Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 280
Filter
1.
J Reprod Immunol ; 164: 104255, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38797133

ABSTRACT

Women with antiphospholipid syndrome (APS) are at high risk for miscarriage and preeclampsia. Unlike pro-thrombotic systemic APS, obstetric APS is associated with insufficient placentation, as well as inflammation and vascular dysfunction at the maternal-fetal interface. Antiphospholipid antibodies (aPL) can target the placental trophoblast and induce inflammation. We reported that aPL trigger trophoblast cells to produce elevated levels of IL-8 through activation of Toll-like receptor 4 (TLR4). Downstream of TLR4, we found this IL-8 response is mediated by a TLR8-activating microRNA (miR), miR-146a-3p, which is also released by the trophoblast via extracellular vesicles (EVs). Since endothelial dysfunction is a feature of obstetric APS, we sought to determine if other miRs that can activate the RNA sensors, TLR7 and/or TLR8, are released by the trophoblast via EVs after exposure to aPL, and if these EVs can activate human endometrial endothelial cells (HEECs). Using a human first trimester extravillous trophoblast cell line we found that aPL elevated their release of small EVs (<150 nm). These extracellular vesicles released from trophoblast cells exposed to aPL expressed elevated levels of TLR7/8-activating miR-21a and miR-29a, in addition to the previously reported miR-146a-3p. Extracellular vesicles from aPL-exposed human trophoblast cells triggered human endometrial endothelial cells to generate an inflammatory IL-8 response, in part through TLR7. This study highlights EVs as a mode of communication between the placenta and the maternal vasculature, as well as a potential role for TLR7/8-activating miRs in contributing to inflammation at the maternal-fetal interface in obstetric APS.


Subject(s)
Antibodies, Antiphospholipid , Antiphospholipid Syndrome , Extracellular Vesicles , MicroRNAs , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Trophoblasts , Humans , Female , Trophoblasts/metabolism , Trophoblasts/immunology , MicroRNAs/metabolism , MicroRNAs/genetics , Toll-Like Receptor 7/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Pregnancy , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/immunology , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/metabolism , Antibodies, Antiphospholipid/immunology , Antibodies, Antiphospholipid/metabolism , Endometrium/metabolism , Endometrium/immunology , Endometrium/pathology , Endothelial Cells/metabolism , Endothelial Cells/immunology , Cell Line , Interleukin-8/metabolism
2.
Adv Sci (Weinh) ; 11(24): e2309140, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38639399

ABSTRACT

Antiphospholipid syndrome (APS) is characterized by thrombus formation, poor pregnancy outcomes, and a proinflammatory response. H3K4me3-related monocytes activation are key regulators of APS pathogenesis. Therefore, H3K4me3 CUT&Tag and ATAC-seq are performed to examine the epigenetic profiles. The results indicate that the H3K4me3 signal and chromatin accessibility at the FOXJ2 promoter are enhanced in an in vitro monocyte model by stimulation with ß2GPI/anti-ß2GPI, which mimics APS, and decreases after OICR-9429 administration. Furthermore, FOXJ2 is highly expressed in patients with primary APS (PAPS) and is the highest in patients with triple-positive antiphospholipid antibodies (aPLs). Mechanistically, FOXJ2 directly binds to the SLAMF8 promoter and activates SLAMF8 transcription. SLAMF8 further interacts with TREM1 to stimulate TLR4/NF-κB signaling and prohibit autophagy. Knockdown of FOXJ2, SLAMF8, or TREM1 blocks TLR4/NF-κB and provokes autophagy, subsequently inhibiting the release of inflammatory and thrombotic indicators. A mouse model of vascular APS is established via ß2GPI intraperitoneal injection, and the results suggest that OICR-9429 administration attenuates the inflammatory response and thrombus formation by inactivating FOXJ2/SLAMF8/TREM1 signaling. These findings highlight the overexpression of H3K4me3-mediated FOXJ2 in APS, which consequently accelerates APS pathogenesis by triggering inflammation and thrombosis via boosting the SLAMF8/TREM1 axis. Therefore, OICR-9429 is a promising candidate drug for APS therapy.


Subject(s)
Disease Models, Animal , Forkhead Transcription Factors , Inflammation , Monocytes , Thrombosis , Animals , Female , Humans , Mice , Antibodies, Antiphospholipid/metabolism , Antiphospholipid Syndrome/metabolism , Antiphospholipid Syndrome/genetics , beta 2-Glycoprotein I/metabolism , beta 2-Glycoprotein I/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Histones/metabolism , Histones/genetics , Inflammation/metabolism , Inflammation/genetics , Monocytes/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Thrombosis/metabolism , Thrombosis/genetics
3.
Blood ; 143(19): 2005-2011, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38437497

ABSTRACT

ABSTRACT: Antiprothrombin antibodies are found in antiphospholipid patients, but how they interact with prothrombin remains elusive. Prothrombin adopts closed and open forms. We recently discovered type I and type II antibodies and proposed that type I recognizes the open form. In this study, we report the discovery and structural and functional characterization in human plasma of a type I antibody, POmAb (prothrombin open monoclonal antibody). Using surface plasmon resonance and single-molecule spectroscopy, we show that POmAb interacts with kringle-1 of prothrombin, shifting the equilibrium toward the open form. Using single-particle cryogenic electron microscopy (cryo-EM), we establish that the epitope targeted by POmAb is in kringle-1, comprising an extended binding interface centered at residues R90-Y93. The 3.2-Å cryo-EM structure of the complex reveals that the epitope overlaps with the position occupied by the protease domain of prothrombin in the closed state, explaining the exclusive binding of POmAb to the open form. In human plasma, POmAb prolongs phospholipid-initiated and diluted Russell's viper venom clotting time, which could be partly rescued by excess phospholipids, indicating POmAb is an anticoagulant but exerts a weak lupus anticoagulant effect. These studies reveal the structural basis of prothrombin recognition by a type I antiphospholipid antibody and uncover an exciting new strategy to achieve anticoagulation in human plasma.


Subject(s)
Antibodies, Antiphospholipid , Cryoelectron Microscopy , Prothrombin , Humans , Antibodies, Antiphospholipid/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Blood Coagulation , Epitopes/immunology , Kringles , Protein Binding , Prothrombin/chemistry , Prothrombin/immunology , Prothrombin/metabolism
4.
J Nanobiotechnology ; 21(1): 419, 2023 Nov 13.
Article in English | MEDLINE | ID: mdl-37957714

ABSTRACT

Exosomes originating from human umbilical cord mesenchymal stem cells (hucMSC-exos) have become a novel strategy for treating various diseases owing to their ability to regulate intercellular signal communication. However, the potential of hucMSC-exos to improve placental injury in obstetric antiphospholipid syndrome and its underlying mechanism remain unclear. Our objective was to explore the potential application of hucMSC-exos in the treatment of obstetric antiphospholipid syndrome and elucidate its underlying mechanism. In our study, hucMSC-exos ameliorated the functional impairment of trophoblasts caused by antiphospholipid antibodies in vitro and attenuated placental dysfunction in mice with obstetric antiphospholipid syndrome by delivering miR-146a-5p. Exosomal miR-146a-5p suppressed the expression of tumor necrosis factor receptor-associated factor 6 (TRAF6) and inhibited the activation of NF-κB signaling, leading to the down-regulation of IL-1ß and IL-18 to rescue inflammation and modulation of Cleaved-CASP3, BAX, and BCL2 to inhibit apoptosis in HTR8/SVneo cells and mice placenta. This study identified the potential molecular basis of how hucMSC-exos improved antiphospholipid antibody-induced placental injury and highlighted the functional importance of the miR-146a-5p/TRAF6 axis in the progression of obstetric antiphospholipid syndrome. More importantly, this study provided a fresh outlook on the promising use of hucMSC-exos as a novel and effective treatment approach in obstetric antiphospholipid syndrome.


Subject(s)
Antiphospholipid Syndrome , Mesenchymal Stem Cells , MicroRNAs , Animals , Female , Humans , Mice , Pregnancy , Antibodies, Antiphospholipid/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Placenta/metabolism , TNF Receptor-Associated Factor 6/metabolism , Trophoblasts , Umbilical Cord
5.
J Clin Invest ; 132(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35289310

ABSTRACT

A close association with its vertebrate and tick hosts allows Borrelia burgdorferi, the bacterium responsible for Lyme disease, to eliminate many metabolic pathways and instead scavenge key nutrients from the host. A lipid-defined culture medium was developed to demonstrate that exogenous lipids are an essential nutrient of B. burgdorferi, which can accumulate intact phospholipids from its environment to support growth. Antibody responses to host phospholipids were studied in mice and humans using an antiphospholipid ELISA. Several of these environmentally acquired phospholipids including phosphatidylserine and phosphatidic acid, as well as borrelial phosphatidylcholine, are the targets of antibodies that arose early in infection in the mouse model. Patients with acute infections demonstrated antibody responses to the same lipids. The elevation of antiphospholipid antibodies predicted early infection with better sensitivity than did the standardized 2-tier tests currently used in diagnosis. Sera obtained from patients with Lyme disease before and after antibiotic therapy showed declining antiphospholipid titers after treatment. Further study will be required to determine whether these antibodies have utility in early diagnosis of Lyme disease, tracking of the response to therapy, and diagnosis of reinfection, areas in which current standardized tests are inadequate.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Animals , Antibodies, Antiphospholipid/metabolism , Antibodies, Bacterial , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Phospholipids/metabolism
6.
Arthritis Rheumatol ; 74(6): 1001-1012, 2022 06.
Article in English | MEDLINE | ID: mdl-35044724

ABSTRACT

OBJECTIVE: Miscarriage affects 1 in 7 pregnancies, and antiphospholipid autoantibodies (aPLs) are one of the biggest risk factors for recurrent pregnancy loss. While aPLs target the endometrial stroma, little is known about their impact. Endometrial stromal cells (EnSCs) undergo decidualization each menstrual cycle, priming the uterus to receive implanting embryos. Thus, appropriate decidualization and EnSC function is key for establishment of a successful pregnancy. This study was undertaken to explore the effects of aPL on EnSC decidualization, senescence, and inflammation. METHODS: EnSCs under decidualizing conditions were exposed to aPL or control IgG alone or in the presence of either a Toll-like receptor 4 (TLR-4) antagonist, a p38 MAPK inhibitor, a reactive oxygen species (ROS) inhibitor, low molecular weight heparin (LMWH), or acetyl salicylic acid. Secretion of decidualization markers and inflammatory interleukin-8 were quantified by enzyme-linked immunosorbent assay, and senescence-associated ß-galactosidase activity was evaluated. In a mouse model of decidualization, aPL or control IgG was administered, and uterine expression levels of decidualization and inflammatory markers were quantified by real-time quantitative polymerase chain reaction. RESULTS: Antiphospholipid antibodies increased human EnSC decidualization, senescence, and inflammation. This phenotype was recapitulated in the mouse model. The decidualization and inflammatory responses were partially mediated by TLR-4 and p38 MAPK, while the decidualization and senescence responses were ROS-dependent. LMWH, commonly used to treat aPL-positive women at risk of obstetric complications, reduced the ability of aPL to increase EnSC decidualization and inflammation. CONCLUSION: These findings shed new light on the pathogenesis of pregnancy complications in women with aPLs and underscore the benefit of heparin in preventing pregnancy loss in this high-risk population.


Subject(s)
Antibodies, Antiphospholipid , MAP Kinase Signaling System , Reactive Oxygen Species , Stromal Cells , Toll-Like Receptor 4 , p38 Mitogen-Activated Protein Kinases , Animals , Antibodies, Antiphospholipid/metabolism , Endometrium/metabolism , Female , Heparin, Low-Molecular-Weight/pharmacology , Immunoglobulin G/metabolism , Inflammation/metabolism , Mice , Pregnancy , Reactive Oxygen Species/metabolism , Stromal Cells/metabolism , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Int J Mol Sci ; 22(8)2021 Apr 18.
Article in English | MEDLINE | ID: mdl-33919627

ABSTRACT

The antiphospholipid syndrome (APS) is characterized by thrombosis and/or pregnancy morbidity with the persistent presence of antiphospholipid antibodies (aPLs). Laboratory criteria for the classification of APS include the detection of lupus anticoagulant (LAC), anti-cardiolipin (aCL) antibodies and anti-ß2glycoprotein I (aß2GPI) antibodies. Clinical criteria for the classification of thrombotic APS include venous and arterial thrombosis, along with microvascular thrombosis. Several aPLs, including LAC, aß2GPI and anti-phosphatidylserine/prothrombin antibodies (aPS/PT) have been associated with arterial thrombosis. The Von Willebrand Factor (VWF) plays an important role in arterial thrombosis by mediating platelet adhesion and aggregation. Studies have shown that aPLs antibodies present in APS patients are able to increase the risk of arterial thrombosis by upregulating the plasma levels of active VWF and by promoting platelet activation. Inflammatory reactions induced by APS may also provide a suitable condition for arterial thrombosis, mostly ischemic stroke and myocardial infarction. The presence of other cardiovascular risk factors can enhance the effect of aPLs and increase the risk for thrombosis even more. These factors should therefore be taken into account when investigating APS-related arterial thrombosis. Nevertheless, the exact mechanism by which aPLs can cause thrombosis remains to be elucidated.


Subject(s)
Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/metabolism , Blood Platelets/metabolism , Thrombosis/metabolism , von Willebrand Factor/metabolism , Animals , Antibodies, Antiphospholipid/immunology , Antibodies, Antiphospholipid/metabolism , Female , Humans , Male , Thrombosis/immunology
9.
Clin Appl Thromb Hemost ; 27: 10760296211002914, 2021.
Article in English | MEDLINE | ID: mdl-33784835

ABSTRACT

The relationship between antiphospholipid antibodies (aPL) and sickle cell disease (SCD) has never been systematically addressed. Our aim was to evaluate potential links between SCD and aPL in all age groups. EMBASE/PubMed was screened from inception to May 2020 and Peto odds ratios for rare events were calculated. The pooled prevalence (PP) of IgG anticardiolipin antibodies (aCL) was higher in individuals with SCD than in controls (27.9% vs 8.7%, P < 0.0001), that of IgM aCL was similar in the two groups (2.9% vs 2.7%); only individuals with SCD were positive for lupus anticoagulant (LA) (7.7% vs 0%, P < 0.0001). The PP of leg ulcers was similar between aPL positive and negative individuals (44% vs 53%) and between patients in acute crisis and stable patients (5.6% vs 7.3%). Reporting of aPL as a binary outcome and not as a titer precluded further interpretation. The results indicate that a prospective case-control study with serial measurements of a panel of aPL in SCD patients might be warranted, in order to understand further the possible pathogenic role of aPL in SCD.


Subject(s)
Anemia, Sickle Cell/blood , Antibodies, Antiphospholipid/metabolism , Female , Humans , Male
10.
Sci China Life Sci ; 64(10): 1702-1719, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33481164

ABSTRACT

Kidney is a major target organ in both antiphospholipid syndrome (APS) and systemic lupus erythematosus (SLE). The etiology of antiphospholipid syndrome nephropathy associated lupus nephritis (APSN-LN) is intricate and remains largely unrevealed. We proposed in present work, that generation of antiphospholipid antibodies (aPLs), especially those directed towards the oxidized neoepitopes, are largely linked with the redox status along with disease progression. Moreover, we observed that compromised antioxidative capacity coincided with turbulence of inflammatory cytokine profile in the kidney of male NZW×BXSB F1 mice suffered from APSN-LN. SM934 is an artemisinin derivative that has been proved to have potent immunosuppressive properties. In current study, we elaborated the therapeutic benefits of SM934 in male NZW×BXSB F1 mice, a murine model develops syndrome resembled human APS associated with SLE, for the first time. SM934 treatment comprehensively impeded autoantibodies production, inflammatory cytokine accumulation and excessive oxidative stress in kidney. Among others, we interpreted in present work that both anti-inflammatory and antioxidative effects of SM934 is closely correlated with the enhancement of Nrf2 signaling and expression of its targets. Collectively, our finding confirmed that therapeutic strategy simultaneously exerting antioxidant and anti-inflammatory efficacy provide a novel feasible remedy for treating APSN-LN.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antiphospholipid Syndrome/drug therapy , Artemisinins/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , NF-E2-Related Factor 2/metabolism , Animals , Antibodies, Antiphospholipid/metabolism , Antiphospholipid Syndrome/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation , Kidney/drug effects , Kidney/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Nephritis/metabolism , Lupus Nephritis/prevention & control , Male , Mice , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
11.
Am J Reprod Immunol ; 85(2): e13312, 2021 02.
Article in English | MEDLINE | ID: mdl-32715546

ABSTRACT

Antiphospholipid antibodies (aPL) are autoantibodies that target phospholipid-binding proteins, such as ß2 glycoprotein I (ß2GPI), and can induce thrombosis systemically, as well as increase the risk of obstetric complications such as recurrent miscarriage and preeclampsia. Due to the expression of ß2GPI by placental trophoblasts, aPL readily target the maternal-fetal interface during pregnancy and many studies have investigated the deleterious effects of aPL on placental trophoblast function. This review will focus on studies that have examined the effects of aPL on the production and modification of extracellular vesicles (EVs) from trophoblasts, as EVs are a key mode of feto-maternal communication in both normal and pathological pregnancy. A more comprehensive understanding of the effects of aPL on the quantity and cargo of EVs extruded by the human placenta may contribute to our current knowledge of how aPL induce both systemic and obstetric disease.


Subject(s)
Antibodies, Antiphospholipid/metabolism , Extracellular Vesicles/metabolism , Pre-Eclampsia/immunology , Pregnancy/immunology , Trophoblasts/immunology , Animals , Female , Humans , beta 2-Glycoprotein I/immunology
12.
Curr Rheumatol Rev ; 17(1): 7-16, 2021.
Article in English | MEDLINE | ID: mdl-33019935

ABSTRACT

Viruses can induce autoimmune diseases, in addition to genetic predisposition and environmental factors. Particularly, coronaviruses are mentioned among the viruses implicated in autoimmunity. Today, the world's greatest threat derives from the pandemic of a new human coronavirus, called "severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the responsible agent of coronavirus disease 2019 (COVID-19). First case of COVID-19 was identified in Wuhan, the capital of Hubei, China, in December 2019 and quickly spread around the world. This review focuses on autoimmune manifestations described during COVID-19, including pro-thrombotic state associated with antiphospholipid antibodies (aPL), acute interstitial pneumonia, macrophage activation syndrome, lymphocytopenia, systemic vasculitis, and autoimmune skin lesions. This offers the opportunity to highlight the pathogenetic mechanisms common to COVID-19 and several autoimmune diseases in order to identify new therapeutic targets. In a supposed preliminary pathogenetic model, SARS-CoV-2 plays a direct role in triggering widespread microthrombosis and microvascular inflammation, because it is able to induce transient aPL, endothelial damage and complement activation at the same time. Hence, endothelium might represent the common pathway in which autoimmunity and infection converge. In addition, autoimmune phenomena in COVID-19 can be explained by regulatory T cells impairment and cytokines cascade.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/physiology , COVID-19/immunology , Animals , Antibodies, Antiphospholipid/immunology , Antibodies, Antiphospholipid/metabolism , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , COVID-19/complications , COVID-19/metabolism , Cytokines/immunology , Cytokines/metabolism , Humans , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Macrophage Activation/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
13.
Int J Mol Sci ; 21(21)2020 Nov 09.
Article in English | MEDLINE | ID: mdl-33182499

ABSTRACT

Antiphospholipid Syndrome (APS) is an autoimmune disease characterized by arterial and/or venous thrombosis and/or pregnancy morbidity, associated with circulating antiphospholipid antibodies (aPL). In some cases, patients with a clinical profile indicative of APS (thrombosis, recurrent miscarriages or fetal loss), who are persistently negative for conventional laboratory diagnostic criteria, are classified as "seronegative" APS patients (SN-APS). Several findings suggest that aPL, which target phospholipids and/or phospholipid binding proteins, mainly ß-glycoprotein I (ß-GPI), may contribute to thrombotic diathesis by interfering with hemostasis. Despite the strong association between aPL and thrombosis, the exact pathogenic mechanisms underlying thrombotic events and pregnancy morbidity in APS have not yet been fully elucidated and multiple mechanisms may be involved. Furthermore, in many SN-APS patients, it is possible to demonstrate the presence of unconventional aPL ("non-criteria" aPL) or to detect aPL with alternative laboratory methods. These findings allowed the scientists to study the pathogenic mechanism of SN-APS. This review is focused on the evidence showing that these antibodies may play a functional role in the signal transduction pathway(s) leading to thrombosis and pregnancy morbidity in SN-APS. A better comprehension of the molecular mechanisms triggered by aPL may drive development of potential therapeutic strategies in APS patients.


Subject(s)
Antibodies, Antiphospholipid/metabolism , Antiphospholipid Syndrome/metabolism , Antiphospholipid Syndrome/pathology , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Female , Humans , Pregnancy , Signal Transduction/radiation effects , Thrombosis/metabolism , Thrombosis/pathology
15.
J Autoimmun ; 115: 102524, 2020 12.
Article in English | MEDLINE | ID: mdl-32693965

ABSTRACT

Antiphospholipid (aPL) autoantibodies are uncommon in systemic autoimmune diseases (SADs). However, the European PRECISESADS study provides the opportunity to better characterize this rare association. The study was composed of 1818 patients with SADs including 453 with systemic lupus erythematosus (SLE), 359 with rheumatoid arthritis (RA), 385 with systemic sclerosis (SSc), 367 with Sjögren's syndrome (SjS), 94 with mixed connective tissue disease (MCTD), and 160 with undifferentiated connective tissue disease (UCTD). Assays used for aPL determination include the lupus anticoagulant (LAC) analysis using the dilute Russell's viper venom time (dRVVT) assay plus anti-cardiolipin (aCL) and anti-aß2GPI autoantibodies of IgG and IgM isotype. Information regarding clinical and biological characteristics of SAD patients was available. Among SAD patients, the prevalence of aPL differs significantly between two groups: SLE (57.6%) and non-SLE SADs (13.7%, p < 10-4). Next, association between aPL plus thrombosis and miscarriage were observed in both SLE and non-SLE patients. Thrombosis was best predicted in SLE patients by dRVVT (OR = 6.1; IC95:3.5-10.3) and miscarriage by aCL±ß2GPI IgG (OR = 2.5; IC95:1.2-5.2); while in non-SLE SADs the best predictors were aCL±ß2GPI IgG for thrombosis (OR = 6.6; IC95:2.4-18.4) and aCL±ß2GPI IgM for miscarriage (OR = 2.9; IC95:1.2-6.8). In the case of multiple positivity of aPL, the risk for thrombosis and miscarriage was increased. Central nervous system involvement characterized the SLE patients, in contrast to pulmonary and skin fibrosis, valve lesions, hypertension, elevated creatinemia, C4 fraction reduction, platelet reduction and inflammation that characterized the non-SLE SAD patients. Anti-PL determination remains important in SADs patients and should not be restricted to only SLE patients.


Subject(s)
Abortion, Spontaneous/epidemiology , Antibodies, Antiphospholipid/blood , Autoimmune Diseases/complications , Thrombosis/epidemiology , Abortion, Spontaneous/immunology , Adult , Aged , Antibodies, Antiphospholipid/immunology , Antibodies, Antiphospholipid/metabolism , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Complement Activation , Europe/epidemiology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pregnancy , Risk Assessment/methods , Thrombosis/immunology
16.
Clin Appl Thromb Hemost ; 26: 1076029620928392, 2020.
Article in English | MEDLINE | ID: mdl-32633133

ABSTRACT

Given that the presence of antiphospholipid (aPL) antibodies has been proposed to be associated with thrombosis in newly diagnosed patients with lymphoma, we conducted a prospective cohort study on these patients. In all, 154 patients were enrolled. More than half were advanced-stage diffuse large B-cell lymphoma. Approximately one-third (35.7%) of the patients had the presence of aPLs, with single-, double-, and triple-aPL positivities of 29.9%, 5.2%, and 0.6%, respectively. Of the 154 patients, 8 (5.19%) developed symptomatic thrombosis during follow-up. There were no significant differences in the incidences of thrombosis for the aPL-positive and aPL-negative groups (5.5% vs 5.1%; P = 1.000). In a multivariate analysis, patients with male sex and lymphoma stage IV were significant risk factors for aPL positivity, with odds ratio [OR] = 2.22 (95% CI: 1.11-4.45), P = .025, and OR: 2.34 (95% CI: 1.17-4.67), P = .016, respectively. An aPL predictive score of ≥-1 was predictive of aPL positivity, with a sensitivity of 83.6% and specificity of 34.3%.


Subject(s)
Antibodies, Antiphospholipid/metabolism , Genetic Testing/methods , Lymphoma/blood , Female , Humans , Incidence , Male , Middle Aged , Risk Factors
17.
Hemodial Int ; 24(3): 383-396, 2020 07.
Article in English | MEDLINE | ID: mdl-32524729

ABSTRACT

INTRODUCTION: The relationship between autoimmune hemolytic anemia and antiphospholipid antibodies (aPL) and/or antiphospholipid syndrome has never been systematically addressed. METHODS: Systematic review of EMBASE and PubMed databases performed according to PRISMA guidelines from inception to March 2020; meta-analysis performed by Peto's odds ratio for rare events. FINDINGS: Forty-five studies with different outcomes met the inclusion/exclusion criteria. The pooled prevalence (PP) of IgG anticardiolipin antibodies (aCL) positivity was greater in end-stage renal disease (ESRD) than controls (20.2% vs. 2.6%, P = 0.001, I2 >80%; I2 = heterogeneity), particularly in hemodialysis patients (18.3% vs. 8%, I2 = 0%). The PP of lupus anticoagulant was greater in ESRD than controls (8.7% vs. 0.2%, P < 0.0001, I2 = 0%). The standardized mean difference of IgG aCL favored ESRD rather than controls (P < 0.0001, I2 =97%). The PP of fistula occlusion was greater in IgG aCL-positive patients than negative patients (39% vs. 27%, I2 =97%); the PP of IgG aCL positivity was greater in patients with fistula occlusion than without fistula occlusion (26.9% vs. 23.2%, P = 0.01, I2 =72%); the same applied to the PP of lupus anticoagulant positivity (23% vs. 0.3%, P < 0.0001, I2 = 0%). The standardized mean difference of IgG aCL favored fistula occlusion (P = 0.004, I2 = 91%). DISCUSSION: Lupus anticoagulant relates to ESRD regardless of management whereas IgG aCL relates specifically to ESRD on hemodialysis, but only lupus anticoagulant associates with fistula occlusion. The expression of aPL as patients positive for aPL rather than as titers precludes further assumptions on the relationship.


Subject(s)
Antibodies, Antiphospholipid/metabolism , Kidney Failure, Chronic/complications , Renal Dialysis/methods , Humans
18.
J Biol Chem ; 295(31): 10794-10806, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32518155

ABSTRACT

ß2-Glycoprotein I (ß2GPI) is an abundant plasma protein displaying phospholipid-binding properties. Because it binds phospholipids, it is a target of antiphospholipid antibodies (aPLs) in antiphospholipid syndrome (APS), a life-threatening autoimmune thrombotic disease. Indeed, aPLs prefer membrane-bound ß2GPI to that in solution. ß2GPI exists in two almost equally populated redox states: oxidized, in which all the disulfide bonds are formed, and reduced, in which one or more disulfide bonds are broken. Furthermore, ß2GPI can adopt multiple conformations (i.e. J-elongated, S-twisted, and O-circular). While strong evidence indicates that the J-form is the structure bound to aPLs, which conformation exists and predominates in solution remains controversial, and so is the conformational pathway leading to the bound state. Here, we report that human recombinant ß2GPI purified under native conditions is oxidized. Moreover, under physiological pH and salt concentrations, this oxidized form adopts a J-elongated, flexible conformation, not circular or twisted, in which the N-terminal domain I (DI) and the C-terminal domain V (DV) are exposed to the solvent. Consistent with this model, binding kinetics and mutagenesis experiments revealed that in solution the J-form interacts with negatively charged liposomes and with MBB2, a monoclonal anti-DI antibody that recapitulates most of the features of pathogenic aPLs. We conclude that the preferential binding of aPLs to phospholipid-bound ß2GPI arises from the ability of its preexisting J-form to accumulate on the membranes, thereby offering an ideal environment for aPL binding. We propose that targeting the J-form of ß2GPI provides a strategy to block pathogenic aPLs in APS.


Subject(s)
Antibodies, Antiphospholipid/chemistry , Antiphospholipid Syndrome , beta 2-Glycoprotein I/chemistry , Animals , Antibodies, Antiphospholipid/metabolism , Cricetinae , HEK293 Cells , Humans , Kinetics , Mutagenesis , Protein Domains , beta 2-Glycoprotein I/metabolism
19.
Clin Rheumatol ; 39(7): 2111-2114, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32458245

ABSTRACT

Antiphospholipid syndrome (APS) is an autoimmune disease characterised by vascular thrombosis and/or pregnancy morbidity in the presence of persistently positive serum tests for antiphospholipid antibodies. Management of APS centres on preventing these clinical events and in preventing chronic damage caused by these events. In patients with thrombotic APS, long-term anticoagulation is recommended in the majority of cases. Although there were hopes that direct-acting oral anticoagulants could replace warfarin for prevention of thrombosis in patients with APS, this now seems less likely due to recent trial results. There is no evidence for use of anticoagulation in people who are aPL-positive but have never had a thrombosis but low-dose aspirin may be beneficial in those who have a higher-risk aPL profile. Management of obstetric APS is with daily subcutaneous heparin and low-dose aspirin. This gives a live birth rate of 70% or more. Catastrophic APS is rare, occurring in 1% of patients with APS. It is characterised by thrombosis in multiple organs simultaneously, with a high mortality rate. The management is with corticosteroids, anticoagulation, and immunoglobulins or plasma exchange.


Subject(s)
Anticoagulants/therapeutic use , Antiphospholipid Syndrome/therapy , Heparin, Low-Molecular-Weight/therapeutic use , Immunoglobulins, Intravenous/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Pregnancy Complications/therapy , Antibodies, Antiphospholipid/metabolism , Antiphospholipid Syndrome/immunology , Autoimmunity , Disease Management , Drug Therapy, Combination , Female , Humans , Pregnancy , Pregnancy Complications/immunology , Thrombosis
20.
Chin Med J (Engl) ; 133(8): 929-940, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32265421

ABSTRACT

Antiphospholipid syndrome (APS) is a thromboinflammatory disease with a variety of clinical phenotypes. Primary thrombosis prophylaxis should take an individualized risk stratification approach. Moderate-intensity vitamin K antagonist such as warfarin remains the primary strategy for secondary thrombosis prophylaxis among APS patients, especially for patients with predominantly venous disease. For now, direct oral anti-coagulants should be avoided in most APS patients, especially those with history of arterial manifestations. Obstetric APS management should be tailored based on an individual patient's antiphospholipid antibody profile, and obstetric and thrombotic history. Pharmacological agents beyond anticoagulants may be considered for the management of microthrombotic and nonthrombotic manifestations of APS, although more data are needed. A relatively recent discovery in the area of APS pathogenesis is the implication of neutrophil extracellular traps in thrombin generation and initiation of inflammatory cascades. APS is a complex thromboinflammatory disease with a broad clinical spectrum. Personalized therapy according to an individual's unique thrombosis and obstetric risk should be advocated.


Subject(s)
Antiphospholipid Syndrome/drug therapy , Antiphospholipid Syndrome/metabolism , Antibodies, Antiphospholipid/metabolism , Anticoagulants/therapeutic use , Extracellular Traps/metabolism , Humans , Thrombin/metabolism , Warfarin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL