Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 274
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 205(5): 1385-1392, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32759297

RESUMO

ß2-Glycoprotein I (ß2-GPI) is an abundant plasma glycoprotein with unknown physiological function and is currently recognized as the main target of antiphospholipid Abs responsible for complement activation and vascular thrombosis in patients with antiphospholipid syndrome (APS). In this study, we provide evidence that mannose-binding lectin (MBL) binds to ß2-GPI in Ca++ and a dose-dependent manner and that this interaction activates complement and promotes complement-dependent thrombin generation. Surprisingly, a significant binding was observed between MBL and isolated domains II and IV of ß2-GPI, whereas the carbohydrate chains, domain I and domain V, were not involved in the interaction, documenting a noncanonical binding mode between MBL and ß2-GPI. Importantly, this interaction may occur on endothelial cells because binding of MBL to ß2-GPI was detected on the surface of HUVECs, and colocalization of MBL with ß2-GPI was observed on the endothelium of a biopsy specimen of a femoral artery from an APS patient. Because ß2-GPI-mediated MBL-dependent thrombin generation was increased after priming the endothelium with TNF-α, our data suggests that this mechanism could play an important yet unrecognized role under physiological conditions and may be upregulated in pathological situations. Moreover, the complement activation and the procoagulant effects of the ß2-GPI/MBL complex may contribute to amplify similar activities of anti-ß2-GPI Abs in APS and possibly act independently of Abs, raising the issue of developing appropriate therapies to avoid recurrences and disability in patients at risk for these clinical conditions.


Assuntos
Ativação do Complemento/imunologia , Lectina de Ligação a Manose/metabolismo , Trombina/metabolismo , beta 2-Glicoproteína I/metabolismo , Síndrome Antifosfolipídica/imunologia , Síndrome Antifosfolipídica/metabolismo , Cálcio/metabolismo , Linhagem Celular , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Endotélio/imunologia , Endotélio/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Lectina de Ligação a Manose/imunologia , Ligação Proteica/imunologia , Trombina/imunologia , Trombose/imunologia , Trombose/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , beta 2-Glicoproteína I/imunologia
2.
Mikrochim Acta ; 189(1): 32, 2021 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-34932168

RESUMO

The design of a sandwich-type SERS immunoassay (surface-enhanced Raman spectroscopy) is demonstrated operating in dual surface enhancement and dual-tag paradigm. The capture and detection antibodies are linked to two SERS-active substrates and form together the three-dimensional (3D) structure after specific binding to interleukin 6. A variety of metal combinations is tested (Au-Ag, Au-Au, and Ag-Ag), but an enhanced electromagnetic field is generated only due to coupling of Ag and Au nanoparticles with an Au hexagonal nanoarray. The amplified in that way Raman signals improve the limit of detection over 3 times in comparison to the assay with only one SERS-active substrate. It is also shown that the proper readout of the true-positive signal can be achieved in assays with two Raman tags, and this approach also improves LOD. For the optimal combination of the metal-metal junction and Raman tags, a linear relationship between the Raman signal and the concentration of IL-6 is obtained in the range 0-1000 pg⋅mL-1with LOD of 25.2 pg mL-1and RSD < 10%. The presented proof-of-concept of the SERS immunoassay with the dual-enhancement and dual-tag opens additional opportunities for engineering reliable SERS biosensing.


Assuntos
Imunoensaio/métodos , Interleucina-6/análise , Nanopartículas Metálicas/química , Análise Espectral Raman/métodos , Anticorpos Imobilizados/imunologia , Ouro/química , Humanos , Interleucina-6/imunologia , Limite de Detecção , Prata/química , Trombina/análise , Trombina/imunologia
3.
Paediatr Respir Rev ; 35: 20-24, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32653469

RESUMO

Since the initial description in 2019, the novel coronavirus SARS-Cov-2 infection (COVID-19) pandemic has swept the globe. The most severe form of the disease presents with fever and shortness of breath, which rapidly deteriorates to respiratory failure and acute lung injury (ALI). COVID-19 also presents with a severe coagulopathy with a high rate of venous thromboembiolism. In addition, autopsy studies have revealed co-localized thrombosis and inflammation, which is the signature of thromboinflammation, within the pulmonary capillary vasculature. While the majority of published data is on adult patients, there are parallels to pediatric patients. In our experience as a COVID-19 epicenter, children and young adults do develop both the coagulopathy and the ALI of COVID-19. This review will discuss COVID-19 ALI from a hematological perspective with discussion of the distinct aspects of coagulation that are apparent in COVID-19. Current and potential interventions targeting the multiple thromboinflammatory mechanisms will be discussed.


Assuntos
Lesão Pulmonar Aguda/sangue , Infecções por Coronavirus/sangue , Inflamação/sangue , Pneumonia Viral/sangue , Trombose/sangue , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/fisiopatologia , Anticoagulantes/uso terapêutico , Antitrombinas/uso terapêutico , Betacoronavirus , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/imunologia , Transtornos da Coagulação Sanguínea/fisiopatologia , COVID-19 , Capilares/imunologia , Capilares/fisiopatologia , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/fisiopatologia , Endotélio Vascular/imunologia , Endotélio Vascular/fisiopatologia , Inibidores do Fator Xa/uso terapêutico , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/fisiopatologia , Pandemias , Ativação Plaquetária , Inibidores da Agregação Plaquetária/uso terapêutico , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/imunologia , Pneumonia Viral/fisiopatologia , Embolia Pulmonar/sangue , Embolia Pulmonar/imunologia , Embolia Pulmonar/fisiopatologia , SARS-CoV-2 , Trombina/imunologia , Trombina/metabolismo , Trombose/tratamento farmacológico , Trombose/imunologia , Trombose/fisiopatologia , Tratamento Farmacológico da COVID-19
4.
J Thromb Thrombolysis ; 49(3): 446-450, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32036558

RESUMO

The anti-phospholipid syndrome (APS) is defined by the laboratory detection of at least one of three anti-phospholipid autoantibodies (lupus anticoagulant, or anti-cardiolipin or anti-ß2-glycoprotein I antibodies) in the clinical setting of thrombosis or pregnancy morbidity in a patient. Recognising APS and administering appropriate secondary thromboprophylaxis is important to reduce risk of recurrent thrombosis and/or pregnancy morbidity. In some instances, patients having clinical manifestations highly suggestive of APS are persistently negative for these antibodies but instead have other autoantibodies. Autoantibodies directed against prothrombin (PT) have been associated with increased thrombotic risk and comprise anti-prothrombin (aPT) and anti-phosphatidylserine/prothrombin (aPS/PT) antibodies. Detection of aPT and aPS/PT may help stratify patients for more effective treatment, however, their prevalence among patients with unprovoked venous thromboembolism (VTE) is unknown and determination of their frequencies is the objective of this study. Sera from 148 patients with unprovoked VTE were analysed. Autoantibodies directed against PT collectively, aPT and aPS/PT were present in 24.3%, 14.9% and 13.5%, respectively. Prevalence of these autoantibodies in unprovoked VTE is much lower compared to cohorts comprising mainly patients with systemic autoimmune disorders. Detection of these autoantibodies in unprovoked VTE has potential therapeutic implications for patients including the duration of anticoagulation and administration, or otherwise, of direct oral anticoagulants. Data from this study will assist in the design of future clinical studies to estimate risk of recurrent VTE and to determine optimal management.


Assuntos
Autoanticorpos , Trombina , Tromboembolia Venosa , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , Estudos Retrospectivos , Trombina/antagonistas & inibidores , Trombina/imunologia , Trombina/metabolismo , Tromboembolia Venosa/sangue , Tromboembolia Venosa/epidemiologia , Tromboembolia Venosa/imunologia
5.
Proc Natl Acad Sci U S A ; 114(21): E4213-E4222, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28473418

RESUMO

Effective control of endotoxins and bacteria is crucial for normal wound healing. During injury, the key enzyme thrombin is formed, leading to generation of fibrin. Here, we show that human neutrophil elastase cleaves thrombin, generating 11-kDa thrombin-derived C-terminal peptides (TCPs), which bind to and form amorphous amyloid-like aggregates with both bacterial lipopolysaccharide (LPS) and gram-negative bacteria. In silico molecular modeling using atomic resolution and coarse-grained simulations corroborates our experimental observations, altogether indicating increased aggregation through LPS-mediated intermolecular contacts between clusters of TCP molecules. Upon bacterial aggregation, recombinantly produced TCPs induce permeabilization of Escherichia coli and phagocytic uptake. TCPs of about 11 kDa are present in acute wound fluids as well as in fibrin sloughs from patients with infected wounds. We noted aggregation and colocalization of LPS with TCPs in such fibrin material, which indicates the presence of TCP-LPS aggregates under physiological conditions. Apart from identifying a function of proteolyzed thrombin and its fragments, our findings provide an interesting link between the coagulation system, innate immunity, LPS scavenging, and protein aggregation/amyloid formation.


Assuntos
Escherichia coli/imunologia , Lipopolissacarídeos/imunologia , Fragmentos de Peptídeos/imunologia , Agregados Proteicos/imunologia , Trombina/imunologia , Animais , Linhagem Celular , Humanos , Imunidade Inata/imunologia , Elastase de Leucócito/metabolismo , Camundongos , Células RAW 264.7 , Trombina/metabolismo , Ferimentos e Lesões/imunologia , Ferimentos e Lesões/microbiologia
6.
J Biol Chem ; 293(35): 13578-13591, 2018 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-30002122

RESUMO

Some strains of the bacterial pathogen Streptococcus pyogenes secrete protein SIC (streptococcal inhibitor of complement), including strains of the clinically relevant M1 serotype. SIC neutralizes the effect of a number of antimicrobial proteins/peptides and interferes with the function of the host complement system. Previous studies have shown that some S. pyogenes proteins bind and modulate coagulation and fibrinolysis factors, raising the possibility that SIC also may interfere with the activity of these factors. Here we show that SIC interacts with both human thrombin and plasminogen, key components of coagulation and fibrinolysis. We found that during clot formation, SIC binds fibrin through its central region and that SIC inhibits fibrinolysis by interacting with plasminogen. Flow cytometry results indicated that SIC and plasminogen bind simultaneously to S. pyogenes bacteria, and fluorescence microscopy revealed co-localization of the two proteins at the bacterial surface. As a consequence, SIC-expressing bacteria entrapped in clots inhibit fibrinolysis, leading to delayed bacterial escape from the clots as compared with mutant bacteria lacking SIC. Moreover, within the clots SIC-expressing bacteria were protected against killing. In an animal model of subcutaneous infection, SIC-expressing bacteria exhibited a delayed systemic spread. These results demonstrate that the bacterial protein SIC interferes with coagulation and fibrinolysis and thereby enhances bacterial survival, a finding that has significant implications for S. pyogenes virulence.


Assuntos
Proteínas de Bactérias/imunologia , Fibrinólise , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Trombose/imunologia , Animais , Proteínas do Sistema Complemento/imunologia , Feminino , Fibrina/imunologia , Fibrinogênio/imunologia , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Infecções Estreptocócicas/complicações , Infecções Estreptocócicas/microbiologia , Trombina/imunologia , Trombose/complicações , Trombose/microbiologia
7.
Blood ; 130(24): 2678-2681, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29021229

RESUMO

Sepsis concurrently activates both coagulation and complement systems. Although complement activation by bacteria is well documented, work in mice and in vitro suggests that coagulation proteases can directly cleave complement proteins. We aimed to determine whether generation of coagulation proteases in vivo can activate the complement cascade in 2 highly coagulopathic models. We compared temporal changes in activation biomarkers of coagulation (thrombin-antithrombin [TAT]), fibrinolysis (plasmin-antiplasmin [PAP]), and complement (C3b, C5a, C5b-9) in baboons infused with factor Xa (FXa) and phospholipids (FXa/phosphatidylcholine-phosphatidylserine [PCPS]) vs LD100 Escherichia coli We found that, albeit with different timing, both FXa/PCPS and E coli infusion led to robust thrombin and plasmin generation. Conversely, only E coli challenge activated the complement system, reaching a maximum at 2 hours postchallenge during the peaks of lipopolysaccharide and bacteremia but not of TAT and PAP. Despite inducing a strong burst of thrombin and plasmin, FXa/PCPS infusion did not produce measurable levels of complement activation in vivo. Similarly, ex vivo incubation of baboon serum with thrombin, plasmin, or FXa did not show noticeable complement cleavage unless supraphysiologic amounts of enzymes were used. Our results suggest that in vivo-generated thrombin and plasmin do not directly activate the complement in nonhuman primates.


Assuntos
Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Fibrinolisina/imunologia , Trombina/imunologia , Animais , Ativação do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Escherichia coli/imunologia , Escherichia coli/metabolismo , Fator Xa/imunologia , Fator Xa/farmacologia , Fibrinolisina/metabolismo , Humanos , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Papio , Fosfatidilcolinas/imunologia , Fosfatidilcolinas/farmacologia , Fosfatidilserinas/imunologia , Fosfatidilserinas/farmacologia , Trombina/metabolismo
8.
Clin Immunol ; 180: 25-32, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28366510

RESUMO

The terminal complement split product C5a has been described as an important mediator in inflammatory diseases. C5a is generated upon cleavage of C5 and earlier research suggests that, besides the known C5 convertases formed upon activation of the complement pathways, various enzymes could activate C5 directly. We demonstrate that eculizumab effectively blocks C5 activation when mediated by C5-convertase formation, but fails to block C5a generation resulting from direct enzymatic cleavage by trypsin and thrombin. C5a generated by these enzymes is shown to be fully biologically functional and can be blocked by IFX-1, a specific monoclonal anti-human C5a antibody. We further report clinical cases of atypical hemolytic uremic syndrome (aHUS) and C3 Glomerulonephritis (C3GN) patients under treatment with eculizumab presenting substantially elevated C5a levels. Thus, blocking the C5 convertase mediated activation of C5 may not be efficient to control C5a-mediated effects in human disease and that a targeted approach is warranted.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C5a/imunologia , Glomerulonefrite/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Convertases de Complemento C3-C5/imunologia , Glomerulonefrite/tratamento farmacológico , Humanos , Trombina/imunologia , Tripsina/imunologia , Zimosan/farmacologia
9.
Pharmacol Res ; 121: 33-41, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28428115

RESUMO

Asthma and chronic obstructive pulmonary disease (COPD) are common chronic lung inflammatory diseases. Thrombin and interleukin (IL)-8/C-X-C chemokine ligand 8 (CXCL8) play critical roles in lung inflammation. Our previous study showed that c-Src-dependent IκB kinase (IKK)/IκBα/nuclear factor (NF)-κB and mitogen-activated protein kinase kinase kinase 1 (MEKK1)/extracellular signal-regulated kinase (ERK)/ribosomal S6 protein kinase (RSK)-dependent CAAT/enhancer-binding protein ß (C/EBPß) activation are involved in thrombin-induced IL-8/CXCL8 expression in human lung epithelial cells. In this study, we aimed to investigate the roles of p300 and C/EBPß-reliant IKKß expression in thrombin-induced IL-8/CXCL8 expression. Thrombin-induced increases in IL-8/CXCL8-luciferase activity and IL-8/CXCL8 release were inhibited by p300 small interfering (siRNA). Thrombin-caused histone H3 acetylation was attenuated by p300 siRNA. Stimulation of cells with thrombin for 12h resulted in increases in IKKß expression and phosphorylation in human lung epithelial cells. However, thrombin did not affect p65 expression. Moreover, 12h of thrombin stimulation produced increases in IKKß expression and phosphorylation, and IκBα phosphorylation, which were inhibited by C/EBPß siRNA. Finally, treatment of cells with thrombin caused increases in p300 and C/EBPß complex formation, p65 and C/EBPß complex formation, and recruitment of p300, p65, and C/EBPß to the IL-8/CXCL8 promoter. These results imply that p300-dependent histone H3 acetylation and C/EBPß-regulated IKKß expression contribute to thrombin-induced IL-8/CXCL8 expression in human lung epithelial cells. Results of this study will help clarify C/EBPß signaling pathways involved in thrombin-induced IL-8/CXCL8 expression in human lung epithelial cells.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/imunologia , Proteína p300 Associada a E1A/imunologia , Quinase I-kappa B/genética , Inflamação/imunologia , Interleucina-8/genética , Mucosa Respiratória/imunologia , Trombina/imunologia , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Pulmão/citologia , Pulmão/imunologia , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo
10.
J Immunol ; 194(11): 5397-406, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25911750

RESUMO

Host defense peptides have recently gained much interest as novel anti-infectives owing to their ability to kill bacteria and simultaneously modulate host cell responses. The cationic host defense peptide GKY25 (GKYGFYTHVFRLKKWIQKVIDQFGE), derived from the C terminus of human thrombin, inhibits proinflammatory responses in vitro and in vivo, but the mode of action is unclear. In this study, we show that GKY25, apart from binding bacterial LPS, also interacts directly with monocytes and macrophages in vitro, ex vivo, and in vivo. Moreover, GKY25 inhibits TLR4- and TLR2-induced NF-κB activation in response to several microbe-derived agonists. Furthermore, GKY25 reduces LPS-induced phosphorylation of MAPKs p38α and JNK1/2/3. FACS and electron microscopy analyses showed that GKY25 interferes with TLR4/myeloid differentiation protein-2 dimerization. The results demonstrate a previously undisclosed activity of the host defense peptide GKY25, based on combined LPS and cell interactions leading to inhibition of TLR4 dimerization and subsequent reduction of NF-κB activity and proinflammatory cytokine production in monocytes and macrophages.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Lipopolissacarídeos/imunologia , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Trombina/farmacologia , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/imunologia , Linhagem Celular , Citocinas/biossíntese , Ativação Enzimática/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Antígeno 96 de Linfócito/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Monócitos/imunologia , NF-kappa B/metabolismo , Fragmentos de Peptídeos/imunologia , Fosforilação/efeitos dos fármacos , Multimerização Proteica , Transdução de Sinais/imunologia , Trombina/imunologia , Trombina/metabolismo , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia
11.
Rheumatol Int ; 37(4): 579-584, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27807637

RESUMO

A hallmark feature of antiphospholipid syndrome (APS) is the presence of a wide spectrum of antiphospholipid antibodies. In this study, we evaluated the clinical relevance of antibodies to prothrombin (PT) (aPT) and thrombin (aThr) in Chinese patients with APS. A total of 229 subjects were tested, including 86 patients with APS [35 patients with primary APS (PAPS), 51 patients with APS associated with other diseases (APSAOD)], 104 patients with non-APS diseases (disease controls), and 39 healthy controls. Serum IgG/IgM/IgA aPT and aThr were determined by ELISA. The levels of both IgG/IgM/IgA aPT and IgG/IgM/IgA aThr were significantly increased in patients with PAPS and APSAOD compared with patients with non-APS thrombosis and non-APS PRM, and HC. Both IgG aPT and IgG aThr exhibited promising diagnostic potentials for APS with sensitivities and specificities of 16.3 and 95.8% (IgG aPT), and 19.8 and 99.3% (IgG aThr), respectively. Importantly, both IgG aPT (OR 4.06; 95% CI 1.49-11.05) and IgG aThr (OR 4.49; 95% CI 1.62-12.45) were significantly correlated with arterial, but not venous, thrombotic events. Our findings highlighted that IgG aPT and IgG aThr could serve as promising biomarkers to identify patients at risk of arterial thrombosis in China.


Assuntos
Síndrome Antifosfolipídica/diagnóstico , Autoanticorpos/sangue , Protrombina/imunologia , Trombina/imunologia , Trombose/diagnóstico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Síndrome Antifosfolipídica/complicações , Síndrome Antifosfolipídica/imunologia , Biomarcadores/sangue , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Trombose/complicações , Trombose/imunologia , Adulto Jovem
13.
Infect Immun ; 83(9): 3515-25, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26099589

RESUMO

Platelets are rapidly responsive sentinel cells that patrol the bloodstream and contribute to the host response to infection. Platelets have been reported to form heterotypic aggregates with leukocytes and may modulate their function. Here, we have investigated platelet-neutrophil complex formation and neutrophil function in response to distinct agonists. The endogenous platelet activator thrombin gave rise to platelet-dependent neutrophil activation, resulting in enhanced phagocytosis and bacterial killing. Streptococcus pyogenes is an important causative agent of severe infectious disease, which can manifest as sepsis and septic shock. M1 protein from S. pyogenes also mediated platelet-neutrophil complex formation; however, these neutrophils were dysfunctional and exhibited diminished chemotactic ability and bacterial killing. This reveals an important agonist-dependent neutrophil dysfunction during platelet-neutrophil complex formation and highlights the role of platelets during the immune response to streptococcal infection.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Transporte/imunologia , Ativação de Neutrófilo/imunologia , Ativação Plaquetária/imunologia , Infecções Estreptocócicas/imunologia , Adulto , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Plaquetas/imunologia , Proteínas de Transporte/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Masculino , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Neutrófilos/imunologia , Fagocitose , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/imunologia , Streptococcus pyogenes/metabolismo , Trombina/imunologia
14.
J Virol ; 88(23): 13759-68, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25231318

RESUMO

UNLABELLED: Dengue virus (DENV) is the most common cause of viral hemorrhagic fever, and it may lead to life-threating dengue hemorrhagic fever and shock syndrome (DHF/DSS). Because most cases of DHF/DSS occur in patients with secondary DENV infection, anti-DENV antibodies are generally considered to play a role in the pathogenesis of DHF/DSS. Previously, we have found that antithrombin antibodies (ATAs) with both antithrombotic and profibrinolytic activities are present in the sera of dengue patients. However, the mechanism by which these autoantibodies are induced is unclear. In this study, we demonstrated that antibodies induced by DENV immunization in mice and rabbits could bind to DENV antigens as well as to human thrombin and plasminogen (Plg). The binding of anti-DENV antibodies to thrombin and Plg was inhibited by preadsorption with DENV nonstructural protein 1. In addition, affinity-purified ATAs from DENV-immunized rabbit sera could inhibit thrombin activity and enhance Plg activation both in vitro and in vivo. Taken together, our results suggest that molecular mimicry between DENV and coagulation factors can induce the production of autoantibodies with biological effects similar to those of ATAs found in dengue patients. These coagulation-factor cross-reactive anti-DENV antibodies can interfere with the balance of coagulation and fibrinolysis, which may lead to the tendency of DHF/DSS patients to bleed. IMPORTANCE: Dengue virus (DENV) infection is the most common mosquito-borne viral disease in tropical and subtropical areas. Over 50 million DENV infection cases develop each year, and more than 2.5 billion people are at risk of dengue-induced hemorrhagic fever and shock syndrome. Currently, there is no vaccine or drug treatment for DENV. In the present study, we demonstrated that DENV immunization could induce thrombin and plasminogen (Plg) cross-reactive antibodies, which were able to inhibit thrombin activity and enhance Plg activation. These results suggest that molecular mimicry between DENV antigens, thrombin, and Plg may elicit antibodies that disturb hemostasis. The selection of appropriate candidate antigens for use in DENV vaccines should prevent these potentially dangerous autoimmune responses.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Dengue/imunologia , Mimetismo Molecular , Plasminogênio/imunologia , Plasminogênio/metabolismo , Trombina/antagonistas & inibidores , Trombina/imunologia , Animais , Anticorpos Antivirais/metabolismo , Antígenos Virais/imunologia , Autoanticorpos/sangue , Autoanticorpos/metabolismo , Reações Cruzadas , Camundongos Endogâmicos BALB C , Ligação Proteica , Coelhos
15.
Blood ; 121(14): 2785-95, 2013 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-23349389

RESUMO

Approximately 30% of patients with severe hemophilia A develop inhibitory anti-factor VIII (fVIII) antibodies (Abs). We characterized 29 anti-human A2 monoclonal Abs (mAbs) produced in a murine hemophilia A model. A basis set of nonoverlapping mAbs was defined by competition enzyme-linked immunosorbent assay, producing 5 major groups. The overlapping epitopes covered nearly the entire A2 surface when mapped by homolog-scanning mutagenesis. Most group A mAbs recognized a previously described epitope bounded by Arg484-Ile508 in the N-terminal A2 subdomain, resulting in binding to activated fVIII and noncompetitive inhibition of the intrinsic fXase complex. Group B and C mAbs displayed little or no inhibitory activity. Group D and E mAbs recognized epitopes in the C-terminal A2 subdomain. A subset of group D mAbs inhibited the activation of fVIII by interfering with thrombin-catalyzed cleavage at Arg372 at the A1-A2 domain junction. Other group D mAbs displayed indeterminate or no inhibitory activity despite inhibiting cleavage at Arg740 at the A2-B domain junction. Group E mAbs inhibited fVIII light-chain cleavage at Arg1689. Inhibition of cleavages at Arg372 and Arg1689 represent novel mechanisms of inhibitor function and, along with the extensive epitope spectrum identified in this study, reveal hitherto unrecognized complexity in the immune response to fVIII.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos/imunologia , Mapeamento de Epitopos , Fator VIII/imunologia , Hemofilia A/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Reações Cruzadas/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Fator VIII/química , Fator VIII/genética , Hemofilia A/tratamento farmacológico , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Suínos , Trombina/imunologia , Trombina/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 34(2): 313-20, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24311376

RESUMO

OBJECTIVE: Transfusion of aged blood has been associated with increased morbidity and mortality in critically ill patients. During storage, erythrocytes release increasing numbers of microvesicles (red blood cell-derived microvesicles [RBC-MV]). We hypothesized that RBC-MV mediate some of the deleterious effects of aged blood transfusions. APPROACH AND RESULTS: We established a murine transfusion model using RBC-MV purified from aged mouse erythrocytes. Injection of RBC-MV into healthy mice had no effect. However, they aggravated pulmonary leukocyte sequestration and peripheral blood leukopenia induced by lipopolysaccharides. Lipopolysaccharide-induced proinflammatory cytokines were significantly increased in plasma after RBC-MV injection. These effects were not seen in C5aR-deficient mice. In vitro, RBC-MV bound C3 fragments after incubation with plasma but failed to bind immunoglobulins, C1q, or mannose-binding lectin. Preventing thrombin generation inhibited complement activation in vitro and in vivo and reversed the proinflammatory effects of RBC-MV in lipopolysaccharide-primed mice. Finally, the RBC-MV-induced phenotype was recapitulated using phosphatidylserine-expressing liposomes, suggesting that surface expression of phosphatidylserine by RBC-MV was mechanistically involved. CONCLUSIONS: These results point toward a thrombin-dependent mechanism of complement activation by RBC-MV independent of the classical, lectin, or alternative pathway. Besides identifying RBC-MV as potential mediators of transfusion-related morbidity, our findings may be relevant for other inflammatory disorders involving intravascular microvesicle release, for example, sickle cell disease or thrombotic microangiopathy.


Assuntos
Micropartículas Derivadas de Células/imunologia , Ativação do Complemento , Transfusão de Eritrócitos/efeitos adversos , Eritrócitos/imunologia , Inflamação/imunologia , Trombina/imunologia , Animais , Micropartículas Derivadas de Células/metabolismo , Complemento C3/imunologia , Complemento C3/metabolismo , Eritrócitos/metabolismo , Genótipo , Inflamação/sangue , Inflamação/induzido quimicamente , Leucopenia/sangue , Leucopenia/imunologia , Lipopolissacarídeos , Lipossomos , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Infiltração de Neutrófilos , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Fenótipo , Fosfatidilserinas/imunologia , Fosfatidilserinas/metabolismo , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/genética , Trombina/metabolismo , Fatores de Tempo
17.
J Biol Chem ; 288(30): 21569-80, 2013 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-23754290

RESUMO

The staphylococcal superantigen-like protein (SSL) family is composed of 14 exoproteins sharing structural similarity with superantigens but no superantigenic activity. Target proteins of four SSLs have been identified to be involved in host immune responses. However, the counterparts of other SSLs have been functionally uncharacterized. In this study, we have identified porcine plasma prothrombin as SSL10-binding protein by affinity purification using SSL10-conjugated Sepharose. The resin recovered the prodomain of prothrombin (fragment 1 + 2) as well as factor Xa in pull-down analysis. The equilibrium dissociation constant between SSL10 and prothrombin was 1.36 × 10(-7) M in surface plasmon resonance analysis. On the other hand, the resin failed to recover γ-carboxyglutamic acid (Gla) domain-less coagulation factors and prothrombin from warfarin-treated mice, suggesting that the Gla domain of the coagulation factors is essential for the interaction. SSL10 prolonged plasma clotting induced by the addition of Ca(2+) and factor Xa. SSL10 did not affect the protease activity of thrombin but inhibited the generation of thrombin activity in recalcified plasma. S. aureus produces coagulase that non-enzymatically activates prothrombin. SSL10 attenuated clotting induced by coagulase, but the inhibitory effect was weaker than that on physiological clotting, and SSL10 did not inhibit protease activity of staphylothrombin, the complex of prothrombin with coagulase. These results indicate that SSL10 inhibits blood coagulation by interfering with activation of coagulation cascade via binding to the Gla domain of coagulation factor but not by directly inhibiting thrombin activity. This is the first finding that the bacterial protein inhibits blood coagulation via targeting the Gla domain of coagulation factors.


Assuntos
Ácido 1-Carboxiglutâmico/imunologia , Proteínas de Bactérias/imunologia , Fator Xa/imunologia , Protrombina/imunologia , Staphylococcus aureus/imunologia , Superantígenos/imunologia , Ácido 1-Carboxiglutâmico/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/metabolismo , Sítios de Ligação/imunologia , Ligação Competitiva/imunologia , Coagulação Sanguínea/imunologia , Cálcio/imunologia , Cálcio/metabolismo , Coagulase/imunologia , Coagulase/metabolismo , Eletroforese em Gel de Poliacrilamida , Fator Xa/metabolismo , Humanos , Soros Imunes/imunologia , Soros Imunes/metabolismo , Camundongos , Dados de Sequência Molecular , Ligação Proteica/imunologia , Protrombina/metabolismo , Staphylococcus aureus/metabolismo , Superantígenos/metabolismo , Ressonância de Plasmônio de Superfície , Suínos , Trombina/imunologia , Trombina/metabolismo
18.
Bioconjug Chem ; 25(8): 1421-7, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25010569

RESUMO

To increase the affinities of antibodies or aptamers for their targets, we designed antibody-aptamer pincers (AAPs) or heterodimers for thrombin or human epidermal growth factor 2 (HER2) as a model system. For this purpose, we first conjugated a 15-mer or 29-mer anti-thrombin aptamer, which are well-known to bind to thrombin in two specific epitopes, with an anti-thrombin antibody to enable each binding part of the AAP to simultaneously recognize a different part of the thrombin molecule. The AAP comprising a 15-mer aptamer and an anti-thrombin antibody has an apparent dissociation constant (Kd(app)) value of 567 pM, and this value is approximately 1/100 of that of the antibody alone or 1/35 of that of the aptamer monomer alone. The AAP comprising a 29-mer aptamer and an anti-thrombin antibody has a much lower Kd(app) value than that of 15-mer aptamer-conjugated antibody. Furthermore, this concept of the AAP system was employed to HER2-targeted drug delivery system (DDS) based on both antibody and drug-loaded aptamer. Anti-HER2 aptamer was conjugated with anti-HER2 antibody and loaded with doxorubicin, and the resulting AAP-HER2-Dox was found to have approximately 3- and 6-fold higher cytotoxicity than drug alone and antibody alone, respectively. Therefore, this novel AAP system constructed by conjugation of the antibody with the aptamer can effectively improve the affinities of the resulting AAPs for their target molecules and the drug-loaded AAP system can possibly serve as a platform for targeted DDS against many malignancies.


Assuntos
Anticorpos/metabolismo , Aptâmeros de Nucleotídeos/metabolismo , Anticorpos/química , Anticorpos/imunologia , Aptâmeros de Nucleotídeos/química , Sequência de Bases , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/farmacologia , Humanos , Ligantes , Multimerização Proteica , Estrutura Quaternária de Proteína , Receptor ErbB-2/metabolismo , Trombina/imunologia , Trombina/metabolismo
19.
Blood ; 120(8): 1717-25, 2012 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-22802338

RESUMO

The coagulation and complement pathways simultaneously promote homeostasis in response to injury but cause tissue damage when unregulated. Mechanisms by which they cooperate are poorly understood. To delineate their interactions, we studied the effects of thrombin and C5 convertase on C5 in purified and plasma-based systems, measuring release of the anaphylatoxin C5a, and generation of C5b, the initial component of the lytic membrane attack complex. Thrombin cleaved C5 poorly at R751, yielding minimal C5a and C5b. However, thrombin efficiently cleaved C5 at a newly identified, highly conserved R947 site, generating previously undescribed intermediates C5(T) and C5b(T). Tissue factor-induced clotting of plasma led to proteolysis of C5 at a thrombin-sensitive site corresponding to R947 and not R751. Combined treatment of C5 with thrombin and C5 convertase yielded C5a and C5b(T), the latter forming a C5b(T)-9 membrane attack complex with significantly more lytic activity than with C5b-9. Our findings provide a new paradigm for complement activation, in which thrombin and C5 convertase are invariant partners, enhancing the terminal pathway via the generation of newly uncovered C5 intermediates. Delineating the molecular links between coagulation and complement will provide new therapeutic targets for diseases associated with excess fibrin deposition and complement activation.


Assuntos
Ativação do Complemento , Complemento C5/imunologia , Trombina/imunologia , Animais , Coagulação Sanguínea , Galinhas , Convertases de Complemento C3-C5/metabolismo , Complemento C5/metabolismo , Eritrócitos/citologia , Eritrócitos/imunologia , Hemólise , Humanos , Proteólise , Transdução de Sinais , Trombina/metabolismo , Tromboplastina/metabolismo
20.
Vox Sang ; 107(1): 97-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24517203

RESUMO

Factor V (FV) inhibitor arises rarely after using fresh frozen plasma (FFP) to treat inherited FV deficiency and is often a real therapeutic challenge. Here, we report a patient with a severe FV deficiency who developed such an inhibitor and was then treated with recombinant activated FVII (rFVIIa) and platelet concentrates (PC). Monitoring was assessed by thrombin generation assay (TGA). PC were more effective than rFVIIa in treating bleeding, but there was no correlation between the TGA results and clinical efficacy.


Assuntos
Deficiência do Fator V/complicações , Fator VIIa/farmacologia , Fator V/antagonistas & inibidores , Hemorragia/tratamento farmacológico , Deficiência do Fator V/genética , Hemoglobinas/metabolismo , Hemorragia/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto/genética , Plasma , Proteínas Recombinantes/farmacologia , Trombina/imunologia , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA