Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 124(25): 3808-16, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25301710

RESUMEN

The effects of immunoglobulin G (IgG) from patients with the antiphospholipid syndrome (APS) upon monocyte activation have not been fully characterized. We carried out a comprehensive proteomic analysis of human monocytes treated with IgG from patients with different manifestations of the APS. Using 2-dimensional differential gel electrophoresis (2D DiGE), 4 of the most significantly regulated proteins (vimentin [VIM], zinc finger CCH domain-containing protein 18, CAP Gly domain-containing linker protein 2, and myeloperoxidase) were differentially regulated in monocytes treated with thrombotic or obstetric APS IgG, compared with healthy control (HC) IgG. These findings were confirmed by comparing monocytes isolated from APS patients and HC. Anti-VIM antibodies (AVAs) were significantly increased in 11 of 27 patients (40.7%) with APS. VIM expression on HC monocytes was stimulated more strongly by APS IgG from patients with higher-avidity serum AVA. We further characterized the proteome of thrombotic APS IgG-treated monocytes using a label-free proteomics technique. Of 12 proteins identified with the most confidence, 2 overlapped with 2D DiGE and many possessed immune response, cytoskeletal, coagulation, and signal transduction functions which are all relevant to APS and may therefore provide potential new therapeutic targets of this disease.


Asunto(s)
Síndrome Antifosfolípido/inmunología , Inmunoglobulina G/inmunología , Monocitos/inmunología , Proteoma/inmunología , Proteómica/métodos , Adulto , Síndrome Antifosfolípido/sangre , Western Blotting , Células Cultivadas , Cromatografía Liquida , Electroforesis en Gel Bidimensional , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Proteoma/genética , Proteoma/metabolismo , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masas en Tándem , Células U937
2.
Rheumatology (Oxford) ; 54(4): 722-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25273993

RESUMEN

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.


Asunto(s)
Anticuerpos Antifosfolípidos/farmacología , Síndrome Antifosfolípido/inducido químicamente , Modelos Animales de Enfermedad , Inmunoglobulina G/farmacología , Ratones , Trombosis/inducido químicamente , beta 2 Glicoproteína I/inmunología , Animales , Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/inmunología , Inmunoglobulina G/inmunología , Masculino , Estructura Terciaria de Proteína , Trombosis/complicaciones , Trombosis/inmunología
3.
Ann Rheum Dis ; 73(6): 1176-80, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23933625

RESUMEN

OBJECTIVE: To determine if proinflammatory and prothrombotic biomarkers are differentially upregulated in persistently antiphospholipid antibody (aPL)-positive patients, and to examine the effects of fluvastatin on these biomarkers. METHODS: Four groups of patients (age 18-65) were recruited: (a) primary antiphospholipid syndrome; (b) systemic lupus erythematosus (SLE) with antiphospholipid syndrome (APS) (SLE/APS); (c) persistent aPL positivity without SLE or APS (Primary aPL); and (d) persistent aPL positivity with SLE but no APS (SLE/aPL). The frequency-matched control group, used for baseline data comparison, was identified from a databank of healthy persons. Patients received fluvastatin 40 mg daily for 3 months. At 3 months, patients stopped the study medication and they were followed for another 3 months. Blood samples for 12 proinflammatory and prothrombotic biomarkers were collected monthly for 6 months. RESULTS: Based on the comparison of the baseline samples of 41 aPL-positive patients with 30 healthy controls, 9/12 (75%) biomarkers (interleukin (IL)-6, IL1ß, vascular endothelial growth factor (VEGF), tumour necrosis factor (TNF)-α, interferon (IFN)-α, inducible protein-10 (IP10), soluble CD40 ligand (sCD40L), soluble tissue factor (sTF) and intracellular cellular adhesion molecule (ICAM)-1) were significantly elevated. Twenty-four patients completed the study; fluvastatin significantly and reversibly reduced the levels of 6/12 (50%) biomarkers (IL1ß, VEGF, TNFα, IP10, sCD40L and sTF). CONCLUSIONS: Our prospective mechanistic study demonstrates that proinflammatory and prothrombotic biomarkers, which are differentially upregulated in persistently aPL-positive patients, can be reversibly reduced by fluvastatin. Thus, statin-induced modulation of the aPL effects on target cells can be a valuable future approach in the management of aPL-positive patients.


Asunto(s)
Síndrome Antifosfolípido/tratamiento farmacológico , Ácidos Grasos Monoinsaturados/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Indoles/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Adulto , Síndrome Antifosfolípido/inmunología , Síndrome Antifosfolípido/metabolismo , Biomarcadores/sangre , Moléculas de Adhesión Celular/metabolismo , Citocinas/inmunología , Femenino , Fluvastatina , Humanos , Mediadores de Inflamación/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Masculino , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Tromboplastina/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
J Autoimmun ; 54: 72-80, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24972993

RESUMEN

Tolerogenic dendritic cells (tDCs) have the potential to control the outcome of autoimmunity by modulating the immune response. The aim of this study was to uncover the tolerance efficacy attributed to beta-2-glycoprotein-I (ß2GPI) tDCs or ß2GPI domain-I (D-I) and domain-V (D-V)-tDCs in mice with antiphospholipid syndrome (APS). tDCs were pulsed with ß2GPI or D-I or D-V derivatives. Our results revealed that ß2GPI related tDCs phenotype includes CD80(high), CD86(high) CD40(high) MHC class II(high). The miRNA profiling encompass miRNA 23b(high), miRNA 142-3p(low) and miRNA 221(low). In addition the ß2GPI related tDCs showed reduced secretion of IL-1ß, IL-12 and IL-23. D-I tDCs treatment was more efficient than ß2GPI tDCs in inducing of tolerance in APS mice, manifested by lowered titers of anti- ß2GPI antibodies (Abs) and reduced percentage of fetal loss. Tolerance induction was accompanied by poor T cell response to ß2GPI, high numbers of CD4 + CD25 + FOXP3 + T-regulatory cells (Treg), reduced levels of IFNγ, IL-17 and increased expression of IL-10 and TGFß. Tolerance was successfully transferred by Treg cells from the tolerized mice to ß2GPI immunized mice. We conclude that predominantly D-I-tDCs and ß2GPI tDCs have the potential to attenuate experimental APS by induction of Treg cells, reduction of anti- ß2GPI Abs titers and increased expression of anti-inflammatory cytokines. We suggest that ß2-GPI-D-I-tDCs may offer a novel approach for developing therapy for APS patients.


Asunto(s)
Síndrome Antifosfolípido , Células Dendríticas/inmunología , Tolerancia Inmunológica/efectos de los fármacos , beta 2 Glicoproteína I , Animales , Antígenos de Diferenciación/inmunología , Síndrome Antifosfolípido/tratamiento farmacológico , Síndrome Antifosfolípido/inmunología , Síndrome Antifosfolípido/patología , Citocinas/inmunología , Células Dendríticas/patología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos BALB C , Compuestos Orgánicos/inmunología , Compuestos Orgánicos/farmacología , Estructura Terciaria de Proteína , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , beta 2 Glicoproteína I/inmunología , beta 2 Glicoproteína I/farmacología
5.
Arthritis Rheum ; 65(12): 3186-93, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23983008

RESUMEN

OBJECTIVE: To examine the prevalence of isolated IgA anti-ß2 -glycoprotein I (anti-ß2 GPI) positivity and the association of these antibodies, and a subgroup that bind specifically to domain IV/V of ß2 GPI, with clinical manifestations of the antiphospholipid syndrome (APS) in 3 patient groups and to evaluate the pathogenicity of IgA anti-ß2 GPI in a mouse model of thrombosis. METHODS: Patients with systemic lupus erythematosus (SLE) from a multiethnic, multicenter cohort (LUpus in MInorities, NAture versus nurture [LUMINA]) (n = 558), patients with SLE from the Hopkins Lupus Cohort (n = 215), and serum samples referred to the Antiphospholipid Standardization Laboratory (APLS) (n = 5,098) were evaluated. IgA anti-ß2 GPI titers and binding to domain IV/V of ß2 GPI were examined by enzyme-linked immunosorbent assay (ELISA). CD1 mice were inoculated with purified IgA anti-ß2 GPI antibodies, and surgical procedures and ELISAs were performed to evaluate thrombus development and tissue factor (TF) activity. RESULTS: A total of 198 patients were found to be positive for IgA anti-ß2 GPI isotype, and 57 patients were positive exclusively for IgA anti-ß2 GPI antibodies. Of these, 13 of 23 patients (56.5%) in the LUMINA cohort, 17 of 17 patients (100%) in the Hopkins cohort, and 10 of 17 patients (58.9%) referred to APLS had at least one APS-related clinical manifestation. Fifty-four percent of all the IgA anti-ß2 GPI-positive serum samples reacted with domain IV/V of anti-ß2 GPI, and 77% of those had clinical features of APS. Isolated IgA anti-ß2 GPI positivity was associated with an increased risk of arterial thrombosis (P < 0.001), venous thrombosis (P = 0.015), and all thrombosis (P < 0.001). The association between isolated IgA anti-ß2 GPI and arterial thrombosis (P = 0.0003) and all thrombosis (P = 0.0003) remained significant after adjusting for other risk factors for thrombosis. In vivo mouse studies demonstrated that IgA anti-ß2 GPI antibodies induced significantly larger thrombi and higher TF levels compared to controls. CONCLUSION: Isolated IgA anti-ß2 GPI-positive titers may identify additional patients with clinical features of APS. Testing for these antibodies when other antiphospholipid tests are negative and APS is suspected is recommended. IgA anti-ß2 GPI antibodies directed to domain IV/V of ß2 GPI represent an important subgroup of clinically relevant antiphospholipids.


Asunto(s)
Anticuerpos Antiidiotipos/sangre , Síndrome Antifosfolípido/diagnóstico , Autoanticuerpos/sangre , Inmunoglobulina A/sangre , beta 2 Glicoproteína I/inmunología , Animales , Síndrome Antifosfolípido/sangre , Síndrome Antifosfolípido/inmunología , Humanos , Estudios Longitudinales , Ratones , Prevalencia , Trombosis/diagnóstico , Trombosis/inmunología
6.
Blood ; 117(4): 1408-14, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21119114

RESUMEN

Antiphospholipid (aPL)/anti-ß(2) glycoprotein I (anti-ß(2)GPI) antibodies stimulates tissue factor (TF) expression within vasculature and in blood cells, thereby leading to increased thrombosis. Several cellular receptors have been proposed to mediate these effects, but no convincing evidence for the involvement of a specific one has been provided. We investigated the role of Apolipoprotein E receptor 2 (ApoER2') on the pathogenic effects of a patient-derived polyclonal aPL IgG preparation (IgG-APS), a murine anti-ß(2)GPI monoclonal antibody (E7) and of a constructed dimeric ß(2)GPI I (dimer), which in vitro mimics ß(2)GPI-antibody immune complexes, using an animal model of thrombosis, and ApoER2-deficient (-/-) mice. In wild type mice, IgG-APS, E7 and the dimer increased thrombus formation, carotid artery TF activity as well as peritoneal macrophage TF activity/expression. Those pathogenic effects were significantly reduced in ApoER2 (-/-) mice. In addition, those effects induced by the IgG-APS, by E7 and by the dimer were inhibited by treatment of wild-type mice with soluble binding domain 1 of ApoER2 (sBD1). Altogether these data show that ApoER2 is involved in pathogenesis of antiphospholipids antibodies.


Asunto(s)
Síndrome Antifosfolípido/genética , Proteínas Relacionadas con Receptor de LDL/fisiología , Trombosis/genética , Animales , Anticuerpos Antifosfolípidos/efectos adversos , Anticuerpos Antifosfolípidos/metabolismo , Anticuerpos Antifosfolípidos/farmacología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Anticuerpos Fosfo-Específicos/administración & dosificación , Anticuerpos Fosfo-Específicos/efectos adversos , Anticuerpos Fosfo-Específicos/farmacología , Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/patología , Síndrome Antifosfolípido/prevención & control , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/efectos adversos , Inmunoglobulina G/farmacología , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Trombosis/etiología , Trombosis/patología , beta 2 Glicoproteína I/inmunología
7.
Ann Rheum Dis ; 71(1): 120-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21914629

RESUMEN

OBJECTIVES: The importance of ß(2)-glycoprotein I (ß(2)GPI)-specific CD4(+) T cells in the development of pathogenic processes in patients with antiphospholipid syndrome (APS) and APS mouse models is well established. Therefore, our objective is to manipulate the ß2GPI specific CD4(+) T cells using tolerogenic dendritic cells (tDCs) to induce tolerance. We aim to evaluate the capability of tDCs to induce antigen-specific tolerance in effector/memory T cells from patients with APS and to elucidate the involved mechanism. METHODS: DCs and tDCs were produced from patients with APS peripheral-blood-monocytes, using specific cytokines. ß(2)GPI-specific tolerance induction was investigated by coculturing control DC (cDC) or tDC, ß(2)GPI-loaded, with autologous effector/memory T cells, evaluating the proliferative response, phenotype, cytokines secretion, viability and regulatory T cells. RESULTS: Human monocyte-derived DCs treated with interleukin (IL)-10 and transforming growth factor ß-1 (10/TGF-DC) induced ß(2)GPI-specific-unresponsiveness in effector/memory CD4(+) T cells (46.5% ± 26.0 less proliferation) in 16 of 20 analysed patients with APS, without affecting the proliferative response to an unrelated candidin. In five analysed patients, 10/TGF-DC-stimulated T cells acquired an IL-2(low)interferon γ(low)IL-10(high) cytokine profile, with just a propensity to express higher numbers of Foxp3(+)CTLA-4(+) cells, but with an evident suppressive ability. In four of 10 analysed patients, 10/TGF-DC-stimulated T cell hyporesponsiveness could not be reverted and showed higher percentages of late apoptosis, p<0.02. CONCLUSIONS: The inherent tolerance induction resistance of activated T cells present during the development of autoimmune diseases has delayed the application of tDC as an alternative therapy. This study highlights the 10/TGF-DC feasibility to induce antigen-specific unresponsiveness in autoreactive T cells generated in patients with APS by inducing apoptosis or T cells with regulatory abilities.


Asunto(s)
Síndrome Antifosfolípido/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , beta 2 Glicoproteína I/inmunología , Adulto , Anciano , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/biosíntesis , Femenino , Humanos , Tolerancia Inmunológica/inmunología , Memoria Inmunológica/inmunología , Inmunofenotipificación , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad
8.
Semin Thromb Hemost ; 38(4): 360-74, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22576664

RESUMEN

Positive results in the anticardiolipin antibody (aCL), anti-ß2-glycoprotein I antibody (aß2GPI), and/or lupus anticoagulant (LA) assays constitute the laboratory criteria for the definite diagnosis of the antiphospholipid syndrome (APS). These tests became available from as early as the1980s, and since then several novel assays have become available that have varied usefulness in the diagnosis and prognosis of APS patients. For almost three decades there has been an ongoing effort to standardize the aCL, aß2GPI, and LA assays, but there are still reports of significant intra- and interlaboratory variation in the results of all three assays. There have also been numerous issues with the implementation of novel (noncriteria) antiphospholipid antibody (aPL) tests in standard testing panels, due to either lack of standardized testing methods or limited evidence of their clinical utility in APS patients. At the recent 13th International Congress on Antiphospholipid Antibodies (APLA 2010, 13-16 April 2010, Galveston, TX), two task forces were set up to address these problems. This review gives a general description of current problems hindering the standardization of aPL tests and the implementation of novel assays as standard components of aPL testing panels. It also highlights the approach used by APLA 2010 Task Forces to address these problems and presents their recommendations.


Asunto(s)
Anticuerpos Antifosfolípidos/análisis , Síndrome Antifosfolípido/diagnóstico , Anticuerpos Antifosfolípidos/sangre , Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/inmunología , Femenino , Humanos , Agencias Internacionales , Inhibidor de Coagulación del Lupus/sangre , Inhibidor de Coagulación del Lupus/inmunología , Embarazo , Complicaciones del Embarazo , beta 2 Glicoproteína I/sangre , beta 2 Glicoproteína I/inmunología
9.
Semin Thromb Hemost ; 38(4): 305-21, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22510982

RESUMEN

The presence of pathogenic antiphospholipid antibodies (aPL) is the characterizing feature of the antiphospholipid syndrome (APS), mediating the recurrent pregnancy loss and thrombosis typical of the disease, through their action on various antigenic targets. Despite the available knowledge regarding the mechanisms by which aPL induce a procoagulant phenotype in the vasculature and abnormal cellular proliferation and differentiation in placental tissues to cause the typical clinical features, these processes still remain incompletely understood. It is also known that inflammation serves as a necessary link between the observed procoagulant phenotype and actual thrombus development, and is an important mediator of the placental injury in APS patients. Even less well understood are the processes underlying the ontogeny of these pathogenic antibodies. This review seeks to highlight what is known about the mechanisms that contribute to the origin of pathogenic aPL and to the action of these antibodies on target antigens that produce the pathological features of APS. We will also examine the feasibility of classifying patients in clinical phenotypes related to underlying pathophysiological mechanisms, and how this could impact the management of patients with novel "targeted" therapeutic strategies.


Asunto(s)
Síndrome Antifosfolípido/etiología , Animales , Síndrome Antifosfolípido/genética , Síndrome Antifosfolípido/patología , Síndrome Antifosfolípido/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Embarazo , Complicaciones del Embarazo/etiología
10.
J Autoimmun ; 38(2-3): J209-15, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22196923

RESUMEN

ß2 glycoprotein I (ß2GPI)-dependent anti-phospholipid antibodies (aPL) induce thrombosis and affect pregnancy. The CMV-derived synthetic peptide TIFI mimics the PL-binding site of ß2GPI and inhibits ß2GPI cell-binding in vitro and aPL-mediated thrombosis in vivo. Here we investigated the effect of TIFI on aPL-induced fetal loss in mice. TIFI inhibitory effect on in vitro aPL binding to human trophoblasts was evaluated by indirect immunofluorescence and ELISA. TIFI effect on aPL-induced fetal loss was investigated in pregnant C57BL/6 mice treated with aPL or normal IgG (NHS). Placenta/fetus weight and histology and RNA expression were analyzed. TIFI, but not the control peptide VITT, displayed a dose-dependent inhibition of aPL binding to trophoblasts in vitro. Injection of low doses of aPL at day 0 of pregnancy caused growth retardation and increased fetal loss rate, both significantly reduced by TIFI but not VITT. Consistent with observations in humans, histological analysis showed no evidence of inflammation in this model, as confirmed by the absence of an inflammatory signature in gene expression analysis, which in turn revealed a TIFI-dependent modulation of molecules involved in differentiation and development processes. These findings support the non-inflammatory pathogenic role of aPL and suggest innovative therapeutic approaches to aPL-dependent fetal loss.


Asunto(s)
Aborto Espontáneo/inmunología , Anticuerpos Antifosfolípidos/inmunología , Péptidos/metabolismo , beta 2 Glicoproteína I/química , beta 2 Glicoproteína I/metabolismo , Aborto Espontáneo/prevención & control , Animales , Anticuerpos Antifosfolípidos/metabolismo , Sitios de Unión , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Péptidos/administración & dosificación , Péptidos/farmacología , Placenta/metabolismo , Placenta/patología , Embarazo , Unión Proteica , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo
11.
J Immunol ; 184(12): 6622-8, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20483743

RESUMEN

A major mechanism of hypercoagulability in the antiphospholipid syndrome (APS) is antiphospholipid Ab-mediated upregulation of tissue factor (TF) on monocytes via activation of TLRs, p38 MAPK, and NF-kappaB pathways. We examined whether monocyte signaling pathways are differentially activated by IgG from patients with vascular thrombosis (VT) alone compared with IgG from patients with pregnancy morbidity (PM) alone. We purified IgG from 49 subjects. A human monocyte cell line and ex vivo healthy monocytes were treated with 100 microg/ml IgG for 6 h, and cell extracts were examined by immunoblot using Abs to p38 MAPK and NF-kappaB. To further investigate intracellular signaling pathways induced by these IgGs, specific inhibitors of p38 MAPK, NF-kappaB, TLR4, and TLR2 were used to determine their effect on TF activity. Only IgG from patients with VT but no PM (VT+/PM-) caused phosphorylation of NF-kappaBand p38 MAPK and upregulation of TF activity in monocytes. These effects were not seen with IgG from patients with PM alone (VT-/PM+), anti-phospholipid Ab-positive patients without APS, or healthy controls. TF upregulation caused by the VT+/PM- samples was reduced by inhibitors of p38 MAPK, NF-kappaB, and TLR4. The effects of VT+/PM- IgG on signaling and TF upregulation were concentrated in the fraction that bound beta-2-glycoprotein I. Our findings demonstrate that IgGs from patients with diverse clinical manifestations of APS have differential effects upon phosphorylation of NF-kappaB and p38 MAPK and TF activity that may be mediated by differential activation of TLR4.


Asunto(s)
Síndrome Antifosfolípido/inmunología , Inmunoglobulina G/inmunología , Monocitos/inmunología , Transducción de Señal/inmunología , Tromboplastina/inmunología , Adulto , Síndrome Antifosfolípido/metabolismo , Western Blotting , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Fosforilación , Embarazo , Complicaciones del Embarazo/inmunología , Tromboplastina/metabolismo , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Blood ; 114(14): 3074-83, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19628708

RESUMEN

Antiphospholipid (aPL) antibodies recognize receptor-bound beta(2) glycoprotein I (beta(2)GPI) on target cells, and induce an intracellular signaling and a procoagulant/proinflammatory phenotype that leads to thrombosis. Evidence indicates that annexin A2 (A2), a receptor for tissue plasminogen activator and plasminogen, binds beta(2)GPI on target cells. However, whether A2 mediates pathogenic effects of aPL antibodies in vivo is unknown. In this work, we studied the effects of human aPL antibodies in A2-deficient (A2(-/-)) mice. A2(-/-) and A2(+/+) mice were injected with immunoglobulin G (IgG) isolated from either a patient with antiphospholipid syndrome (IgG-APS), a healthy control subject (IgG-normal human serum), a monoclonal anti-beta(2)GPI antibody (4C5), an anti-A2 monoclonal antibody, or monoclonal antibody of irrelevant specificity as control. We found that, after IgG-APS or 4C5 injections and vascular injury, mean thrombus size was significantly smaller and tissue factor activity was significantly less in A2(-/-) mice compared with A2(+/+) mice. The expression of vascular cell adhesion molecule-1 induced by IgG-APS or 4C5 in explanted A2(-/-) aorta was also significantly reduced compared with A2(+/+) mice. Interestingly, anti-A2 monoclonal antibody significantly decreased aPL-induced expression of intercellular cell adhesion molecule-1, E-selectin, and tissue factor activity on cultured endothelial cells. Together, these data indicate for the first time that A2 mediates the pathogenic effects of aPL antibodies in vivo and in vitro APS.


Asunto(s)
Anexina A2/fisiología , Anticuerpos Antifosfolípidos/farmacología , Síndrome Antifosfolípido/metabolismo , Síndrome Antifosfolípido/patología , Aorta/patología , Arterias Carótidas/patología , Animales , Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/inmunología , Aorta/lesiones , Aorta/metabolismo , Western Blotting , Enfermedades Cardiovasculares , Arterias Carótidas/metabolismo , Células Cultivadas , Selectina E/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Técnicas In Vitro , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Tromboplastina/metabolismo , Venas Umbilicales/citología , Venas Umbilicales/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
13.
Arthritis Rheum ; 62(7): 2064-72, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20201076

RESUMEN

OBJECTIVE: Free radical-mediated reactions have been implicated as contributors in a number of autoimmune diseases, including systemic lupus erythematosus (SLE). However, the potential for oxidative/nitrosative stress to elicit an autoimmune response or to contribute to disease pathogenesis, and thus be useful when determining a prognosis, remains largely unexplored in humans. This study was undertaken to investigate the status and contribution of oxidative/nitrosative stress in patients with SLE. METHODS: Sera from 72 SLE patients with varying levels of disease activity according to the SLE Disease Activity Index (SLEDAI) and 36 age- and sex-matched healthy controls were evaluated for serum levels of oxidative/nitrosative stress markers, including antibodies to malondialdehyde (anti-MDA) protein adducts and to 4-hydroxynonenal (anti-HNE) protein adducts, MDA/HNE protein adducts, superoxide dismutase (SOD), nitrotyrosine (NT), and inducible nitric oxide synthase (iNOS). RESULTS: Serum analysis showed significantly higher levels of both anti-MDA/anti-HNE protein adduct antibodies and MDA/HNE protein adducts in SLE patients compared with healthy controls. Interestingly, not only was there an increased number of subjects positive for anti-MDA or anti-HNE antibodies, but also the levels of both of these antibodies were statistically significantly higher among SLE patients whose SLEDAI scores were > or = 6 as compared with SLE patients with lower SLEDAI scores (SLEDAI score <6). In addition, a significant correlation was observed between the levels of anti-MDA or anti-HNE antibodies and the SLEDAI score (r = 0.734 and r = 0.647, respectively), suggesting a possible causal relationship between these antibodies and SLE. Furthermore, sera from SLE patients had lower levels of SOD and higher levels of iNOS and NT compared with healthy control sera. CONCLUSION: These findings support an association between oxidative/nitrosative stress and SLE. The stronger response observed in serum samples from patients with higher SLEDAI scores suggests that markers of oxidative/nitrosative stress may be useful in evaluating the progression of SLE and in elucidating the mechanisms of disease pathogenesis.


Asunto(s)
Lupus Eritematoso Sistémico/sangre , Estrés Oxidativo , Adulto , Anciano , Biomarcadores/sangre , Progresión de la Enfermedad , Femenino , Estado de Salud , Humanos , Lupus Eritematoso Sistémico/patología , Lupus Eritematoso Sistémico/fisiopatología , Masculino , Malondialdehído/sangre , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/sangre , Índice de Severidad de la Enfermedad , Superóxido Dismutasa/sangre , Tirosina/análogos & derivados , Tirosina/sangre , Adulto Joven
14.
J Immunol ; 182(8): 4836-43, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19342662

RESUMEN

The mechanisms by which antiphospholipid Abs (aPL) cause thrombosis are not fully understood. It is clear that binding to a number of phospholipid-associated Ags is important but it is difficult to identify which Ag-binding properties are most closely linked to the ability to cause biologic effects such as promotion of thrombosis and activation of endothelial cells. We have previously used an in vitro expression system to produce a panel of human monoclonal IgG molecules between which we engineered small differences in sequence leading to significant well-defined changes in binding properties. In this study, we assess the properties of five of these IgG molecules in assays of biologic function in vitro and in vivo. The i.p. injection of these IgG into mice subjected to a femoral vein pinch stimulus showed that only those IgG that showed strong binding to thrombin promoted in vivo venous thrombosis and leukocyte adherence. However, this finding did not hold true for the effects of these IgG on activation of cultured endothelial cells in vitro, where there was a less clear relationship between binding properties and biologic effects.


Asunto(s)
Anticuerpos Antifosfolípidos/inmunología , Anticuerpos Antifosfolípidos/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Trombina/metabolismo , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/inmunología , Humanos , Ratones , Unión Proteica , Cordón Umbilical/citología , Cordón Umbilical/inmunología
15.
J Exp Med ; 195(2): 211-20, 2002 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-11805148

RESUMEN

The antiphospholipid syndrome (APS) is characterized by recurrent fetal loss, vascular thrombosis, and thrombocytopenia occurring in the presence of antiphospholipid (aPL) antibodies. The pathogenesis of fetal loss and tissue injury in APS is incompletely understood, but is thought to involve platelet and endothelial cell activation as well as procoagulant effects of aPL antibodies acting directly on clotting pathway components. Recent studies have shown that uncontrolled complement activation in the placenta leads to fetal death in utero. We hypothesized that aPL antibodies activate complement in the placenta, generating split products that mediate placental injury and lead to fetal loss and growth retardation. To test this hypothesis, we used a murine model of APS in which pregnant mice are injected with human IgG containing aPL antibodies. We found that inhibition of the complement cascade in vivo, using the C3 convertase inhibitor complement receptor 1-related gene/protein y (Crry)-Ig, blocks fetal loss and growth retardation. Furthermore, mice deficient in complement C3 were resistant to fetal injury induced by aPL antibodies. While antigenic epitopes recognized by aPL antibodies are important in the pathogenesis of APS, our data show that in vivo complement activation is required for aPL antibody-induced fetal loss and growth retardation.


Asunto(s)
Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/inmunología , Complemento C3/inmunología , Muerte Fetal/inmunología , Animales , Síndrome Antifosfolípido/sangre , Síndrome Antifosfolípido/fisiopatología , Activación de Complemento/inmunología , Complemento C3/genética , Epítopos/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Embarazo
17.
J Neuroimmunol ; 196(1-2): 101-6, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18455242

RESUMEN

C5 deficient mice are highly resistant to experimental autoimmune myasthenia gravis (EAMG) despite intact immune response to acetylcholine receptor (AChR), validating the pivotal role played by membrane attack complex (MAC, C5b-9) in neuromuscular junction destruction. To distinguish the significance of C5a from that of C5b in EAMG pathogenesis, C5a receptor (C5aR) knockout (KO) and wild-type (WT) mice were immunized with AChR to induce pathogenic anti-AChR antibodies. In contrast with C5 deficient mice, C5aR KO mice were equally susceptible to EAMG as WT mice and exhibited comparable antibody and lymphocyte proliferation response to AChR implicating that C5a is not involved in EAMG development.


Asunto(s)
Miastenia Gravis Autoinmune Experimental/inmunología , Receptor de Anafilatoxina C5a/fisiología , Receptores Colinérgicos/inmunología , Animales , Anticuerpos/metabolismo , Complejo Antígeno-Anticuerpo , Complemento C5a/genética , Complemento C5a/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Centro Germinal/inmunología , Ratones , Ratones Noqueados , Miastenia Gravis Autoinmune Experimental/genética , Radioinmunoensayo/métodos , Receptor de Anafilatoxina C5a/deficiencia , Estadísticas no Paramétricas
18.
Semin Thromb Hemost ; 34(4): 313-28, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18814064

RESUMEN

The anticardiolipin (aCL) test has been widely used by physicians since the mid-1980s for diagnosing patients with antiphospholipid syndrome (APS). Establishment of this diagnosis has enabled effective management of patients with recurrent thrombosis and recurrent pregnancy losses. The test was first established in 1983 as a radioimmunoassay and soon thereafter converted into an enzyme-linked immunosorbent assay (ELISA). The other test commonly used in the diagnosis of APS is the lupus anticoagulant (LA) test. The aCL ELISA is sensitive for the diagnosis of APS but lacks specificity. On the other hand, the LA assay, although more specific, is not as sensitive as the aCL ELISA. More specific tests are now available such as the anti-beta2 glycoprotein I (anti-beta2GPI) assay, the antiprothrombin assay, and other ELISAs that use negatively charged phospholipids instead of cardiolipin to coat the plates. In the past 25 years, there have been numerous efforts to standardize aCL, LA, and anti-beta2GPI tests but there are still reports of significant intra- and interlaboratory variation in results for all three assays. This article discusses in detail the clinical value of these tests, technical problems associated with their use, the current laboratory classification criteria for diagnosis of APS, and possible new and better assays that will be available in the near future for diagnosis of APS.


Asunto(s)
Anticuerpos Anticardiolipina/sangre , Síndrome Antifosfolípido/sangre , Síndrome Antifosfolípido/diagnóstico , Ensayo de Inmunoadsorción Enzimática/normas , Ensayo de Inmunoadsorción Enzimática/tendencias , Aborto Habitual/sangre , Aborto Habitual/diagnóstico , Aborto Habitual/historia , Aborto Habitual/inmunología , Anticuerpos Anticardiolipina/inmunología , Síndrome Antifosfolípido/historia , Síndrome Antifosfolípido/inmunología , Ensayo de Inmunoadsorción Enzimática/historia , Femenino , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Masculino , Embarazo , Trombina/inmunología , Trombosis/sangre , Trombosis/diagnóstico , Trombosis/historia , Trombosis/inmunología , beta 2 Glicoproteína I/inmunología
20.
Expert Rev Mol Med ; 9(30): 1-15, 2007 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-17997880

RESUMEN

Antiphospholipid (aPL) antibodies (Abs) are associated with thrombosis and pregnancy loss in antiphospholipid syndrome (APS), a disorder initially characterised in patients with systemic lupus erythematosus (SLE) but now known to occur in the absence of other autoimmune disease. There is strong evidence that aPL Abs are pathogenic in vivo, from studies of animal models of thrombosis, endothelial cell activation and pregnancy loss. In recent years, progress has been made in characterising the molecular basis of this pathogenicity, which includes direct effects on platelets, endothelial cells and monocytes as well as activation of complement. This review summarises the clinical manifestations of APS and current modalities of treatment, and explains recent advances in understanding the molecular events triggered by aPL Abs on target cells in coagulation pathways as well as effects of aPL Abs on complement activation. Based on this information and on additional scientific evidence using in vitro and in vivo models, new potential targeted therapies for treatment and/or prevention of thrombosis in APS are proposed and discussed.


Asunto(s)
Anticuerpos Antifosfolípidos/sangre , Síndrome Antifosfolípido , Trombosis/tratamiento farmacológico , Animales , Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/tratamiento farmacológico , Síndrome Antifosfolípido/inmunología , Síndrome Antifosfolípido/metabolismo , Síndrome Antifosfolípido/fisiopatología , Plaquetas/inmunología , Plaquetas/fisiología , Activación de Complemento , Células Endoteliales/fisiología , Humanos , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Monocitos/fisiología , Protrombina/inmunología , Protrombina/metabolismo , Trombosis/inmunología , Trombosis/metabolismo , beta 2 Glicoproteína I/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA