Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Int J Mol Sci ; 24(13)2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-37446134

RESUMO

Anti-ß2-glycoprotein I/HLA-DR (anti-ß2GPI/HLA-DR) antibody has been reported to be associated with antiphospholipid syndrome and recurrent pregnancy loss (RPL). We conducted a prospective multicenter cross-sectional study aimed at evaluating whether the anti-ß2GPI/HLA-DR antibody is associated with adverse obstetric outcomes and RPL. From 2019 to 2021, serum anti-ß2GPI/HLA-DR antibody levels (normal, <73.3 U) were measured in 462 women with RPL, 124 with fetal growth restriction (FGR), 138 with hypertensive disorders of pregnancy (HDP), 71 with preterm delivery before 34 gestational weeks (preterm delivery (PD) ≤ 34 GWs), and 488 control women who experienced normal delivery, by flow cytometry analysis. The adjusted odds ratios (aORs) of anti-ß2GPI/HLA-DR antibody positivity for adverse obstetric outcomes and RPL were evaluated on the basis of comparisons between the control and each patient group, using multivariable logistic regression analysis. The following were the positivity rates for the anti-ß2GPI/HLA-DR antibody in the patient and control groups: RPL, 16.9%; FGR, 15.3%; HDP, 17.4%; PD ≤ 34 GWs, 11.3%; and the control, 5.5%. It was demonstrated that anti-ß2GPI/HLA-DR antibody positivity was a significant risk factor for RPL (aOR, 3.3 [95% confidence interval {CI} 1.9-5.6], p < 0.001), FGR (2.7 [1.3-5.3], p < 0.01), and HDP (2.7 [1.4-5.3], p < 0.01) although not for PD ≤ 34 GWs. For the first time, our study demonstrated that the anti-ß2GPI/HLA-DR antibody is involved in the pathophysiology underlying FGR and HDP, as well as RPL.


Assuntos
Síndrome Antifosfolipídica , Pré-Eclâmpsia , Nascimento Prematuro , Gravidez , Recém-Nascido , Humanos , Feminino , Estudos Transversais , Estudos Prospectivos , Antígenos HLA-DR , Autoanticorpos , beta 2-Glicoproteína I
2.
Pharmacol Res ; 164: 105372, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33316382

RESUMO

Cardiovascular disease continues to be the most common cause of death worldwide. The global burden is so high that numerous organizations are providing counseling recommendations and annual revisions of current pharmacological and non-pharmacological treatments as well as risk prediction for disease prevention and further progression. Although primary preventive interventions targeting risk factors such as obesity, hypertension, smoking, and sedentarism have led to a global decline in hospitalization rates, the aging population has overwhelmed these efforts on a global scale. This review focuses on peptidic vaccines, with the known and not well-known autoantigens in atheroma formation or acquired cardiac diseases, as novel potential immunotherapy approaches to counteract harmful heart disease continuance. We summarize how cancer immunomodulatory strategies started novel approaches to modulate the innate and adaptive immune responses, and how they can be targeted for therapeutic purposes in the cardiovascular system. Brief descriptions focused on the processes that start as either immunologic or non-immunologic, and the ultimate loss of cardiac muscle cell contractility as the outcome, are discussed. We conclude debating how novel strategies with nanoparticles and nanovaccines open a promising therapeutic option to reduce or prevent cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/terapia , Imunoterapia , Vacinas de Subunidades Antigênicas/uso terapêutico , Animais , Autoantígenos/imunologia , Doenças Cardiovasculares/imunologia , Endotélio Vascular , Humanos , Placa Aterosclerótica/prevenção & controle , Sistema Renina-Angiotensina
3.
J Thromb Thrombolysis ; 52(4): 1133-1136, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33914240

RESUMO

Although a few antiphospholipid syndrome (APS) occurs with acquired thrombotic thrombocytopenic purpura (TTP), the relationship between antiphospholipid antibodies (aPL) and anti-ADAMTS13 (anti-a disintegrin and metalloprotease with thrombospondin type 1 motif, member 13) antibody remains uncertain. We investigated the relationship between high-risk thrombotic aPL and anti-ADAMTS13 antibody. Two hundred and thirty-seven patients with positive lupus anticoagulant and/or anticardiolipin antibody were included. Anti-ß2GPI (anti-ß2-glycoprotein I), anti-ß2GPIdI (anti-ß2-glycoprotein I domain I), anti-PS/PT (anti-phosphatidylserine and prothrombin), ADAMTS13 activity, and anti-ADAMTS13 antibody were measured. Double positivity of anti-ß2GPI and anti-PS/PT increased thrombotic risk more than three-fold and showed increased positivity of anti-ADAMTS13 antibody in comparison with the double negative group. Double positivity of anti-ß2GPIdI and anti-PS/PT presented both effects even more. In the linear regression analysis, double positivity of anti-ß2GPI and anti-PS/PT independently affected the anti-ADAMTS13 antibody level (ß = 1.982, P = 0.042). Our results revealed that double positivity of anti-ß2GPI or anti-ß2GPIdI and anti-PS/PT increased not only thrombotic risk but also the positivity of anti-ADAMTS13 antibody, especially indicating anti-ß2GPIdI showed a higher synergistic effect with anti-PS/PT. We suggest a possible association of anti-ADAMTS13 antibody with a high thrombotic risk of APS. Double positivity of anti-ß2GPI (anti-ß2-glycoprotein I) and anti-PS/PT (anti-phosphatidylserine and prothrombin) antibodies enhanced not only thrombotic risk but also positivity of anti-ADAMTS13 (anti-a disintegrin and metalloprotease with thrombospondin type 1 motif, member 13) antibody. Furthermore, double positivity of anti-ß2GPIdI (anti-ß2-glycoprotein I domain I) combined with anti-PS/PT even more elevated both thrombosis and positivity of anti-ADAMTS13 antibody. Double positivity of ß2GPI and anti-PS/PT was found as an independently significant contributing factor to anti-ADAMTS13 antibody level. We suggest the association between anti-ADAMTS13 antibody and the pathophysiology of antiphospholipid syndrome, which should be further evaluated.


Assuntos
Síndrome Antifosfolipídica , Trombose , Anticorpos Antifosfolipídeos , Desintegrinas , Humanos , Fosfatidilserinas , Protrombina , Trombospondinas , beta 2-Glicoproteína I
4.
Molecules ; 26(9)2021 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-33923336

RESUMO

Antiphospholipid syndrome (APS) is an autoimmune disease characterized by the production of ß2-glycoprotein I (ß2GPI)-dependent autoantibodies, with vascular thrombosis or obstetrical complications. Around 20% of APS patients are refractory to current treatments. Crassolide, a cembranoid diterpene extracted from soft corals, is a potential therapeutic candidate. Here, to examine the anti-inflammatory properties of crassolide, we first determined its effects on bone marrow-derived and splenic dendritic cells (DC). Specifically, we applied lipopolysaccharide (LPS) or ß2GPI stimulation and measured the expressions of CD80 and CD86, and secretions of cytokines. We also determined in the OT-II mice, if bone marrow-derived DC was able to stimulate antigen-specific T cells. Moreover, we examined the therapeutic potential of crassolide postimmunization in a murine model of APS that depended on active immunization with ß2GPI. The vascular manifestations were evaluated in terms of fluorescein-induced thrombi in mesenteric microvessels, whereas the obstetric manifestations were evaluated based on the proportion of fetal loss after pregnancy. We also measured blood titers of anti-ß2GPI antibody, splenic cell proliferative responses and cytokine secretions after ß2GPI stimulation ex vivo. Finally, we determined in these mice, hematological, hepatic and renal toxicities of crassolide. Crassolide after LPS stimulation suppressed DC maturation and secretion of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-12 and IL-23, and downstream T cell activation. Crassolide could partially ameliorate both the vascular and obstetric manifestations of APS in BALB/c mice. Both blood titers of anti-ß2GPI antibody and splenic cell proliferation after ß2GPI stimulation were reduced. Splenic Th1 and Th17 responses were also lowered after ß2GPI stimulation. Finally, within therapeutic doses of crassolide, we found no evidence of its toxicity. In conclusion, we showed the ability of crassolide to suppress DC and downstream T cell responses. Crassolide is therefore a potential candidate for adjunctive therapy in APS.


Assuntos
Síndrome Antifosfolipídica/tratamento farmacológico , Células Dendríticas/efeitos dos fármacos , Diterpenos/farmacologia , Inflamação/tratamento farmacológico , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Síndrome Antifosfolipídica/genética , Síndrome Antifosfolipídica/imunologia , Síndrome Antifosfolipídica/patologia , Antígeno B7-1/genética , Antígeno B7-2/genética , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos/toxicidade , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Gravidez , beta 2-Glicoproteína I/toxicidade
5.
J Thromb Thrombolysis ; 49(2): 228-234, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31612355

RESUMO

Antiphospholipid antibodies (aPL) are heterogeneous and there is evidence that binding specificity determines which cellular effects they can trigger. We have therefore hypothesised that the induction of tissue factor (TF) in monocytes and endothelial cells by aPL depends on their binding specificity. To further investigate this, we have analyzed the ability of three human monoclonal aPL with distinctly different binding specificities to induce transcription and cell surface expression of TF in monocytes and endothelial cells. Results with human monoclonal aPL were validated with IgG-fractions obtained from patients with APS. We confirmed previous results that a lipid reactive human monoclonal aPL rapidly induced TF transcription and cell surface expression in monocytes and endothelial cells. A monoclonal aPL reactive against ß2 glycoprotein I (ß2GPI) induced TF with a delayed time course. This was fully dependent on the induction of tumor necrosis factor alpha (TNFα) secretion as capture of TNFα by adalimumab prevented TF induction. This pattern was confirmed with patient IgG fractions. Both lipid reactive and anti-ß2GPI induced TF transcription. Unexpectedly, this activity of anti-ß2GPI was mediated fully by TNFα which was secreted in response to incubation with anti-ß2GPI. The role of TNFα in mediating TF induction by anti-ß2GPI may have wider implications for APS pathogenesis.


Assuntos
Anticorpos Monoclonais/farmacologia , Tromboplastina/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , beta 2-Glicoproteína I/farmacologia , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Tromboplastina/genética , Fator de Necrose Tumoral alfa/genética
6.
Crit Rev Clin Lab Sci ; 56(8): 511-525, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31448653

RESUMO

Antiphospholipid antibody syndrome (APS) is a systemic autoimmune disease characterized by arterial and venous thrombosis and/or pregnancy morbidity. The incidence is around five new cases/100,000 persons per year and the prevalence is around 40-50 cases/100,000. The prevalence is higher (about 30%) among patients with systemic lupus erythematosus. APS is associated with circulating antiphospholipid antibodies (aPLs), a heterogeneous group of autoantibodies directed against negatively charged molecules and a combination of protein-complexed phospholipids. The predominant protein antigen in this disorder is beta 2-glycoprotein I (ß2GPI). Despite the discovery of "new" antigenic targets and development of "new" methodological approaches, the laboratory diagnosis of APS is still an evolving field and studies to identify further antigenic target(s) as potential diagnostic markers and risk predictors are in progress. In particular, recent studies were aimed at analyzing the pathogenic role of post-translational modifications (PTMs) of proteins induced by inflammation and/or oxidative stress as modulators of protein structure and function and possibly as a source of antigenic epitopes. The present review is focused on PTMs of self-proteins responsible for autoimmune reactions in patients with APS. At present, the known PTMs in APS involve ß2GPI. In particular, the PTM of ß2GPI via thiol-exchange reactions is a highly specific phenomenon that makes the protein more antigenic. Other PTMs, including sialylation and acetylation, may affect ß2GPI antigenicity. Moreover, the addition or loss of carbohydrate chains affects ß2GPI immunoreactivity since carbohydrates are determining factors for ß2GPI conformation. In addition to ß2GPI, PTMs of other self proteins such as vimentin and annexins may play a role in the immune response during APS. The study of PTMs is useful to clarify the role of modified proteins in the pathogenesis of APS as well as to design more efficient diagnostic/prognostic tools and more targeted therapeutic approaches.


Assuntos
Síndrome Antifosfolipídica/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas/metabolismo , Autoantígenos/imunologia , Autoimunidade , Humanos , Estresse Oxidativo
7.
Cancer Sci ; 110(6): 1974-1986, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31012976

RESUMO

We previously found that circulating ß2 -glycoprotein I inhibits human endothelial cell migration, proliferation, and angiogenesis by diverse mechanisms. In the present study, we investigated the antitumor activities of ß2 -glycoprotein I using structure-function analysis and mapped the critical region within the ß2 -glycoprotein I peptide sequence that mediates anticancer effects. We constructed recombinant cDNA and purified different ß2 -glycoprotein I polypeptide domains using a baculovirus expression system. We found that purified ß2 -glycoprotein I, as well as recombinant ß2 -glycoprotein I full-length (D12345), polypeptide domains I-IV (D1234), and polypeptide domain I (D1) significantly inhibited melanoma cell migration, proliferation and invasion. Western blot analyses were used to determine the dysregulated expression of proteins essential for intracellular signaling pathways in B16-F10 treated with ß2 -glycoprotein I and variant recombinant polypeptides. Using a melanoma mouse model, we found that D1 polypeptide showed stronger potency in suppressing tumor growth. Structural analysis showed that fragments A and B within domain I would be the critical regions responsible for antitumor activity. Annexin A2 was identified as the counterpart molecule for ß2 -glycoprotein I by immunofluorescence and coimmunoprecipitation assays. Interaction between specific amino acids of ß2 -glycoprotein I D1 and annexin A2 was later evaluated by the molecular docking approach. Moreover, five amino acid residues were selected from fragments A and B for functional evaluation using site-directed mutagenesis, and P11A, M42A, and I55P mutations were shown to disrupt the anti-melanoma cell migration ability of ß2 -glycoprotein I. This is the first study to show the therapeutic potential of ß2 -glycoprotein I D1 in the treatment of melanoma progression.


Assuntos
Movimento Celular/efeitos dos fármacos , Melanoma Experimental/tratamento farmacológico , Peptídeos/farmacologia , beta 2-Glicoproteína I/química , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Linhagem Celular Tumoral , Masculino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Mutagênese Sítio-Dirigida , Peptídeos/química , Peptídeos/metabolismo , Domínios Proteicos , Homologia de Sequência de Aminoácidos , beta 2-Glicoproteína I/genética , beta 2-Glicoproteína I/metabolismo
8.
Immunol Cell Biol ; 97(9): 799-814, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31187539

RESUMO

Systemic lupus erythematosus (SLE) is characterized by the development of autoantibodies against diverse self-antigens with damage to multiple organs. Immunization with the SLE autoantigen ß2 -glycoprotein I (ß2 GPI) and lipopolysaccharide (LPS), a known trigger of necroptosis, induces a murine model of SLE. We hypothesized that necroptotic cells, like apoptotic cells, provide a "scaffold" of cellular self-antigens, but, unlike apoptotic cells, necroptotic cells do so in a proinflammatory and immunogenic context. We demonstrate that ß2 GPI indeed binds to necroptotic cells and serves as a target for anti-ß2 GPI autoantibodies. We further demonstrate that necroptotic, but not apoptotic, cells promote antigenic presentation of ß2 GPI to CD4 T cells by dendritic cells. Finally, we show that ß2 GPI/LPS-immunized mice deficient in RIPK3 (receptor-interacting serine/threonine-protein kinase 3) or MLKL (mixed lineage kinase domain like), and consequently unable to undergo necroptosis, have reduced SLE autoantibody production and pathology. RIPK3-/- mice had low levels of SLE autoantibodies and no renal pathology, while MLKL-/- mice produced low levels of SLE autoantibodies initially, but later developed levels comparable with wild type (WT) mice and pathology intermediate to that of WT and RIPK3-/- mice. Serum cytokine levels induced by LPS tended to be lower in RIPK3-/- and MLKL-/- mice than in WT mice, suggesting that impaired proinflammatory cytokine production may impact the initiation of autoantibody production in both strains. Our data suggest that self-antigen (i.e. ß2 GPI) presented in the context of necroptosis and proinflammatory signals may be sufficient to overcome immune tolerance and induce SLE.


Assuntos
Autoantígenos/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Necroptose/imunologia , beta 2-Glicoproteína I/metabolismo , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/metabolismo , Apoptose , Autoanticorpos/imunologia , Linhagem Celular , Citocinas/metabolismo , Células Dendríticas/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Ligação Proteica , Proteínas Quinases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
9.
Clin Chem Lab Med ; 55(6): 882-889, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28002027

RESUMO

BACKGROUND: Highly specific assays for measuring antiphospholipid antibodies (aPLs) are required for accurate assessment of thrombotic risk. aPLs against ß2-glycoprotein I domain I (anti-ß2GPIdI) and against prothrombin complexed with phosphatidylserine (anti-PS/PT) have been recently identified as being associated with a hypercoagulable state. This study evaluated the synergism between anti-ß2GPIdI and anti-PS/PT for predicting thrombotic events. METHODS: A total of 180 patients with clinical suspicion of hypercoagulability were evaluated. The plasma levels of lupus anticoagulant (LA) and antibodies against anticardiolipin (anti-CL) (IgG and IgM), ß2GPI (IgG and IgM), PS/PT (IgG and IgM), and ß2GPI dI (IgG) were measured. RESULTS: IgG anti-ß2GPIdI and LA were highly associated with thrombosis. Mean values and positivity rates of IgG anti-ß2GPI dI and IgG anti-PS/PT were significantly higher in the triple-positive group (LA+, IgG anti-CL+, IgG anti-ß2GPI+) than in the other groups. Interestingly, the thrombotic risk [odds ratio (OR) 24.400, 95% confidence interval (CI) 1.976-63.273, p<0.001] of the newly defined triple positive group (LA+, IgG anti-CL+, IgG anti-ß2GPIdI+; OR 11.182, 95% CI 1.976-63.273, p=0.006) was more than twice that of the triple-positive group (LA+, IgG anti-CL+, IgG anti-ß2GPI+). Double positivity for IgG anti-PS/PT and IgG anti-ß2GPI also indicated significant thrombotic risk (OR 7.467, 95% CI 2.350-23.729, p=0.001). Furthermore, the thrombotic risk associated with double positivity for IgG anti-PS/PT and IgG anti-ß2GPIdI was markedly elevated (OR 33.654, 95% CI 6.322-179.141, p<0.001). CONCLUSIONS: Our data suggest that simultaneous measurement of IgG anti-ß2GPIdI and IgG anti-PS/PT may improve clinical decision-making for aPL-positive patients.


Assuntos
Anticorpos Antifosfolipídeos/sangue , Anticorpos Antifosfolipídeos/imunologia , Fosfatidilserinas/imunologia , Protrombina/imunologia , Trombose/sangue , beta 2-Glicoproteína I/imunologia , Adulto , Feminino , Humanos , Masculino , Prognóstico , Risco , Trombose/diagnóstico , Trombose/imunologia
10.
Lipids Health Dis ; 16(1): 173, 2017 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-28903783

RESUMO

BACKGROUND: New evidence has shown that reduced ß2-glycoprotein I (ß2GPI) has anti-oxidative stress and anti-inflammatory activity. However, the details are still poorly understood. This study aims to prepare stable reduced ß2GPI with its native bioactivity in vitro. METHODS: Human ß2GPI was purified from plasma first with perchloric acid precipitation and then purified with a series of chromatography methods including Sephadex G-25 desalting, SP HP, AF-heparin HC-650 M, and Sephacryl S-200. The purified human ß2GPI was reduced with thioredoxin-1 (TRX-1) activated by DL-dithiothreitol (DTT). Glutathione (GSH) was selected to block the free thiols in reduced ß2GPI. LC/MS was used to verify the location of free thiols. Western blot analysis was used to detect ß2GPI immunoreactivity. MTS and flow cytometry were conducted to investigate its biological effect on oxidative-stress-induced death of human umbilical vein endothelial cells (HUVECs). The levels of tumour necrosis factor-alpha (TNF-α),interleukin-6 (IL-6) interleukin-10 (IL-10),interleukin-12P70 (IL-12P70),interferon-gamma (IFN-γ) and monocyte chemoattractant protein -1(MCP-1) in mouse serum were quantified to assess its anti-inflammatory activity in lipopolysaccharide (LPS)-mediated systemic inflammation. RESULTS: We obtained approximately 10 mg ß2GPI (purity 98.7%) from 200 ml plasma. The protein yield was 0.05 mg/ml plasma. ß2GPI was then reduced by TRX-1/DTT in vitro; the free thiols were detected on Cys288 and Cys326 in domain V of ß2GPI. The GSH blockage stabilized the reduced ß2GPI in vitro. This reduced ß2GPI can be recognized by the anti-ß2GPI antibody, can significantly reduce the death of HUVECs after H2O2 treatment and can significantly decrease the levels of TNF-α, IL-6,IFN-γ and MCP-1 in mice upon LPS stimulation. CONCLUSION: Stable reduced ß2GPI can be obtained in vitro by TRX-1 deoxidation followed by the blockage of thiols with GSH. This reduced ß2GPI maintains the same immunological activity as oxidized ß2GPI and has the ability to counter the oxidative stress induced by H2O2 in HUVECs and inflammation in LPS-mediated inflammation in mice.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , beta 2-Glicoproteína I/isolamento & purificação , beta 2-Glicoproteína I/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Compostos de Sulfidrila/química , beta 2-Glicoproteína I/química , beta 2-Glicoproteína I/imunologia
11.
Biochem J ; 473(24): 4629-4650, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27760842

RESUMO

ß2-Glycoprotein I (ß2GpI) is the major autoantigen in the antiphospholipid syndrome, a thrombotic autoimmune disease. Nonetheless, the physiological role of ß2GpI is still unclear. In a recent work, we have shown that ß2GpI selectively inhibits the procoagulant functions of human α-thrombin (αT; i.e. prolongs fibrin clotting time, tc, and inhibits αT-induced platelet aggregation) without affecting the unique anticoagulant activity of the protease, i.e. the proteolytic generation of the anticoagulant protein C (PC) from the PC zymogen, which interacts with αT exclusively at the protease catalytic site. Here, we used several different biochemical/biophysical techniques and molecular probes for mapping the binding sites in the αT-ß2GpI complex. Our results indicate that αT exploits the highly electropositive exosite-II, which is also responsible for anchoring αT on the platelet GpIbα (platelet receptor glycoprotein Ibα) receptor, for binding to a continuous negative region on ß2GpI structure, spanning domain IV and (part of) domain V, whereas the protease active site and exosite-I (i.e. the fibrinogen-binding site) remain accessible for substrate/ligand binding. Furthermore, we provided evidence that the apparent increase in tc, previously observed with ß2GpI, is more likely caused by alteration in the ensuing fibrin structure rather than by the inhibition of fibrinogen hydrolysis. Finally, we produced a theoretical docking model of αT-ß2GpI interaction, which was in agreement with the experimental results. Altogether, these findings help to understand how ß2GpI affects αT interactions and suggest that ß2GpI may function as a scavenger of αT for binding to the GpIbα receptor, thus impairing platelet aggregation while enabling normal cleavage of fibrinogen and PC.


Assuntos
Trombina/química , Trombina/metabolismo , beta 2-Glicoproteína I/química , beta 2-Glicoproteína I/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Difusão Dinâmica da Luz , Fibrina/química , Fibrina/metabolismo , Humanos , Cinética , Concentração Osmolar , Agregação Plaquetária/efeitos dos fármacos , Espectrometria de Fluorescência , Ressonância de Plasmônio de Superfície , Trombina/farmacologia
12.
J Biol Chem ; 290(9): 5543-55, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25555913

RESUMO

Systemic lupus erythematosus (SLE) is a prototypic model for B cell epitope spread in autoimmunity. Autoantibodies to numerous and molecularly distinct self-antigens emerge in a sequential manner over several years, leading to disease manifestations. Among the earliest autoantibodies to appear are those targeting the apoptotic cell-binding protein ß2-glycoprotein I (ß2GPI). Notably, mice immunized with ß2GPI and LPS display a remarkably similar pattern of autoantibody emergence to that seen in human SLE. Here, we used this model to investigate whether epitope spread to SLE-related autoantibodies is associated with a unique or limited ß2GPI-specific T cell response. We ask whether MHC class II haplotype and its associated T cell epitope restriction impact epitope spread to SLE-related autoantibodies. We found that ß2GPI/LPS-immunized mice produced similar SLE-related autoantibody profiles regardless of their ß2GPI T cell epitope specificity or MHC class II haplotype. Although ß2GPI T cell epitope specificity was clearly determined by MHC class II haplotype, a number of different ß2GPI T cell epitopes were associated with epitope spread to SLE-related autoantibodies. Notably, one ß2GPI T cell epitope (peptide 23, NTGFYLNGADSAKCT) was also recognized by T cells from an HLA-DRB1*0403(+) autoimmune patient. These data suggest that the generation of a ß2GPI-reactive T cell response is associated with epitope spread to SLE-related autoantibodies, independent of epitope specificity or MHC class II restriction. On the basis of these findings, we propose that factors enabling a ß2GPI-reactive T cell response may predispose individuals to the development of SLE-related autoantibodies independent of their MHC class II haplotype.


Assuntos
Autoanticorpos/imunologia , Epitopos de Linfócito T/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Linfócitos T/imunologia , beta 2-Glicoproteína I/imunologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito T/metabolismo , Feminino , Cadeias HLA-DRB1/genética , Cadeias HLA-DRB1/imunologia , Haplótipos/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Hibridomas , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Linfócitos T/metabolismo , beta 2-Glicoproteína I/metabolismo
13.
Stroke ; 47(10): 2589-95, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27625376

RESUMO

BACKGROUND AND PURPOSE: The burden of stroke is high in sub-Saharan Africa, and improved knowledge of risk factors is needed. Antiphospholipid antibodies are a common acquired stroke risk factor in young individuals. Antiphospholipid antibodies may be induced by infectious diseases. Sub-Saharan Africa has a high infectious burden, and we analyzed the contribution of antiphospholipid antibodies to the risk of stroke in an incident population from rural and urban Tanzania. METHODS: Stroke cases and age- and sex-matched community-acquired controls from the rural Hai district and urban Dar-es-Salaam areas of Tanzania were recruited in a wider study of stroke incidence between June 2003 and June 2006. Lupus anticoagulant, anticardiolipin, anti-ß2-glycoprotein I, and antiphosphatidylserine/prothrombin antibodies were determined in stored plasma, as well as IgG antibodies against Treponema pallidum. RESULTS: Data from 158 stroke cases and 369 controls were analyzed. Thirty cases (19%) and 4 controls (1%) had a lupus anticoagulant (odds ratio, 20.8; 95% confidence interval, 7.2-60.5). Anticardiolipin IgG was the only other antiphospholipid antibody subtype associated with increased stroke risk (odds ratio, 2.1; 95% confidence interval, 1.0-4.3), but this association disappeared when corrected for IgG antibodies against Treponema pallidum results. The prevalence of anti-ß2-glycoprotein I IgG antibodies in the Tanzanian healthy population was high when Dutch cutoff values were applied (67%), whereas presence of anti-ß2-glycoprotein I IgM was associated with a reduced stroke risk (odds ratio 0.3; 95% confidence interval, 0.1-1.1). CONCLUSIONS: The presence of lupus anticoagulant is a strong, and to date unrecognized, risk factor for stroke in Tanzania, especially in young and middle-aged individuals.


Assuntos
Anticorpos Antifosfolipídeos/sangue , Acidente Vascular Cerebral/epidemiologia , Idoso , Idoso de 80 Anos ou mais , Anticorpos Anticardiolipina/sangue , Estudos de Casos e Controles , Feminino , Humanos , Incidência , Inibidor de Coagulação do Lúpus/sangue , Masculino , Pessoa de Meia-Idade , Prevalência , População Rural , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/imunologia , Tanzânia/epidemiologia , População Urbana
14.
Clin Immunol ; 160(2): 198-210, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26065621

RESUMO

Antiphospholipid (aPL)/anti-ß2-glycoprotein I (ß2GPI) antibodies are considered to play a pivotal pathogenic role in antiphospholipid syndrome (APS) by inducing an intracellular signaling and procoagulant/proinflammatory phenotype that leads to thrombosis. There is increasing evidence that Toll-like receptor 4 (TLR4) could serve as an important molecule for anti-ß2GPI recognition on target cells. However, few studies have focused on the effects of TLR4 in in vivo models. Here, we investigated the role of TLR4 in the pathogenic effects of aPL/anti-ß2GPI more precisely using TLR4-intact (C3H/HeN) and TLR4-defective (C3H/HeJ) mice. C3H/HeN and C3H/HeJ mice were injected with either IgG isolated from patient with APS (IgG-APS) or epitope-specific anti-ß2GPI purified from ß2GPI peptide-immunized rabbits. We found that, following anti-ß2GPI injections and vascular injury, thrombus formation in both the carotid artery and femoral vein was markedly reduced in C3H/HeJ mice when compared with C3H/HeN mice. IgG-APS or anti-ß2GPI-induced carotid artery and peritoneal macrophage tissue factor activity/expression was significantly lesser in C3H/HeJ than in C3H/HeN mice. Furthermore, the IgG-APS or anti-ß2GPI induced expression of VCAM-1, ICAM-1, and E-selectin in the aorta and of IL-1ß, IL-6, and TNF-α in peritoneal macrophages of C3H/HeJ mice was also significantly reduced compared to C3H/HeN mice. Together, these data suggest that TLR4 is involved in the pathogenic effects of aPL/anti-ß2GPI antibodies in vivo.


Assuntos
Anticorpos Antifosfolipídeos/imunologia , Síndrome Antifosfolipídica/imunologia , Trombose das Artérias Carótidas/imunologia , Receptor 4 Toll-Like/imunologia , Trombose Venosa/imunologia , Animais , Trombose das Artérias Carótidas/induzido quimicamente , Adesão Celular , Cloretos/toxicidade , Modelos Animais de Doenças , Selectina E/metabolismo , Veia Femoral , Compostos Férricos/toxicidade , Imunoglobulina G/imunologia , Mediadores da Inflamação/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Lipopolissacarídeos , Camundongos , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo , Trombose Venosa/induzido quimicamente , beta 2-Glicoproteína I/imunologia
15.
Rheumatology (Oxford) ; 54(4): 722-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25273993

RESUMO

OBJECTIVE: IgG aPL against domain I of ß2-glycoprotein I (ß2GPI) [anti-DI (aDI)] is associated with the pathogenesis of APS, an autoimmune disease defined by thrombosis and pregnancy morbidity. To date, however, no study has demonstrated direct pathogenicity of IgG aDI in vivo. In this proof-of-concept study, we designed a novel system to affinity purify polyclonal aDI aPL in order to assess its prothrombotic ability in a well-characterized mouse microcirculation model for APS. METHODS: Two polyclonal IgG fractions were isolated from serum of a patient with APS, both with high aPL activity but differing in aDI activity (aDI-rich and aDI-poor). These IgG fractions were tested for their pathogenic ability in an in vivo mouse model of thrombosis. Male CD1 mice were injected intraperitoneally with either aDI-rich or aDI-poor IgG; as a control, IgG isolated from healthy serum was used. A pinch injury was applied to the right femoral vein and thrombus dynamics and tissue factor activity in isolated tissue were evaluated. RESULTS: Both aDI-rich and aDI-poor IgG retained aCL and anti-ß2GPI activity, while only aDI-rich IgG displayed high aDI activity, as defined by our in-house cut-offs for positivity in each assay. aDI-rich IgG induced significantly larger thrombi in vivo compared with aDI-poor IgG (P < 0.0001). Similarly, aDI-rich IgG significantly enhanced the procoagulant activity of carotid artery endothelium and peritoneal macrophages isolated from experimental animals (P < 0.01). CONCLUSION: These data directly demonstrate that the ability to cause thrombosis in vivo is concentrated in the aDI fraction of aPL.


Assuntos
Anticorpos Antifosfolipídeos/farmacologia , Síndrome Antifosfolipídica/induzido quimicamente , Modelos Animais de Doenças , Imunoglobulina G/farmacologia , Camundongos , Trombose/induzido quimicamente , beta 2-Glicoproteína I/imunologia , Animais , Anticorpos Antifosfolipídeos/imunologia , Síndrome Antifosfolipídica/complicações , Síndrome Antifosfolipídica/imunologia , Imunoglobulina G/imunologia , Masculino , Estrutura Terciária de Proteína , Trombose/complicações , Trombose/imunologia
16.
Lupus ; 24(4-5): 448-62, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25801888

RESUMO

Oxidative stress has been shown to play a role in modifying antibodies in favor of higher auto-immunoreactivity. We studied the immunoreactivity of oxidized IgG (oxIgG) to ß2-glycoprotein I (ß2GPI), six peptide sequences corresponding to amino acid clusters on its different domains, to determine their effects on human coronary artery endothelial cells (HCAEC). Human IgG was purified from seven donors, electro-oxidized and checked for immunoreactivity and avidity to ß2GPI and to peptides by ELISA. Conformational stability and antibody-antigen complex formation of oxIgG was analyzed by fluorescence spectroscopy and dynamic light scattering. Resting and activated sub-confluent HCAEC were stimulated with oxIgG or IgG. Secreted cytokines were measured by ELISA. Immunoreactivity of seven oxIgG samples increased to 7.5-fold against ß2GPI and to 3.8-fold against six peptides as compared to IgG. oxIgG showed low avidity "properties." Conformational changes and exposure of protein hydrophobic regions were confirmed by an elevation in fluorescence (2.4- to 5.0-fold) on bis-ANS dye binding to oxIgG. oxIgG significantly elevated the release of GROα and IL-8 in resting and activated states of HCAEC. Oxidation alters IgG in favor of autoreactivity toward whole ß2GPI and corresponding peptides on different domains of ß2GPI and could lead to dysfunction of arterial endothelium by upregulation of chemokines.


Assuntos
Células Endoteliais/citologia , Imunoglobulina G/isolamento & purificação , Estresse Oxidativo , Peptídeos/imunologia , beta 2-Glicoproteína I/imunologia , Reações Antígeno-Anticorpo , Autoanticorpos/imunologia , Células Cultivadas , Vasos Coronários/citologia , Citocinas/imunologia , Difusão Dinâmica da Luz , Voluntários Saudáveis , Humanos , Imunoglobulina G/metabolismo
17.
Molecules ; 20(6): 10253-63, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-26046322

RESUMO

Copper(I)-catalyzed azide-alkyne cycloaddition, or CuAAC click chemistry, is an efficient method for bioconjugation aiming at chemical and biological applications. Herein, we demonstrate how the CuAAC method can provide novel phospholipid-protein conjugates with a high potential for the diagnostics and therapy of autoimmune conditions. In doing this, we, for the first time, covalently bind via 1,2,3-triazole linker biologically complementary molecules, namely phosphoethanol amine with human ß2-glycoprotein I and prothrombin. The resulting phospholipid-protein conjugates show high binding affinity and specificity for the autoimmune antibodies against autoimmune complexes. Thus, the development of this work might become a milestone in further diagnostics and therapy of autoimmune diseases that involve the production of autoantibodies against the aforementioned phospholipids and proteins, such as antiphospholipid syndrome and systemic lupus erythematosus.


Assuntos
Antígenos/química , Autoanticorpos/química , Etanolaminas/química , Imunoconjugados/química , Protrombina/química , beta 2-Glicoproteína I/química , Alcinos/química , Complexo Antígeno-Anticorpo/química , Antígenos/imunologia , Síndrome Antifosfolipídica/sangue , Síndrome Antifosfolipídica/imunologia , Azidas/química , Química Click , Reação de Cicloadição , Humanos , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Ligação Proteica , Soluções , Triazóis/química
18.
Cell Immunol ; 290(2): 226-32, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25108557

RESUMO

Our previous study has demonstrated that Toll-like receptor 4 (TLR4) can contribute to anti-ß2-glycoprotein I/ß2-glycoprotein I (anti-ß2GPI/ß2GPI)-induced tissue factor (TF) expression in human acute monocytic leukemia cell line THP-1. However, the role of TLR4 in the activation of autoimmune response in antiphospholipid syndrome (APS) has rarely been reported. In this study, we focused on the role of TLR4 in ß2GPI-induced maturation of bone marrow-derived dendritic cells (BMDCs). iDCs from C3H/HeN mice stimulated with ß2GPI were more mature than that from C3H/HeJ mice, yields of CD11c(+)MHCII(+)DCs, CD11c(+)CD80(+)DCs and CD11c(+)CD86(+)DCs and production of some pro-inflammatory cytokines in iDCs from C3H/HeN were higher than those from C3H/HeJ (p<0.05). Moreover, the ability of ß2GPI-treated iDCs from C3H/HeJ to stimulate proliferation of allogeneic mixed lymphocytes was lower than that of iDCs from C3H/HeN. In conclusion, our results indicate that TLR4 may play a significant role in ß2-glycoprotein I-induced BMDCs maturation.


Assuntos
Síndrome Antifosfolipídica/imunologia , Células Dendríticas/citologia , Receptor 4 Toll-Like/metabolismo , beta 2-Glicoproteína I/imunologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H
19.
J Autoimmun ; 55: 42-50, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24685231

RESUMO

The thrombogenic effect of ß2-glycoprotein I (ß2GPI)-dependent anti-phospholipid antibodies (aPL) in animal models was found to be LPS dependent. Since ß2GPI behaves as LPS scavenger, LPS/ß2GPI complex was suggested to account for in vitro cell activation through LPS/TLR4 involvement being LPS the actual bridge ligand between ß2GPI and TLR4 at least in monocytes/macrophages. However, no definite information is available on the interaction among ß2GPI, LPS and endothelial TLR4 in spite of the main role of endothelial cells (EC) in clotting. To analyse at the endothelial level the need of LPS, we investigated the in vitro interaction of ß2GPI with endothelial TLR4 and we assessed the role of LPS in such an interaction. To do this, we evaluated the direct binding and internalization of ß2GPI by confocal microscopy in living TLR4-MD2 transfected CHO cells (CHO/TLR4-MD2) and ß2GPI binding to CHO/TLR4-MD2 cells and human umbilical cord vein EC (HUVEC) by flow cytometry and cell-ELISA using anti-ß2GPI monoclonal antibodies in the absence or presence of various concentrations of exogenous LPS. To further investigate the role of TLR4, we performed anti-ß2GPI antibody binding and adhesion molecule up-regulation in TLR4-silenced HUVEC. Confocal microscopy studies show that ß2GPI does interact with TLR4 at the cell membrane and is internalized in cytoplasmic granules in CHO/TLR4-MD2 cells. ß2GPI binding to CHO/TLR4-MD2 cells and HUVEC is also confirmed by flow cytometry and cell-ELISA, respectively. The interaction between ß2GPI and TLR4 is confirmed by the reduction of anti-ß2GPI antibody binding and by the up-regulation of E-selectin or ICAM-1 by TLR4 silencing in HUVEC. ß2GPI binding is not affected by LPS at concentrations comparable to those found in both ß2GPI and antibody preparations. Only higher amount of LPS that can activate EC and up-regulate TLR4 expression are found to increase the binding. Our findings demonstrate that ß2GPI interacts directly with TLR4 expressed on EC, and that such interaction may contribute to ß2GPI-dependent aPL-mediated EC activation. At variance of monocytic cells, we also showed a threshold effect for the action of LPS, that is able to enhance anti-ß2GPI antibody EC binding only at cell activating concentrations, shown to increase TLR4 expression. This in vitro model may explain why LPS behaves as a second hit increasing the expression of ß2GPI in vascular tissues and triggering aPL-mediated thrombosis in experimental animals.


Assuntos
Anticorpos Antifosfolipídeos/imunologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Lipopolissacarídeos/toxicidade , Trombose/imunologia , Receptor 4 Toll-Like/imunologia , beta 2-Glicoproteína I/imunologia , Animais , Células CHO , Cricetinae , Cricetulus , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Modelos Imunológicos , Ligação Proteica , Trombose/induzido quimicamente , Trombose/genética , Trombose/patologia , Receptor 4 Toll-Like/genética , beta 2-Glicoproteína I/genética
20.
Lupus ; 23(14): 1468-76, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25164304

RESUMO

OBJECTIVES: The long-term prognosis of individuals fulfilling the laboratory criteria, but not clinical criteria, of antiphospholipid syndrome (APS) has not been widely investigated. The primary aim of this study was to evaluate the incidence of first thrombotic event (deep venous thrombosis (DVT), pulmonary embolism (PE), myocardial infarction (MI), stroke or transient ischaemic attack (TIA) in a nationwide antiphospholipid antibody (aPL) carrier cohort. DESIGN: We conducted a prospective nationwide cohort study. SETTING: The aPL profile of participants was recorded from the laboratory database. Information was collected about thrombotic and pregnancy complications, subsequent medical history, other risk factors for thrombosis, use of prophylactic antithrombotic medication and general health. PARTICIPANTS: Participants included adult asymptomatic aPL carriers recognized in Finland during 1971-2009. MAIN OUTCOME MEASURE: The main outcome measure was incidence of first thrombotic event. RESULTS: A total of 119 (89% female) aPL carriers were followed for mean (SD) of 9.1 (7.5) years (range 3-41 years). Sixty-one per cent of the study participants had autoimmune disease, most often systemic lupus erythematosus (SLE). Thirty-six of 119 (30%) were either double or triple positive, 56% single lupus anticoagulant (LA) positive, and 8% and 5% single anticardiolipin antibodies (aCL) and anti-ß2glycoprotein I antibodies (aß2GPI) positive, respectively. Nine (7.6%) study patients experienced a first thrombotic event (five DVT, one PE, two MI, one TIA) mean (SD) 7.2 (8.3) years (range 1-26 years) after aPL detection (annual incidence rate 0.8%). All individuals who developed thrombotic complications had autoimmune disease. Annual rate of first thrombotic event in carriers of single positivity (0.65%) was equal to the known risk of thrombosis in the healthy Caucasian population, whereas the rate was two times higher in carriers of double or triple positivity (1.27%). Sixteen of 79 (20%) women experienced pregnancy complications. CONCLUSIONS: Double or triple positivity for aPL is a risk factor for future thrombotic events, especially in individuals with an underlying autoimmune disease, whereas single positivity does not seem to carry an elevated risk of thrombosis.


Assuntos
Anticorpos Antifosfolipídeos/sangue , Doenças Autoimunes/epidemiologia , Ataque Isquêmico Transitório/epidemiologia , Infarto do Miocárdio/epidemiologia , Embolia Pulmonar/epidemiologia , Acidente Vascular Cerebral/epidemiologia , Trombose Venosa/epidemiologia , Aborto Espontâneo/epidemiologia , Aborto Espontâneo/imunologia , Adolescente , Adulto , Idoso , Anticorpos Anticardiolipina/sangue , Doenças Assintomáticas , Doenças Autoimunes/sangue , Doenças Autoimunes/complicações , Criança , Feminino , Fibrinolíticos/uso terapêutico , Finlândia/epidemiologia , Seguimentos , Humanos , Ataque Isquêmico Transitório/imunologia , Inibidor de Coagulação do Lúpus/sangue , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/imunologia , Gravidez , Estudos Prospectivos , Embolia Pulmonar/imunologia , Fatores de Risco , Acidente Vascular Cerebral/imunologia , Fatores de Tempo , Trombose Venosa/imunologia , Adulto Jovem , beta 2-Glicoproteína I/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA