidized low-density lipoprotein (oxLDL) and stimulated with lipopolysaccharide (LPS) suggested inflammasome-mediated trained immunity. Consistently, Nlrp3-/-/Ldlr-/- mice lacked WD-induced systemic inflammation, myeloid progenitor proliferation, and reprogramming. Hence, NLRP3 mediates trained immunity following WD and could thereby mediate the potentially deleterious effects of trained immunity in inflammatory diseases.


Assuntos
Reprogramação Celular , Dieta Ocidental , Epigênese Genética , Imunidade Inata , Memória Imunológica , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Adulto , Idoso , Animais , Células Cultivadas , Feminino , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Células Mieloides/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Locos de Características Quantitativas , Receptores de LDL/genética
2.
Nat Immunol ; 15(8): 727-37, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24952505

RESUMO

Microbes or danger signals trigger inflammasome sensors, which induce polymerization of the adaptor ASC and the assembly of ASC specks. ASC specks recruit and activate caspase-1, which induces maturation of the cytokine interleukin 1ß (IL-1ß) and pyroptotic cell death. Here we found that after pyroptosis, ASC specks accumulated in the extracellular space, where they promoted further maturation of IL-1ß. In addition, phagocytosis of ASC specks by macrophages induced lysosomal damage and nucleation of soluble ASC, as well as activation of IL-1ß in recipient cells. ASC specks appeared in bodily fluids from inflamed tissues, and autoantibodies to ASC specks developed in patients and mice with autoimmune pathologies. Together these findings reveal extracellular functions of ASC specks and a previously unknown form of cell-to-cell communication.


Assuntos
Apoptose/imunologia , Caspase 1/imunologia , Proteínas do Citoesqueleto/imunologia , Inflamação/imunologia , Interleucina-1beta/imunologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Anticorpos/imunologia , Proteínas Reguladoras de Apoptose , Autoanticorpos/imunologia , Doenças Autoimunes/imunologia , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/genética , Caspase 1/genética , Inibidores de Caspase/farmacologia , Comunicação Celular/imunologia , Proteínas do Citoesqueleto/genética , Humanos , Inflamassomos/imunologia , Lisossomos/patologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fagocitose/imunologia , Príons/química , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Transdução de Sinais/imunologia
3.
J Immunol ; 208(12): 2771-2778, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35675954

RESUMO

Thrombin plays a central role in thromboinflammatory responses, but its activity is blocked in the common ex vivo human whole blood models, making an ex vivo study of thrombin effects on thromboinflammatory responses unfeasible. In this study, we exploited the anticoagulant peptide Gly-Pro-Arg-Pro (GPRP) that blocks fibrin polymerization to study the effects of thrombin on acute inflammation in response to Escherichia coli and Staphylococcus aureus Human blood was anticoagulated with either GPRP or the thrombin inhibitor lepirudin and incubated with either E. coli or S. aureus for up to 4 h at 37°C. In GPRP-anticoagulated blood, there were spontaneous elevations in thrombin levels and platelet activation, which further increased in the presence of bacteria. Complement activation and the expression of activation markers on monocytes and granulocytes increased to the same extent in both blood models in response to bacteria. Most cytokines were not elevated in response to thrombin alone, but thrombin presence substantially and heterogeneously modulated several cytokines that increased in response to bacterial incubations. Bacterial-induced releases of IL-8, MIP-1α, and MIP-1ß were potentiated in the thrombin-active GPRP model, whereas the levels of IP-10, TNF, IL-6, and IL-1ß were elevated in the thrombin-inactive lepirudin model. Complement C5-blockade, combined with CD14 inhibition, reduced the overall cytokine release significantly, both in thrombin-active and thrombin-inactive models. Our data support that thrombin itself marginally induces leukocyte-dependent cytokine release in this isolated human whole blood but is a significant modulator of bacteria-induced inflammation by a differential effect on cytokine patterns.


Assuntos
Infecções por Escherichia coli , Infecções Estafilocócicas , Citocinas/metabolismo , Escherichia coli/fisiologia , Humanos , Inflamação , Staphylococcus aureus/metabolismo , Trombina/metabolismo
4.
Pediatr Res ; 94(2): 512-519, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36725909

RESUMO

BACKGROUND: Escherichia coli and Group B streptococci (GBS) are the main causes of neonatal early-onset sepsis (EOS). Despite antibiotic therapy, EOS is associated with high morbidity and mortality. Dual inhibition of complement C5 and the Toll-like receptor co-factor CD14 has in animal studies been a promising novel therapy for sepsis. METHODS: Whole blood was collected from the umbilical cord after caesarean section (n = 30). Blood was anti-coagulated with lepirudin. C5 inhibitor (eculizumab) and anti-CD14 was added 8 min prior to, or 15 and 30 min after adding E. coli or GBS. Total bacterial incubation time was 120 min (n = 16) and 240 min (n = 14). Cytokines and the terminal complement complex (TCC) were measured using multiplex technology and ELISA. RESULTS: Dual inhibition significantly attenuated TCC formation by 25-79% when adding inhibitors with up to 30 min delay in both E. coli- and GBS-induced inflammation. TNF, IL-6 and IL-8 plasma concentration were significantly reduced by 28-87% in E. coli-induced inflammation when adding inhibitors with up to 30 min delay. The dual inhibition did not significantly reduce TNF, IL-6 and IL-8 plasma concentration in GBS-induced inflammation. CONCLUSION: Dual inhibition of C5 and CD14 holds promise as a potential future treatment for severe neonatal EOS. IMPACT: Neonatal sepsis can cause severe host inflammation with high morbidity and mortality, but there are still no effective adjunctive immunologic interventions available. Adding CD14 and complement C5 inhibitors up to 30 min after incubation of E. coli or Group B streptococci in a human umbilical cord blood model significantly reduced complement activation and cytokine release. Dual inhibition of C5 and CD14 is a potential future therapy to modulate systemic inflammation in severe cases of neonatal sepsis.


Assuntos
Sepse Neonatal , Sepse , Gravidez , Animais , Recém-Nascido , Humanos , Feminino , Complemento C5 , Escherichia coli , Sangue Fetal , Interleucina-6 , Interleucina-8 , Cesárea , Citocinas , Inflamação , Receptores de Lipopolissacarídeos
5.
Mol Divers ; 26(4): 2175-2188, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34668104

RESUMO

IFN-ß is a cytokine that plays a significant role in the immune system. Inhibition of IFN-ß might be used as a therapeutic approach to treat septic shock. A peptidomimetic previously developed by our research team, 1-benzyl-5-methyl-4-(n-octylamino)pyrimidin-2(1H)-one (LT87), was used as an cardioprotective agent in a myocardial ischemia (MI) mouse model. We have developed new LT87 derivatives by synthetizing its dimers in an attempt to extend its structural variety and enhance its biological activity. A dimeric derivative, LT127, exhibited a dose-dependent inhibition of LPS-mediated IFN-ß and subsequent CXCL10 mRNA transcription. The effect was selective and transduced through TLR4- and TRAM/TRIF-mediated signaling, with no significant effect on MyD88-dependent signaling. However, this effect was not specific to TLR4, since a similar effect was observed both on TLR8- and MDA5/RIG-I-stimulated IFN-ß expression. Nevertheless, LT127 might serve as a drug candidate, specifically as an inhibitor for IFN-ß production in order to develop a novel therapeutic approach to prevent septic shock.


Assuntos
Interferon beta , Peptidomiméticos , Choque Séptico , Animais , Citocinas/metabolismo , Interferon beta/metabolismo , Camundongos , Peptidomiméticos/farmacologia , Choque Séptico/tratamento farmacológico , Transdução de Sinais , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
6.
PLoS Pathog ; 15(3): e1007684, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30883606

RESUMO

Phagocytosis is a complex process that eliminates microbes and is performed by specialised cells such as macrophages. Toll-like receptor 4 (TLR4) is expressed on the surface of macrophages and recognizes Gram-negative bacteria. Moreover, TLR4 has been suggested to play a role in the phagocytosis of Gram-negative bacteria, but the mechanisms remain unclear. Here we have used primary human macrophages and engineered THP-1 monocytes to show that the TLR4 sorting adapter, TRAM, is instrumental for phagocytosis of Escherichia coli as well as Staphylococcus aureus. We find that TRAM forms a complex with Rab11 family interacting protein 2 (FIP2) that is recruited to the phagocytic cups of E. coli. This promotes activation of the actin-regulatory GTPases Rac1 and Cdc42. Our results show that FIP2 guided TRAM recruitment orchestrates actin remodelling and IRF3 activation, two events that are both required for phagocytosis of Gram-negative bacteria.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Fagocitose/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Transporte/fisiologia , Endocitose , Endossomos , Escherichia coli/patogenicidade , Células HEK293 , Humanos , Fator Regulador 3 de Interferon , Lipopolissacarídeos , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide , Cultura Primária de Células , Transporte Proteico , Transdução de Sinais , Staphylococcus aureus/patogenicidade , Células THP-1 , Receptor 4 Toll-Like/metabolismo , Proteína cdc42 de Ligação ao GTP , Proteínas rab de Ligação ao GTP , Proteínas rac1 de Ligação ao GTP
7.
Nat Immunol ; 10(6): 579-86, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19412184

RESUMO

Toll-like receptor 4 (TLR4) signals the induction of transcription factor IRF3-dependent genes from the early endosome via the adaptor TRAM. Here we report a splice variant of TRAM, TAG ('TRAM adaptor with GOLD domain'), which has a Golgi dynamics domain coupled to TRAM's Toll-interleukin 1 receptor domain. After stimulation with lipopolysaccharide, TRAM and TAG localized to late endosomes positive for the GTPase Rab7a. TAG inhibited activation of IRF3 by lipopolysaccharide. Knockdown of TAG with small interfering RNA enhanced induction of the chemokine CCL5 (RANTES), but not of interleukin 8, by lipopolysaccharide in human peripheral blood mononuclear cells. TAG displaced the adaptor TRIF from TRAM. TAG is therefore an example of a specific inhibitor of the adaptor MyD88-independent pathway activated by TLR4. Targeting TAG could be useful in the effort to boost the immunostimulatory effect of TLR4 without causing unwanted inflammation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Endossomos/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Quimiocina CCL5/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Lipopolissacarídeos/metabolismo , Camundongos , Dados de Sequência Molecular , Fator 88 de Diferenciação Mieloide/metabolismo , Isoformas de Proteínas , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Especificidade por Substrato , Transfecção , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
8.
J Immunol ; 203(6): 1571-1578, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31413105

RESUMO

Heme is a critical danger molecule liberated from hemeproteins in various conditions, including from hemoglobin in hemolytic diseases. Heme may cause thromboinflammatory damage by activating inflammatory and hemostatic pathways, such as complement, the TLRs, coagulation, and platelets. In this study, we explored the effect of single and dual inhibition of complement component C5 and TLR coreceptor CD14 on heme-induced thromboinflammation in an ex vivo human whole blood model. Heme induced a dose-dependent activation of complement via the alternative pathway. Single inhibition of C5 by eculizumab attenuated the release of IL-6, IL-8, TNF, MCP-1, MIP-1α, IFN-γ, LTB-4, MMP-8 and -9, and IL-1Ra with more than 60% (p < 0.05 for all) reduced the upregulation of CD11b on granulocytes and monocytes by 59 and 40%, respectively (p < 0.05), and attenuated monocytic tissue factor expression by 33% (p < 0.001). Blocking CD14 attenuated IL-6 and TNF by more than 50% (p < 0.05). In contrast to single inhibition, combined C5 and CD14 was required for a significantly attenuated prothrombin cleavage (72%, p < 0.05). Markers of thromboinflammation were also quantified in two patients admitted to the hospital with sickle cell disease (SCD) crisis. Both SCD patients had pronounced hemolysis and depleted plasma hemopexin and haptoglobin. Plasma heme and complement activation was markedly increased in one patient, a coinciding observation as demonstrated ex vivo. In conclusion, heme-induced thromboinflammation was largely attenuated by C5 inhibition alone, with a beneficial effect of adding a CD14 inhibitor to attenuate prothrombin activation. Targeting C5 has the potential to reduce thromboinflammation in SCD crisis patients.


Assuntos
Complemento C5/metabolismo , Heme/metabolismo , Inflamação/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Adulto , Anemia Falciforme/metabolismo , Animais , Coagulação Sanguínea/fisiologia , Ativação do Complemento/fisiologia , Citocinas/metabolismo , Granulócitos/metabolismo , Hemólise/fisiologia , Humanos , Masculino , Monócitos/metabolismo , Suínos , Tromboplastina/metabolismo
9.
J Immunol ; 203(4): 853-863, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31270150

RESUMO

Cholesterol crystals (CC) are strong activators of complement and could potentially be involved in thromboinflammation through complement-coagulation cross-talk. To explore the coagulation-inducing potential of CC, we performed studies in lepirudin-based human whole blood and plasma models. In addition, immunohistological examinations of brain thrombi and vulnerable plaque material from patients with advanced carotid atherosclerosis were performed using polarization filter reflected light microscopy to identify CC. In whole blood, CC exposure induced a time- and concentration-dependent generation of prothrombin fragment 1+2 (PTF1.2), tissue factor (TF) mRNA synthesis, and monocyte TF expression. Blocking Abs against TF abolished CC-mediated coagulation, thus indicating involvement of the TF-dependent pathway. Blockade of FXII by corn trypsin inhibitor had a significant inhibitory effect on CC-induced PTF1.2 in platelet-free plasma, although the overall activation potential was low. CC exposure did not induce platelet aggregation, TF microparticle induction, or TF on granulocytes or eosinophils. Inhibition of complement C3 by CP40 (compstatin), C5 by eculizumab, or C5aR1 by PMX53 blocked CC-induced PTF1.2 by 90% and reduced TF+ monocytes from 18-20 to 1-2%. The physiologic relevance was supported by birefringent CC structures adjacent to monocytes (CD14), TF, and activated complement iC3b and C5b-9 in a human brain thrombus. Furthermore, monocyte influx and TF induction in close proximity to CC-rich regions with activated complement were found in a vulnerable plaque. In conclusion, CC could be active, releasable contributors to thrombosis by inducing monocyte TF secondary to complement C5aR1 signaling.


Assuntos
Coagulação Sanguínea/imunologia , Colesterol/imunologia , Ativação do Complemento/imunologia , Receptor da Anafilatoxina C5a/metabolismo , Tromboplastina/biossíntese , Doenças das Artérias Carótidas/imunologia , Doenças das Artérias Carótidas/metabolismo , Humanos , Monócitos/imunologia , Monócitos/metabolismo , Tromboplastina/imunologia , Trombose/imunologia , Trombose/metabolismo
10.
Immunity ; 33(4): 583-96, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20933442

RESUMO

Toll-like receptor 4 (TLR4) is indispensable for recognition of Gram-negative bacteria. We described a trafficking pathway for TLR4 from the endocytic recycling compartment (ERC) to E. coli phagosomes. We found a prominent colocalization between TLR4 and the small GTPase Rab11a in the ERC, and Rab11a was involved in the recruitment of TLR4 to phagosomes in a process requiring TLR4 signaling. Also, Toll-receptor-associated molecule (TRAM) and interferon regulatory factor-3 (IRF3) localized to E. coli phagosomes and internalization of E. coli was required for a robust interferon-ß induction. Suppression of Rab11a reduced TLR4 in the ERC and on phagosomes leading to inhibition of the IRF3 signaling pathway induced by E. coli, whereas activation of the transcription factor NF-κB was unaffected. Moreover, Rab11a silencing reduced the amount of TRAM on phagosomes. Thus, Rab11a is an important regulator of TLR4 and TRAM transport to E. coli phagosomes thereby controlling IRF3 activation from this compartment.


Assuntos
Fagossomos/metabolismo , Receptor 4 Toll-Like/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia , Endocitose , Escherichia coli/imunologia , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/biossíntese , Fagocitose , Transdução de Sinais , Staphylococcus aureus/imunologia
11.
J Immunol ; 199(8): 2910-2920, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28855312

RESUMO

Cholesterol crystals (CC) are abundant in atherosclerotic plaques and promote inflammatory responses via the complement system and inflammasome activation. Cyclic oligosaccharide 2-hydroxypropyl-ß-cyclodextrin (BCD) is a compound that solubilizes lipophilic substances. Recently we have shown that BCD has an anti-inflammatory effect on CC via suppression of the inflammasome and liver X receptor activation. The putative effects of BCD on CC-induced complement activation remain unknown. In this study, we found that BCD bound to CC and reduced deposition of Igs, pattern recognition molecules, and complement factors on CC in human plasma. Furthermore, BCD decreased complement activation as measured by terminal complement complex and lowered the expression of complement receptors on monocytes in whole blood in response to CC exposure. In line with this, BCD also reduced reactive oxygen species formation caused by CC in whole blood. Furthermore, BCD attenuated the CC-induced proinflammatory cytokine responses (e.g., IL-1α, MIP-1α, TNF, IL-6, and IL-8) as well as regulated a range of CC-induced genes in human PBMC. BCD also regulated complement-related genes in human carotid plaques treated ex vivo. Formation of terminal complement complex on other complement-activating structures such as monosodium urate crystals and zymosan was not affected by BCD. These data demonstrate that BCD inhibits CC-induced inflammatory responses, which may be explained by BCD-mediated attenuation of complement activation. Thus, these findings support the potential for using BCD in treatment of atherosclerosis.


Assuntos
Artérias Carótidas/fisiologia , Colesterol/metabolismo , Ciclodextrinas/metabolismo , Inflamação/imunologia , Leucócitos Mononucleares/fisiologia , Monócitos/fisiologia , Placa Aterosclerótica/imunologia , Células Cultivadas , Colesterol/imunologia , Ativação do Complemento , Proteínas do Sistema Complemento/biossíntese , Ciclodextrinas/química , Citocinas/metabolismo , Humanos , Imunomodulação , Inflamação/induzido quimicamente , Mediadores da Inflamação/metabolismo , Placa Aterosclerótica/terapia , Espécies Reativas de Oxigênio/metabolismo
12.
J Biol Chem ; 292(37): 15408-15425, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28717003

RESUMO

Toll-like receptors (TLRs) are innate immune receptors for sensing microbial molecules and damage-associated molecular patterns released from host cells. Double-stranded RNA and the synthetic analog polyinosinic:polycytidylic acid (poly(I:C)) bind and activate TLR3. This stimulation leads to recruitment of the adaptor molecule TRIF (Toll/IL-1 resistance (TIR) domain-containing adapter-inducing interferon ß) and activation of the transcription factors nuclear factor κB (NF-κB) and interferon regulatory factor 3 (IRF-3), classically inducing IFNß production. Here we report that, unlike non-metastatic intestinal epithelial cells (IECs), metastatic IECs express TLR3 and that TLR3 promotes invasiveness of these cells. In response to poly(I:C) addition, the metastatic IECs also induced the chemokine CXCL10 in a TLR3-, TRIF-, and IRF3-dependent manner but failed to produce IFNß. This was in contrast to healthy and non-metastatic IECs, which did not respond to poly(I:C) stimulation. Endolysosomal acidification and the endosomal transporter protein UNC93B1 was required for poly(I:C)-induced CXCL10 production. However, TLR3-induced CXCL10 was triggered by immobilized poly(I:C), was only modestly affected by inhibition of endocytosis, and could be blocked with an anti-TLR3 antibody, indicating that TLR3 can still signal from the cell surface of these cells. Furthermore, plasma membrane fractions from metastatic IECs contained both full-length and cleaved TLR3, demonstrating surface expression of both forms of TLR3. Our results imply that metastatic IECs express surface TLR3, allowing it to sense extracellular stimuli that trigger chemokine responses and promote invasiveness in these cells. We conclude that altered TLR3 expression and localization may have implications for cancer progression.


Assuntos
Quimiocina CXCL10/agonistas , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/metabolismo , Proteínas de Neoplasias/agonistas , Receptor 3 Toll-Like/agonistas , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Transporte/toxicidade , Linhagem Celular , Linhagem Celular Tumoral , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citocinas/agonistas , Citocinas/genética , Citocinas/metabolismo , Endocitose/efeitos dos fármacos , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Neoplasias Intestinais/tratamento farmacológico , Neoplasias Intestinais/imunologia , Neoplasias Intestinais/patologia , Ligantes , Lipopolissacarídeos/toxicidade , Invasividade Neoplásica/imunologia , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Poli I-C , Polinucleotídeos/toxicidade , Regiões Promotoras Genéticas/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Interferência de RNA , Receptor 3 Toll-Like/antagonistas & inibidores , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo
13.
J Immunol ; 196(5): 2293-9, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26800874

RESUMO

Endothelial cells (EC) play a central role in inflammation. E-selectin and ICAM-1 expression are essential for leukocyte recruitment and are good markers of EC activation. Most studies of EC activation are done in vitro using isolated mediators. The aim of the present study was to examine the relative importance of pattern recognition systems and downstream mediators in bacteria-induced EC activation in a physiological relevant human model, using EC incubated with whole blood. HUVEC were incubated with human whole blood. Escherichia coli- and Staphylococcus aureus-induced EC activation was measured by E-selectin and ICAM-1 expression using flow cytometry. The mAb 18D11 was used to neutralize CD14, and the lipid A analog eritoran was used to block TLR4/MD2. C5 cleavage was inhibited using eculizumab, and C5aR1 was blocked by an antagonist. Infliximab and canakinumab were used to neutralize TNF and IL-1ß. The EC were minimally activated when bacteria were incubated in serum, whereas a substantial EC activation was seen when the bacteria were incubated in whole blood. E. coli-induced activation was largely CD14-dependent, whereas S. aureus mainly caused a C5aR1-mediated response. Combined CD14 and C5 inhibition reduced E-selectin and ICAM-1 expression by 96 and 98% for E. coli and by 70 and 75% for S. aureus. Finally, the EC activation by both bacteria was completely abolished by combined inhibition of TNF and IL-1ß. E. coli and S. aureus activated EC in a CD14- and C5-dependent manner with subsequent leukocyte secretion of TNF and IL-1ß mediating the effect.


Assuntos
Ativação do Complemento/imunologia , Complemento C5/imunologia , Células Endoteliais/metabolismo , Escherichia coli/imunologia , Interleucina-1beta/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Staphylococcus aureus/imunologia , Fatores de Necrose Tumoral/metabolismo , Anticorpos Monoclonais/farmacologia , Biomarcadores , Células Cultivadas , Complemento C5/antagonistas & inibidores , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Antígeno 96 de Linfócito/antagonistas & inibidores , Antígeno 96 de Linfócito/metabolismo
14.
J Immunol ; 196(12): 5064-74, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27183610

RESUMO

Cholesterol crystals (CC) play an essential role in the formation of atherosclerotic plaques. CC activate the classical and the alternative complement pathways, but the role of the lectin pathway is unknown. We hypothesized that the pattern recognition molecules (PRMs) from the lectin pathway bind CC and function as an upstream innate inflammatory signal in the pathophysiology of atherosclerosis. We investigated the binding of the PRMs mannose-binding lectin (MBL), ficolin-1, ficolin-2, and ficolin-3, the associated serine proteases, and complement activation products to CC in vitro using recombinant proteins, specific inhibitors, as well as deficient and normal sera. Additionally, we examined the deposition of ficolin-2 and MBL in human carotid plaques by immunohistochemistry and fluorescence microscopy. The results showed that the lectin pathway was activated on CC by binding of ficolin-2 and MBL in vitro, resulting in activation and deposition of complement activation products. MBL bound to CC in a calcium-dependent manner whereas ficolin-2 binding was calcium-independent. No binding was observed for ficolin-1 or ficolin-3. MBL and ficolin-2 were present in human carotid plaques, and binding of MBL to CC was confirmed in vivo by immunohistochemistry, showing localization of MBL around CC clefts. Moreover, we demonstrated that IgM, but not IgG, bound to CC in vitro and that C1q binding was facilitated by IgM. In conclusion, our study demonstrates that PRMs from the lectin pathway recognize CC and provides evidence for an important role for this pathway in the inflammatory response induced by CC in the pathophysiology of atherosclerosis.


Assuntos
Aterosclerose/imunologia , Aterosclerose/fisiopatologia , Ativação do Complemento , Progressão da Doença , Lectinas/metabolismo , Lectina de Ligação a Manose/metabolismo , Cálcio/metabolismo , Estenose das Carótidas/imunologia , Colesterol/química , Colesterol/imunologia , Colesterol/metabolismo , Colesterol/farmacologia , Complemento C4/metabolismo , Cristalização , Imunofluorescência , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunoglobulina M/imunologia , Imunoglobulina M/metabolismo , Inflamação , Lectinas/imunologia , Lectina de Ligação a Manose/imunologia , Microscopia de Fluorescência , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Proteínas Recombinantes/metabolismo , Ficolinas
15.
Traffic ; 16(7): 677-90, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25707286

RESUMO

Toll-like receptor 4 (TLR4) is responsible for the immediate response to Gram-negative bacteria and signals via two main pathways by recruitment of distinct pairs of adaptor proteins. Mal-MyD88 [Mal (MyD88-adaptor-like) - MYD88 (Myeloid differentiation primary response gene (88))] is recruited to the plasma membrane to initiate the signaling cascade leading to production of pro-inflammatory cytokines while TRAM-TRIF [TRAM (TRIF-related adaptor molecule)-TRIF (TIR-domain-containing adapter-inducing interferon-ß)] is recruited to early endosomes to initiate the subsequent production of type I interferons. We have investigated the dynamics of TLR4 and TRAM during lipopolysaccharide (LPS) stimulation. We found that LPS induced a CD14-dependent immobile fraction of TLR4 in the plasma membrane. Total internal reflection fluorescence microscopy (TIRF) revealed that LPS stimulation induced clustering of TLR4 into small punctate structures in the plasma membrane containing CD14/LPS and clathrin, both in HEK293 cells and the macrophage model cell line U373-CD14. These results suggest that laterally immobilized TLR4 receptor complexes are being formed and prepared for endocytosis. RAB11A was found to be involved in localizing TRAM to the endocytic recycling compartment (ERC) and to early sorting endosomes. Moreover, CD14/LPS but not TRAM was immobilized on RAB11A-positive endosomes, which indicates that TRAM and CD14/LPS can independently be recruited to endosomes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Endocitose , Receptores de Lipopolissacarídeos/metabolismo , Receptor 4 Toll-Like/metabolismo , Membrana Celular/metabolismo , Clatrina/metabolismo , Endossomos/metabolismo , Células HEK293 , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Transporte Proteico , Proteínas rab de Ligação ao GTP
16.
Biochem Biophys Res Commun ; 491(2): 442-448, 2017 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-28712870

RESUMO

Endoplasmic reticulum (ER) stress has been shown to play a key role during the initiation and clinical progression of the cardiovascular diseases, such as atherosclerosis. We have recently shown that expression of tissue factor pathway inhibitor (TFPI) in human monocyte-derived macrophages (MDMs) was induced by cholesterol crystals (CC). In the present study we aimed to determine the role of TFPI under ER stress conditions using human MDMs. qRT-PCR and immunohistochemistry analysis were performed to determine the presence of the ER stress marker CCAAT/enhancer binding protein homologous protein (CHOP) and TFPI in human carotid plaque material and also in human MDMs polarized into pro-inflammatory M1 or anti-inflammatory M2 populations. CHOP mRNA levels were upregulated in the plaques compared to healthy vessels, and CHOP protein was localized in the same area as TFPI in the plaques. Both CHOP and TFPI mRNA levels were upregulated after CC treatment, especially in the M2 phenotype, and the ER stress inhibitor 4-phenylbutyric acid (PBA) reversed this effect. Furthermore, CC treatment increased the levels of the pro-inflammatory cytokines TNF-α, IL-6, and IL-8, which for TNF-α and IL-8 was inhibited by PBA, and reduced the levels of the anti-inflammatory cytokine IL-10 in M2-polarized macrophages. Knockdown of TFPI prior to CC treatment exacerbated TNF-α and IL-6 levels, but reduced IL-8 and IL-10 levels. Our results show that CC induce TFPI and cytokine expression in M2-polarized macrophages through activation of the ER stress pathway and that TFPI has a protective effect against TNF-α and IL-6 mediated inflammation. These mechanisms may have implications for the pathogenesis of atherosclerosis.


Assuntos
Aterosclerose/genética , Colesterol/farmacologia , Estresse do Retículo Endoplasmático/genética , Lipoproteínas/genética , Placa Aterosclerótica/genética , RNA Mensageiro/genética , Aterosclerose/imunologia , Aterosclerose/patologia , Aterosclerose/cirurgia , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/imunologia , Artérias Carótidas/patologia , Artérias Carótidas/cirurgia , Cristalização , Endarterectomia das Carótidas , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Interleucina-8/genética , Interleucina-8/imunologia , Lipoproteínas/antagonistas & inibidores , Lipoproteínas/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Fenilbutiratos/farmacologia , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia , Placa Aterosclerótica/cirurgia , Cultura Primária de Células , RNA Mensageiro/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
17.
J Immunol ; 195(3): 1100-11, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26085680

RESUMO

Staphylococcus aureus may cause serious infections and is one of the most lethal and common causes of sepsis. TLR2 has been described as the main pattern recognition receptor that senses S. aureus and elicits production of proinflammatory cytokines via MyD88 -: NF-κB signaling. S. aureus can also induce the production of IFN-ß, a cytokine that requires IFN regulatory factors (IRFs) for its transcription, but the signaling mechanism for IFN-ß induction by S. aureus are unclear. Surprisingly, we demonstrate that activation of TLR2 by lipoproteins does not contribute to IFN-ß production but instead can suppress the induction of IFN-ß in human primary monocytes and monocyte-derived macrophages. The production of IFN-ß was induced by TLR8-mediated sensing of S. aureus RNA, which triggered IRF5 nuclear accumulation, and this could be antagonized by concomitant TLR2 signaling. The TLR8-mediated activation of IRF5 was dependent on TAK1 and IκB kinase (IKK)ß, which thus reveals a physiological role of the recently described IRF5-activating function of IKKß. TLR8 -: IRF5 signaling was necessary for induction of IFN-ß and IL-12 by S. aureus, and it also contributed to the induction of TNF. In conclusion, our study demonstrates a physiological role of TLR8 in the sensing of entire S. aureus in human primary phagocytes, including the induction of IFN-ß and IL-12 production via a TAK1 -: IKKß -: IRF5 pathway that can be inhibited by TLR2 signaling.


Assuntos
Fatores Reguladores de Interferon/imunologia , Interferon beta/biossíntese , Interleucina-12/biossíntese , RNA Bacteriano/imunologia , Staphylococcus aureus/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia , Ativação Enzimática/imunologia , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/imunologia , Fatores Reguladores de Interferon/genética , Interferon beta/imunologia , Interleucina-12/imunologia , MAP Quinase Quinase Quinases/imunologia , Macrófagos/imunologia , Proteínas de Membrana/genética , Monócitos/imunologia , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , RNA Bacteriano/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética , Fator de Necrose Tumoral alfa/biossíntese
18.
J Immunol ; 195(1): 257-64, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26026058

RESUMO

Chronic inflammation of the arterial wall is a key element in the development of atherosclerosis, and cholesterol crystals (CC) that accumulate in plaques are associated with initiation and progression of the disease. We recently revealed a link between the complement system and CC-induced inflammasome caspase-1 activation, showing that the complement system is a key trigger in CC-induced inflammation. HDL exhibits cardioprotective and anti-inflammatory properties thought to explain its inverse correlation to cardiovascular risk. In this study, we sought to determine the effect of reconstituted HDL (rHDL) on CC-induced inflammation in a human whole blood model. rHDL bound to CC and inhibited the CC-induced complement activation as measured by soluble terminal C5b-9 formation and C3c deposition on the CC surface. rHDL attenuated the amount of CC-induced complement receptor 3 (CD11b/CD18) expression on monocytes and granulocytes, as well as reactive oxygen species generation. Moreover, addition of CC to whole blood resulted in release of proinflammatory cytokines that were inhibited by rHDL. Our results support and extend the notion that CC are potent triggers of inflammation, and that rHDL may have a beneficial role in controlling the CC-induced inflammatory responses by inhibiting complement deposition on the crystals.


Assuntos
Colesterol/efeitos adversos , Ativação do Complemento/efeitos dos fármacos , Lipoproteínas HDL/farmacologia , Células Sanguíneas/citologia , Células Sanguíneas/efeitos dos fármacos , Células Sanguíneas/imunologia , Antígeno CD11b/imunologia , Antígenos CD18/imunologia , Complemento C3c/antagonistas & inibidores , Complemento C3c/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Cristalização , Humanos , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Cultura Primária de Células , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/imunologia
19.
Eur Heart J ; 37(30): 2406-13, 2016 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-27161611

RESUMO

AIMS: Interleukin-6 (IL-6) contributes to atherosclerotic plaque destabilization and is involved in myocardial injury during ischaemia-reperfusion. Interleukin-6 is therefore a potential therapeutic target in myocardial infarction (MI). We hypothesized that the IL-6 receptor antagonist tocilizumab would attenuate inflammation, and secondarily reduce troponin T (TnT) release in non-ST-elevation MI (NSTEMI). METHODS AND RESULTS: In a two-centre, double-blind, placebo-controlled trial, 117 patients with NSTEMI were randomized at a median of 2 days after symptom onset to receive placebo (n = 59) or tocilizumab (n = 58), administered as a single dose prior to coronary angiography. High sensitivity (hs) C-reactive protein and hsTnT were measured at seven consecutive timepoints between Days 1 and 3. The area under the curve (AUC) for high-sensitivity C-reactive protein was the primary endpoint. The median AUC for high-sensitivity C-reactive protein during hospitalization was 2.1 times higher in the placebo than in the tocilizumab group (4.2 vs. 2.0 mg/L/h, P < 0.001). Also, the median AUC for hsTnT during hospitalization was 1.5 times higher in the placebo group compared with the tocilizumab group (234 vs. 159 ng/L/h, P = 0.007). The differences between the two treatment groups were observed mainly in (i) patients included ≤2 days from symptom onset and (ii) patients treated with percutaneous coronary intervention (PCI). No safety issues in the tocilizumab group were detected during 6 months of follow-up. CONCLUSION: Tocilizumab attenuated the inflammatory response and primarily PCI-related TnT release in NSTEMI patients.


Assuntos
Infarto do Miocárdio sem Supradesnível do Segmento ST , Anticorpos Monoclonais Humanizados , Método Duplo-Cego , Humanos , Inflamação , Receptores de Interleucina-6 , Troponina T
20.
J Infect Dis ; 214(1): 140-50, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26977050

RESUMO

BACKGROUND: Single inhibition of the Toll-like receptor 4 (TLR4)-MD2 complex failed in treatment of sepsis. CD14 is a coreceptor for several TLRs, including TLR4 and TLR2. The aim of this study was to investigate the effect of single TLR4-MD2 inhibition by using eritoran, compared with the effect of CD14 inhibition alone and combined with the C3 complement inhibitor compstatin (Cp40), on the bacteria-induced inflammatory response in human whole blood. METHODS: Cytokines were measured by multiplex technology, and leukocyte activation markers CD11b and CD35 were measured by flow cytometry. RESULTS: Lipopolysaccharide (LPS)-induced inflammatory markers were efficiently abolished by both anti-CD14 and eritoran. Anti-CD14 was significantly more effective than eritoran in inhibiting LPS-binding to HEK-293E cells transfected with CD14 and Escherichia coli-induced upregulation of monocyte activation markers (P < .01). Combining Cp40 with anti-CD14 was significantly more effective than combining Cp40 with eritoran in reducing E. coli-induced interleukin 6 (P < .05) and monocyte activation markers induced by both E. coli (P < .001) and Staphylococcus aureus (P < .01). Combining CP40 with anti-CD14 was more efficient than eritoran alone for 18 of 20 bacteria-induced inflammatory responses (mean P < .0001). CONCLUSIONS: Whole bacteria-induced inflammation was inhibited more efficiently by anti-CD14 than by eritoran, particularly when combined with complement inhibition. Combined CD14 and complement inhibition may prove a promising treatment strategy for bacterial sepsis.


Assuntos
Antibacterianos/uso terapêutico , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/microbiologia , Sepse/tratamento farmacológico , Infecções Estafilocócicas/complicações , Infecções Estafilocócicas/tratamento farmacológico , Citocinas/sangue , Escherichia coli/efeitos dos fármacos , Humanos , Inflamação/sangue , Receptores de Lipopolissacarídeos/sangue , Receptores de Lipopolissacarídeos/efeitos dos fármacos , Sepse/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Receptor 4 Toll-Like/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa