1
|
Müller-Calleja N, Ruf W, Lackner KJ. Lipid-binding antiphospholipid antibodies: significance for pathophysiology and diagnosis of the antiphospholipid syndrome. Crit Rev Clin Lab Sci 2024; 61:370-387. [PMID: 38293818 DOI: 10.1080/10408363.2024.2305121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/13/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
The antiphospholipid syndrome (APS) is an autoimmune disease characterized by the presence of pathogenic antiphospholipid antibodies (aPL). Since approximately 30 years ago, lipid-binding aPL, which do not require a protein cofactor, have been regarded as irrelevant for APS pathogenesis even though anticardiolipin are a diagnostic criterion of APS. In this review, we will summarize the available evidence from in vitro studies, animal models, and epidemiologic studies, which suggest that this concept is no longer tenable. Accordingly, we will only briefly touch on the role of other aPL in APS. This topic has been amply reviewed in detail elsewhere. We will discuss the consequences for laboratory diagnostics and future research required to resolve open questions related to the pathogenic role of different aPL specificities.
Collapse
Affiliation(s)
- Nadine Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
2
|
Sun X, Pan H, Lu H, Song S, Jin C, Pu Y. Diagnostic significance of combined anti-extractable nuclear antigens antibody, anti-cardiolipin antibody and anti-β2-glycoprotein 1 in systemic lupus erythematosus patients. Heliyon 2024; 10:e29230. [PMID: 38617903 PMCID: PMC11015451 DOI: 10.1016/j.heliyon.2024.e29230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024] Open
Abstract
Objective This study aimed to investigate the diagnostic value of a combination of anti-extractable nuclear antigens (anti-ENA) antibodies, anti-cardiolipin antibodies (ACA), and anti-β2-glycoprotein 1 (anti-β2 GPI) antibodies in patients with systemic lupus erythematosus (SLE). Methods A total of 646 SLE patients diagnosed in our hospital between January 2020 and April 2023 were randomly selected as study subjects, while 2075 non-SLE subjects during the same period were selected as the control group. The levels of anti-extractable nuclear antigen (ENA) antibody, ACA, and anti-β2 GPI antibodies were measured, and their diagnostic value for SLE was analyzed using binary logistic regression and receiver operating characteristic (ROC) analysis. Results The rates of positive anti-RNP, anti-Sm, anti-Sjögren's syndrome (SS-A), and anti-SS-B antibodies in the SLE patient group were significantly higher than those in the control group, with all differences being statistically significant (P < 0.01). The rates of positive ACA, as well as the levels of ACA IgA, ACA IgG, and ACA IgM, were significantly higher in the SLE patients group compared to the control group, with statistically differences (P < 0.05). Similarly, the rates of positive anti-β2 GPI antibodies, anti-β2 GPI antibody IgA, IgG, and IgM, were significantly higher in the SLE patient group compared to the control group, with all differences being statistically significant (P < 0.01). Gender, age, positive anti-JO1 antibodies, positive anti-RNP antibodies, positive anti-SS-A antibodies, positive ACA, high level ACA IgG and ACA IgM, positive anti-β2 GPI antibodies, and high level of anti-β2 GPI antibody IgA were identified as independent risk factors for the development of SLE (P < 0.05). The combined use of age, sex, anti-ENA antibodies, ACA, and anti-β2 GPI antibodies yielded a sensitivity of 82.12% (80.41%-83.71%) and a specificity of 80.03% (76.77%-82.93%) for the diagnosis of SLE. Conclusion The combination of age, sex, anti-ENA antibodies, ACA, and anti-β2 GPI antibodies shows high diagnostic value for SLE and holds potential for clinical application as an auxiliary diagnostic tool for SLE.
Collapse
Affiliation(s)
| | | | | | - Shanshan Song
- Department of Clinical Laboratory, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chengjun Jin
- Department of Clinical Laboratory, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yingye Pu
- Department of Clinical Laboratory, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
3
|
Ma H, Huang Y, Tian W, Liu J, Yan X, Ma L, Lai J. Endothelial transferrin receptor 1 contributes to thrombogenesis through cascade ferroptosis. Redox Biol 2024; 70:103041. [PMID: 38241836 PMCID: PMC10831316 DOI: 10.1016/j.redox.2024.103041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
Oxidative stress and iron accumulation-induced ferroptosis occurs in injured vascular cells and can promote thrombogenesis. Transferrin receptor 1 (encoded by the TFRC gene) is an initial element involved in iron transport and ferroptosis and is highly expressed in injured vascular tissues, but its role in thrombosis has not been determined. To explore the potential mechanism and therapeutic effect of TFRC on thrombogenesis, a DVT model of femoral veins (FVs) was established in rats, and weighted correlation network analysis (WGCNA) was used to identify TFRC as a hub protein that is associated with thrombus formation. TFRC was knocked down by adeno-associated virus (AAV) or lentivirus transduction in FVs or human umbilical vein endothelial cells (HUVECs), respectively. Thrombus characteristics and ferroptosis biomarkers were evaluated. Colocalization analysis, molecular docking and coimmunoprecipitation (co-IP) were used to evaluate protein interactions. Tissue-specific TFRC knockdown alleviated iron overload and redox stress, thereby preventing ferroptosis in injured FVs. Loss of TFRC in injured veins could alleviate thrombogenesis, reduce thrombus size and attenuate hypercoagulability. The protein level of thrombospondin-1 (THBS1) was increased in DVT tissues, and silencing TFRC decreased the protein level of THBS1. In vitro experiments further showed that TFRC and THBS1 were sensitive to erastin-induced ferroptosis and that TFRC knockdown reversed this effect. TFRC can interact with THBS1 in the domain spanning from TSR1-2 to TSR1-3 of THBS1. Amino acid sites, including GLN320 of TFRC and ASP502 of THBS1, could be potential pharmacological targets. Erastin induced ferroptosis affected extracellular THBS1 levels and weakened the interaction between TFRC and THBS1 both in vivo and in vitro, and promoted the interaction between THBS1 and CD47. This study revealed a linked relationship between venous ferroptosis and coagulation cascades. Controlling TFRC and ferroptosis in endothelial cells can be an efficient approach for preventing and treating thrombogenesis.
Collapse
Affiliation(s)
- Haotian Ma
- NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, China; Institute of Forensic Injury, Bio-evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| | - Yongtao Huang
- Department of Orthopedics, Ruihua Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenrong Tian
- NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, China; Institute of Forensic Injury, Bio-evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| | - Jincen Liu
- NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, China; Institute of Forensic Injury, Bio-evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| | - Xinyue Yan
- NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, China; Institute of Forensic Injury, Bio-evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| | - Lei Ma
- NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, China; Institute of Forensic Injury, Bio-evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| | - Jianghua Lai
- NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, China; Institute of Forensic Injury, Bio-evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
4
|
Müller-Calleja N, Grunz K, Nguyen TS, Posma J, Pedrosa D, Meineck M, Hollerbach A, Braun J, Muth S, Schild H, Saar K, Hübner N, Krishnaswamy S, Royce J, Teyton L, Lemmermann N, Weinmann-Menke J, Lackner KJ, Ruf W. Targeting the tissue factor coagulation initiation complex prevents antiphospholipid antibody development. Blood 2024; 143:1167-1180. [PMID: 38142429 PMCID: PMC10972716 DOI: 10.1182/blood.2023022276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023] Open
Abstract
ABSTRACT Antiphospholipid antibodies (aPL) in primary or secondary antiphospholipid syndrome (APS) are a major cause for acquired thrombophilia, but specific interventions preventing autoimmune aPL development are an unmet clinical need. Although autoimmune aPL cross react with various coagulation regulatory proteins, lipid-reactive aPL, including those derived from patients with COVID-19, recognize the endolysosomal phospholipid lysobisphosphatidic acid presented by the cell surface-expressed endothelial protein C receptor. This specific recognition leads to complement-mediated activation of tissue factor (TF)-dependent proinflammatory signaling and thrombosis. Here, we show that specific inhibition of the TF coagulation initiation complex with nematode anticoagulant protein c2 (NAPc2) prevents the prothrombotic effects of aPL derived from patients with COVID-19 in mice and the aPL-induced proinflammatory and prothrombotic activation of monocytes. The induction of experimental APS is dependent on the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex, and NAPc2 suppresses monocyte endosomal reactive oxygen species production requiring the TF cytoplasmic domain and interferon-α secretion from dendritic cells. Latent infection with murine cytomegalovirus causes TF cytoplasmic domain-dependent development of persistent aPL and circulating phospholipid-reactive B1 cells, which is prevented by short-term intervention with NAPc2 during acute viral infection. In addition, treatment of lupus prone MRL-lpr mice with NAPc2, but not with heparin, suppresses dendritic-cell activation in the spleen, aPL production and circulating phospholipid-reactive B1 cells, and attenuates lupus pathology. These data demonstrate a convergent TF-dependent mechanism of aPL development in latent viral infection and autoimmune disease and provide initial evidence that specific targeting of the TF initiation complex has therapeutic benefits beyond currently used clinical anticoagulant strategies.
Collapse
Affiliation(s)
- Nadine Müller-Calleja
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Kristin Grunz
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - T. Son Nguyen
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Jens Posma
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Denise Pedrosa
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Myriam Meineck
- Department of Medicine I, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Anne Hollerbach
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Johannes Braun
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Sabine Muth
- Institute for Immunology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Hansjörg Schild
- Institute for Immunology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Kathrin Saar
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Sriram Krishnaswamy
- Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA
| | - Jennifer Royce
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
| | - Luc Teyton
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
| | - Niels Lemmermann
- Institute for Virology, Johannes Gutenberg University Medical Center, Mainz, Germany
- Institute of Virology, University Hospital Bonn, Bonn, Germany
| | - Julia Weinmann-Menke
- Department of Medicine I, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Karl J. Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Mainz, Germany
| |
Collapse
|
5
|
Willis R, McDonnell TCR, Pericleous C, Gonzalez EB, Schleh A, Romay-Penabad Z, Giles IP, Rahman A. PEGylated Domain I of Beta-2-Glycoprotein I Inhibits Thrombosis in a Chronic Mouse Model of the Antiphospholipid Syndrome. Front Immunol 2022; 13:842923. [PMID: 35479096 PMCID: PMC9035797 DOI: 10.3389/fimmu.2022.842923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disorder in which autoantibodies cause clinical effects of vascular thrombosis and pregnancy morbidity. The only evidence-based treatments are anticoagulant medications such as warfarin and heparin. These medications have a number of disadvantages, notably risk of haemorrhage. Therefore, there is a pressing need to develop new, more focused treatments that target the actual pathogenic disease process in APS. The pathogenic antibodies exert their effects by interacting with phospholipid-binding proteins, of which the most important is beta-2-glycoprotein I. This protein has five domains, of which the N-terminal Domain I (DI) is the main site for binding of pathogenic autoantibodies. We previously demonstrated bacterial expression of human DI and showed that this product could inhibit the ability of IgG from patients with APS (APS-IgG) to promote thrombosis in a mouse model. Since DI is a small 7kDa protein, its serum half-life would be too short to be therapeutically useful. We therefore used site-specific chemical addition of polyethylene glycol (PEG) to produce a larger variant of DI (PEG-DI) and showed that PEG-DI was equally effective as the non-PEGylated DI in inhibiting thrombosis caused by passive transfer of APS-IgG in mice. In this paper, we have used a mouse model that reflects human APS much more closely than the passive transfer of APS-IgG. In this model, the mice are immunized with human beta-2-glycoprotein I and develop endogenous anti-beta-2-glycoprotein I antibodies. When submitted to a pinch stimulus at the femoral vein, these mice develop clots. Our results show that PEG-DI inhibits production of thromboses in this model and also reduces expression of tissue factor in the aortas of the mice. No toxicity was seen in mice that received PEG-DI. Therefore, these results provide further evidence supporting possible efficacy of PEG-DI as a potential treatment for APS.
Collapse
Affiliation(s)
- Rohan Willis
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Thomas C. R. McDonnell
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - Charis Pericleous
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - Emilio B. Gonzalez
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Alvaro Schleh
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Zurina Romay-Penabad
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Ian P. Giles
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - Anisur Rahman
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
6
|
Bray MA, Sartain SE, Gollamudi J, Rumbaut RE. Microvascular thrombosis: experimental and clinical implications. Transl Res 2020; 225:105-130. [PMID: 32454092 PMCID: PMC7245314 DOI: 10.1016/j.trsl.2020.05.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 02/07/2023]
Abstract
A significant amount of clinical and research interest in thrombosis is focused on large vessels (eg, stroke, myocardial infarction, deep venous thrombosis, etc.); however, thrombosis is often present in the microcirculation in a variety of significant human diseases, such as disseminated intravascular coagulation, thrombotic microangiopathy, sickle cell disease, and others. Further, microvascular thrombosis has recently been demonstrated in patients with COVID-19, and has been proposed to mediate the pathogenesis of organ injury in this disease. In many of these conditions, microvascular thrombosis is accompanied by inflammation, an association referred to as thromboinflammation. In this review, we discuss endogenous regulatory mechanisms that prevent thrombosis in the microcirculation, experimental approaches to induce microvascular thrombi, and clinical conditions associated with microvascular thrombosis. A greater understanding of the links between inflammation and thrombosis in the microcirculation is anticipated to provide optimal therapeutic targets for patients with diseases accompanied by microvascular thrombosis.
Collapse
Key Words
- adamts13, a disintegrin-like and metalloproteinase with thrombospondin type 1 motif 13
- ap, alternate pathway
- apc, activated protein c
- aps, antiphospholipid syndrome
- caps, catastrophic aps
- asfa, american society for apheresis
- atp, adenosine triphosphate
- cfh, complement factor h
- con a, concavalin a
- cox, cyclooxygenase
- damp, damage-associated molecular pattern
- dic, disseminated intravascular coagulation
- gbm, glomerular basement membrane
- hellp, hemolysis, elevated liver enzymes, low platelets
- hitt, heparin-induced thrombocytopenia and thrombosis
- hlh, hemophagocytic lymphohistiocytosis
- hus, hemolytic-uremic syndrome
- isth, international society for thrombosis and haemostasis
- ivig, intravenous immunoglobulin
- ldh, lactate nos, nitric oxide synthase
- net, neutrophil extracellular trap
- pai-1, plasminogen activator inhibitor 1
- pf4, platelet factor 4
- prr, pattern recognition receptor
- rbc, red blood cell
- scd, sickle cell disease
- sle, systemic lupus erythematosus
- tlr, toll-like receptor
- tf, tissue factor
- tfpi, tissue factor pathway inhibitor
- tma, thrombotic microangiopathy
- tnf-α, tumor necrosis factor-α
- tpe, therapeutic plasma exchange
- ulc, ultra large heparin-pf4 complexes
- ulvwf, ultra-large von willebrand factor
- vwf, von willebrand factor
Collapse
Affiliation(s)
- Monica A Bray
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas
| | - Sarah E Sartain
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas
| | - Jahnavi Gollamudi
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas
| | - Rolando E Rumbaut
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
7
|
Schreiber K, Hunt BJ. Managing antiphospholipid syndrome in pregnancy. Thromb Res 2020; 181 Suppl 1:S41-S46. [PMID: 31477227 DOI: 10.1016/s0049-3848(19)30366-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterised by the presence of antiphospholipid antibodies (aPL). The antibodies currently included in the classification criteria include lupus anticoagulant (LA), anticardiolipin antibodies (aCL) and anti-^2-glycoprotein 1 antibodies (^2GPI). APS can present with a variety of clinical phenotypes, including thrombosis in the veins, arteries and microvasculature and obstetrical complications. Pregnancy complications in obstetric APS (OAPS) include unexplained recurrent early pregnancy loss, fetal death, or premature birth due to severe preeclampsia, eclampsia, intrauterine growth restriction or other consequences of placental insufficiency. Careful, well monitored obstetric care with the use of aspirin and heparin has likely improved the pregnancy outcome in obstetric APS and currently approximately 70-80% of pregnant women with APS have a successful pregnancy outcome. However, the current standard of care does not prevent all pregnancy complications as the current treatment fails in 20-30% of APS pregnancies. Other treatments options are currently being explored and retrospective studies suggest that trials with hydroxychloroquine and possibly pravastatin are warranted in pregnant women with aPL. In this review will focus on the current treatment of OAPS.
Collapse
Affiliation(s)
- Karen Schreiber
- The Thrombosis & Haemophilia Centre, Guy's & St Thomas' NHS Foundation Trust, London SE1 7EH, UK; Copenhagen Lupus and Vasculitis Clinic, Center for Rheumatology and Spine Diseases, Copenhagen, Denmark
| | - Beverley J Hunt
- The Thrombosis & Haemophilia Centre, Guy's & St Thomas' NHS Foundation Trust, London SE1 7EH, UK.
| |
Collapse
|
8
|
Urbanski G, Caillon A, Poli C, Kauffenstein G, Begorre MA, Loufrani L, Henrion D, Belizna C. Hydroxychloroquine partially prevents endothelial dysfunction induced by anti-beta-2-GPI antibodies in an in vivo mouse model of antiphospholipid syndrome. PLoS One 2018; 13:e0206814. [PMID: 30399161 PMCID: PMC6219787 DOI: 10.1371/journal.pone.0206814] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/20/2018] [Indexed: 01/18/2023] Open
Abstract
Background Antiphospholipid syndrome is associated with endothelial dysfunction, which leads to thrombosis and early atheroma. Given that hydroxychloroquine has anti-thrombotic properties in lupus, we hypothesized that it could reduce endothelial dysfunction in an animal model of antiphospholipid syndrome. We evaluated the effect of hydroxychloroquine in preventing endothelial dysfunction in a mouse model of antiphospholipid syndrome. Methods Antiphospholipid syndrome was induced by an injection of monoclonal anti-beta-2-GPI antibodies. Vascular reactivity was evaluated in mesenteric resistance arteries isolated from mice 3 weeks (APL3W) after receiving a single injection of anti-beta-2-GPI antibodies and after 3 weeks of daily oral hydroxychloroquine treatment (HCQ3W) compared to control mice (CT3W). We evaluated endothelial dysfunction by measuring acetylcholine-mediated vasodilation. A pharmacological approach was used to evaluate NO synthase uncoupling (tetrahydrobiopterin) and the generation of reactive oxygen species (Tempol). Results Impaired acetylcholine-mediated dilation was evidenced in mice 3 weeks after anti-beta-2-GPI antibodies injection compared to CT3W, by reduced maximal dilation (p<0.0001) and sensitivity (pKd) (p = 0.01) to acetylcholine. Hydroxychloroquine improved acetylcholine-dependent dilation, on pKd (p = 0.02) but not maximal capacity compared to untreated mice. The addition of tetrahydrobiopterin (p = 0.02) and/or Tempol (p = 0.0008) improved acetylcholine-mediated dilation in APL3W but not in HCQ3W. Conclusions We demonstrated that endothelial dysfunction in mouse resistance arteries persisted at 3 weeks after a single injection of monoclonal anti-beta-2-GPI antibodies, and that hydroxychloroquine improved endothelium-dependent dilation at 3 weeks, through improvement of NO synthase coupling and oxidative stress reduction.
Collapse
Affiliation(s)
- Geoffrey Urbanski
- UMR CNRS 6015—INSERM U1083, MITOVASC Institute, Université d’Angers, Faculté de Médecine, Bâtiment IRIS 2, Angers, France
- Service de Médecine Interne et Maladies Vasculaires, Centre Hospitalier Universitaire, Angers, France
- * E-mail:
| | - Antoine Caillon
- UMR CNRS 6015—INSERM U1083, MITOVASC Institute, Université d’Angers, Faculté de Médecine, Bâtiment IRIS 2, Angers, France
| | - Caroline Poli
- Laboratoire d’immunologie et d’allergologie, Centre Hospitalier Universitaire, Angers, France
| | - Gilles Kauffenstein
- UMR CNRS 6015—INSERM U1083, MITOVASC Institute, Université d’Angers, Faculté de Médecine, Bâtiment IRIS 2, Angers, France
| | - Marc-Antoine Begorre
- UMR CNRS 6015—INSERM U1083, MITOVASC Institute, Université d’Angers, Faculté de Médecine, Bâtiment IRIS 2, Angers, France
| | - Laurent Loufrani
- UMR CNRS 6015—INSERM U1083, MITOVASC Institute, Université d’Angers, Faculté de Médecine, Bâtiment IRIS 2, Angers, France
| | - Daniel Henrion
- UMR CNRS 6015—INSERM U1083, MITOVASC Institute, Université d’Angers, Faculté de Médecine, Bâtiment IRIS 2, Angers, France
| | - Cristina Belizna
- UMR CNRS 6015—INSERM U1083, MITOVASC Institute, Université d’Angers, Faculté de Médecine, Bâtiment IRIS 2, Angers, France
- Centre vasculaire et de la coagulation, Centre Hospitalier Universitaire, Angers, France
| |
Collapse
|
9
|
McDonnell TCR, Willis R, Pericleous C, Ripoll VM, Giles IP, Isenberg DA, Brasier AR, Gonzalez EB, Papalardo E, Romay-Penabad Z, Jamaluddin M, Ioannou Y, Rahman A. PEGylated Domain I of Beta-2-Glycoprotein I Inhibits the Binding, Coagulopathic, and Thrombogenic Properties of IgG From Patients With the Antiphospholipid Syndrome. Front Immunol 2018; 9:2413. [PMID: 30405613 PMCID: PMC6204385 DOI: 10.3389/fimmu.2018.02413] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/28/2018] [Indexed: 01/08/2023] Open
Abstract
APS is an autoimmune disease in which antiphospholipid antibodies (aPL) cause vascular thrombosis and pregnancy morbidity. In patients with APS, aPL exert pathogenic actions by binding serum beta-2-glycoprotein I (β2GPI) via its N-terminal domain I (DI). We previously showed that bacterially-expressed recombinant DI inhibits biological actions of IgG derived from serum of patients with APS (APS-IgG). DI is too small (7 kDa) to be a viable therapeutic agent. Addition of polyethylene glycol (PEGylation) to small molecules enhances the serum half-life, reduces proteolytic targeting and can decrease immunogenicity. It is a common method of tailoring pharmacokinetic parameters and has been used in the production of many therapies in the clinic. However, PEGylation of molecules may reduce their biological activity, and the size of the PEG group can alter the balance between activity and half-life extension. Here we achieve production of site-specific PEGylation of recombinant DI (PEG-DI) and describe the activities in vitro and in vivo of three variants with different size PEG groups. All variants were able to inhibit APS-IgG from: binding to whole β2GPI in ELISA, altering the clotting properties of human plasma and promoting thrombosis and tissue factor expression in mice. These findings provide an important step on the path to developing DI into a first-in-class therapeutic in APS.
Collapse
Affiliation(s)
- Thomas C. R. McDonnell
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
| | - Rohan Willis
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Charis Pericleous
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
| | - Vera M. Ripoll
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
| | - Ian P. Giles
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
| | - David. A. Isenberg
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
- Arthritis Research UK Centre for Adolescent Rheumatology, UCL/UCLH/Great Ormond Street Hospital, London, United Kingdom
| | - Allan R. Brasier
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Emilio B. Gonzalez
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Elizabeth Papalardo
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Zurina Romay-Penabad
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Mohammad Jamaluddin
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Yiannis Ioannou
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
- Arthritis Research UK Centre for Adolescent Rheumatology, UCL/UCLH/Great Ormond Street Hospital, London, United Kingdom
| | - Anisur Rahman
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
| |
Collapse
|
10
|
Noureldine MHA, Nour-Eldine W, Khamashta MA, Uthman I. Insights into the diagnosis and pathogenesis of the antiphospholipid syndrome. Semin Arthritis Rheum 2018; 48:860-866. [PMID: 30217394 DOI: 10.1016/j.semarthrit.2018.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/30/2018] [Accepted: 08/14/2018] [Indexed: 11/16/2022]
Abstract
The Antiphospholipid syndrome (APS), formerly known as Anticardiolipin or Hughes syndrome, is a systemic autoimmune disorder characterized by obstetrical complications and thrombotic events affecting almost every organ-system in patients persistently testing positive for antiphospholipid antibodies (aPL). The contribution of the extra-criteria aPL to the pathogenesis of APS have exceeded the expectations of a simple, direct pathologic 'hit' leading to thrombogenesis or obstetrical complications, and more pathologic pathways are being linked directly or indirectly to aPL. The value of extra-criteria aPL is on the rise, and these antibodies are nowadays evaluated as markers for risk assessment in the diagnostic approach to APS. A diagnosis of APS should be considered in pediatric patients with suggestive clinical and laboratory picture. Management of APS remains mostly based on anticoagulation, while other drugs are being tested for efficacy and side effects. Low-dose aspirin may have a role in the management of thrombotic and obstetric APS. Due to the high variability in disease severity and complication recurrence outcomes, new tools are being developed and validated to assess the damage index and quality of life of APS patients.
Collapse
Affiliation(s)
| | - Wared Nour-Eldine
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Imad Uthman
- Division of Rheumatology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Antiphospholipid syndrome (APS) is defined as the association of thrombotic events and/or obstetric morbidity in patients persistently positive for antiphospholipid antibodies (aPL). In this review, we will highlight the most important clinical presentations of APS with a focus on the obstetric morbidity, the current management strategies and the outlook for the future. RECENT FINDINGS The use of aspirin and heparin has improved the pregnancy outcome in obstetric APS and approximately 70% of pregnant women with APS have a successful pregnancy outcome. Unfortunately, the current standard of care does not prevent all pregnancy complications as the current treatment fails in 20-30% of APS pregnancies. This therefore highlights the need for alternative treatments to improve obstetrical outcome. Other treatment options are currently explored and retrospective studies show that pravastatin for example is beneficial in women with aPL-related early preeclampsia. Moreover, the immunmodulator hydroxychloroquine may play a beneficial role in the prevention of aPL-related pregnancy complications. SUMMARY APS is among the most frequent acquired risk factors for a treatable cause of recurrent pregnancy loss and increases the risk of conditions associated with ischaemic placental dysfunction, such as fetal growth restriction, preeclampsia, premature birth and intrauterine death. Current treatment is mainly based on aspirin and heparin. Studies to inform on alternative treatment options are urgently needed.
Collapse
|
12
|
Radic M, Pattanaik D. Cellular and Molecular Mechanisms of Anti-Phospholipid Syndrome. Front Immunol 2018; 9:969. [PMID: 29867951 PMCID: PMC5949565 DOI: 10.3389/fimmu.2018.00969] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 04/18/2018] [Indexed: 01/10/2023] Open
Abstract
The primary anti-phospholipid syndrome (APS) is characterized by the production of antibodies that bind the phospholipid-binding protein β2 glycoprotein I (β2GPI) or that directly recognize negatively charged membrane phospholipids in a manner that may contribute to arterial or venous thrombosis. Clinically, the binding of antibodies to β2GPI could contribute to pathogenesis by formation of immune complexes or modification of coagulation steps that operate along cell surfaces. However, additional events are likely to play a role in pathogenesis, including platelet and endothelial cell activation. Recent studies focus on neutrophil release of chromatin in the form of neutrophil extracellular traps as an important disease contributor. Jointly, the participation of both the innate and adaptive arms of the immune system in aspects of the APS make the complete understanding of crucial steps in pathogenesis extremely difficult. Only coordinated and comprehensive analyses, carried out in different clinical and research settings, are likely to advance the understanding of this complex disease condition.
Collapse
Affiliation(s)
- Marko Radic
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Debendra Pattanaik
- Division of Rheumatology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
13
|
Manukyan D, Müller-Calleja N, Lackner K. Pathophysiological insights into the antiphospholipid syndrome. Hamostaseologie 2017; 37:202-207. [DOI: 10.5482/hamo-16-07-0020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/06/2016] [Indexed: 11/05/2022] Open
Abstract
SummaryThe antiphospholipid syndrome (APS) is characterized by venous and/or arterial thrombosis and severe pregnancy morbidity in presence of antiphospholipid antibodies (aPL). While there is compelling evidence that aPL cause the clinical manifestations of APS, the underlying mechanisms are still a matter of scientific debate. This is mainly related to the broad heterogeneity of aPL. There are three major types of aPL: The first one binds to (anionic) phospholipids, e.g. cardiolipin, in absence of other factors (cofactor independent aPL). The second type binds to phospholipids only in presence of protein cofactors, e.g. ß2-glycoprotein I (ß2GPI) (cofactor dependent aPL). The third type binds to cofactor proteins directly without need for phospholipids. It is widely believed that cofactor independent aPL (type 1) are associated with infections and, more importantly, non-pathogenic, while pathogenic aPL belong to the second and in particular to the third type. This view, in particular with regard to type 1 aPL, has not been undisputed and novel research data have shown that it is in fact untenable. We summarize the available data on the pathogenetic role of aPL and the implications for diagnosis of APS and future research.
Collapse
|
14
|
Papalardo E, Romay-Penabad Z, Willis R, Christadoss P, Carrera-Marin AL, Reyes-Maldonado E, Rudrangi R, Alfieri-Papalardo S, Garcia-Latorre E, Blank M, Pierangeli S, Brasier AR, Gonzalez EB. Major Histocompatibility Complex Class II Alleles Influence Induction of Pathogenic Antiphospholipid Antibodies in a Mouse Model of Thrombosis. Arthritis Rheumatol 2017; 69:2052-2061. [PMID: 28666081 DOI: 10.1002/art.40195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/27/2017] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Both environmental and genetic factors are important in the development of antiphospholipid antibodies (aPL) in patients with antiphospholipid syndrome (APS). Currently, the only available data on predisposing genetic factors have been obtained from epidemiologic studies, without mechanistic evidence. Therefore, we studied the influence of major histocompatibility complex (MHC) class II alleles on the production of aPL in a mouse model of APS. METHODS Three groups of mice, MHC class II-deficient (MHCII-/- ) mice, MHCII-/- mice transgenic for human HLA-DQ6 (DQ6), DQ8, or DR4 alleles, and the corresponding wild-type (WT) mouse strains were immunized; half were immunized with human β2 -glycoprotein I (β2 GPI), and the other half were immunized with control ovalbumin (OVA) protein. Thrombus formation in vivo, tissue factor activity in carotid and peritoneal macrophages, and serum levels of tumor necrosis factor (TNF), IgG anticardiolipin (aCL), antibodies, and anti-OVA antibodies were determined. RESULTS Immunization with β2 GPI induced significant production of aCL and anti-β2 GPI in WT mice compared with control mice immunized with OVA (P < 0.001) but diminished aCL (P < 0.001) and anti-β2 GPI (P = 0.016) production in MHCII-/- mice. Anti-β2 GPI production was fully restored in DQ6 and DQ8 mice, while levels of anti-β2 GPI in DR4 mice and aCL in all transgenic lines were only partially restored (P < 0.001 to P < 0.046). Thrombus size in WT mice was twice that in MHCII-/- mice (P < 0.001) but similar to that in all transgenic lines. Carotid and peritoneal macrophage tissue factor levels decreased by >50% in MHCII-/- mice compared with wild-type B6 mice and were restored in DQ8 mice but not DR4 mice or DQ6 mice. TNF levels decreased 4-fold in MHCII-/- mice (P < 0.001) and were not restored in transgenic mice. CONCLUSION Our mechanistic study is the first to show that MHC class II alleles influence not only quantitative aPL production but also the pathogenic capacity of induced aPL.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Miri Blank
- Sheba Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | | |
Collapse
|
15
|
Abstract
Autoimmune factors are involved in some of the cases of reproductive failure. These factors entail several autoantibodies, especially in patients having systemic lupus erythematosus (SLE) or the antiphospholipid syndrome (APS). These autoantibodies include mainly antibodies directed to phospholipid such as cardiolipin, phosphatidylserine, phosphatidylethanolamine or phospholipids binding glycoproteins such as b2glycoprotein-I, annexin V, prothrombin and protein-Z. There are also some other autoantibodies directed to laminin-I, thromboplastin, mitochondrial antibodies of the M5 type, corpus luteum, prolactin, poly (ADP-ribose), thyroglobulin and more, which were also found in SLE or APS patients with reproductive failure. Moreover, the presence of additional autoantibodies directed to actin, enolase, cubilin and others, needs further investigation to support a firm association to reproductive failure in women. Future studies are likely to help to determine and expand the number of autoantibodies screened in these patients, as well as by the use of proteomics technology, to determine peptides resembling the epitope specificities associated with the specific clinical manifestations.
Collapse
Affiliation(s)
- Y Shoenfeld
- Internal Medicine B and The Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Israel.
| | | |
Collapse
|
16
|
George J, Afek A, Gilburd B, Levy Y, Blank M, Kopolovic J, Harats D, Shoenfeld Y. Atherosclerosis in LDL-receptor knockout mice is accelerated by immunization with anticardiolipin antibodies. Lupus 2016. [DOI: 10.1177/096120339700600908] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Atherosclerosis is a process initiated by accumulation of macrophages in distinct areas of endothelial cell damage and uptake of large amounts of lipids. Recently, it has been shown that the immune system plays an active part in the progression of the atherosclerotic plaque although its precise role has not yet been elucidated. Anticardiolipin antibodies (aCL) are generally found in the sera of patients with the antiphospholipid syndrome (APS) and are associated with a prothrombotic state. Several authors have demonstrated that aCL can activate platelets and endothelial cells as well as increase oxidized low density lipoprotein (LDL) uptake by macrophages. In the present study we sought to assess the effect of immunization with aCL (Ab1, leading to the production of mouse aCL-Ab3) on the progression of atherosclerosis. Two groups of 8-weeks old female LDL-receptor knockout mice (n = 13 per group) were immunized with IgG purified from the serum of an APS patient or with normal human IgG, respectively. The aCL immunized mice developed high titres of 'self' aCL (detected using the standard aCL ELISA) as compared with the normal human IgG immunized mice, whereas no differences were noted between both study groups with respect to the serum lipid levels. The extent of fatty streak formation was significantly higher in the aCL immunized mice in comparison with the human IgG injected mice (mean aortic lesion size of 5308 ± 471 μm2 vs 1027 ± 184 μm2, respectively, P < 0.01). The immunohistochemical analysis of the atherosclerotic plaques from both mouse groups did not display differences in cellular composition. The results of the study show that mouse aCL induced by immunization with human aCL from an APS patient enhance atherogenesis in LDL-RKO mice and imply that these antibodies may play a role in atherosclerosis development in patients with the APS.
Collapse
Affiliation(s)
- J. George
- Research Unit of Autoimmune Diseases, Department of Medicine 'B', Tel Aviv University
| | - A. Afek
- Institute of Pathology, Tel Aviv University
| | - B. Gilburd
- Institute of Lipid and Atherosclerosis Research, Sheba Medical Centre, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Y. Levy
- Research Unit of Autoimmune Diseases, Department of Medicine 'B', Tel Aviv University
| | - M. Blank
- Research Unit of Autoimmune Diseases, Department of Medicine 'B', Tel Aviv University
| | | | - D. Harats
- Institute of Lipid and Atherosclerosis Research, Sheba Medical Centre, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Y. Shoenfeld
- Research Unit of Autoimmune Diseases, Department of Medicine 'B', Tel Aviv University
| |
Collapse
|
17
|
Antiphospholipid Antibodies: Their Origin and Development. Antibodies (Basel) 2016; 5:antib5020015. [PMID: 31557996 PMCID: PMC6698834 DOI: 10.3390/antib5020015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/18/2016] [Accepted: 05/19/2016] [Indexed: 11/25/2022] Open
Abstract
Antiphospholipid antibodies (aPL) are a hallmark of the antiphospholipid syndrome (APS), which is the most commonly acquired thrombophilia. To date there is consensus that aPL cause the clinical manifestations of this potentially devastating disorder. However, there is good evidence that not all aPL are pathogenic. For instance, aPL associated with syphilis show no association with the manifestations of APS. While there has been intensive research on the pathogenetic role of aPL, comparably little is known about the origin and development of aPL. This review will summarize the current knowledge and understanding of the origin and development of aPL derived from animal and human studies.
Collapse
|
18
|
Lackner KJ, Müller-Calleja N. Pathogenesis of the antiphospholipid syndrome revisited: time to challenge the dogma. J Thromb Haemost 2016; 14:1117-20. [PMID: 26998919 DOI: 10.1111/jth.13320] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/10/2016] [Indexed: 01/27/2023]
Abstract
For more than a decade the antiphospholipid syndrome (APS) has been reported to be caused mainly by antiphospholipid antibodies (aPL), which are not directed against phospholipids but against a complex of phospholipids and phospholipid binding proteins, so called cofactors (e.g. β2-glycoprotein I [β2GPI]). In fact, many researchers propose that the only relevant antigens in the APS are the cofactors themselves, with β2GPI being the most important. Antibodies that bind to phospholipids in a cofactor-independent manner are considered insignificant for the pathogenesis of the APS. We review the evidence for this current pathophysiologic concept and argue that it has never been proven and is now clearly no longer tenable. First, there is undisputable evidence that cofactor-independent aPL are pathogenic and present in the blood of APS patients. Second, available epidemiologic and clinical studies do not support a dominant pathogenic role for anti-β2GPI.
Collapse
Affiliation(s)
- K J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Mainz, Mainz, Germany
| | - N Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Mainz, Mainz, Germany
| |
Collapse
|
19
|
Manukyan D, Müller-Calleja N, Jäckel S, Luchmann K, Mönnikes R, Kiouptsi K, Reinhardt C, Jurk K, Walter U, Lackner KJ. Cofactor-independent human antiphospholipid antibodies induce venous thrombosis in mice. J Thromb Haemost 2016; 14:1011-20. [PMID: 26786324 DOI: 10.1111/jth.13263] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
Abstract
UNLABELLED Essentials Cofactor-independent antiphospholipid antibodies (CI-aPL) are generally considered non-pathogenic. We analyzed the effects of human monoclonal CI-aPL in a mouse model of venous thrombosis. As shown in vitro, CI-aPL induce a procoagulant state in vivo by activation of endosomal NADPH-oxidase. Contrary to common belief, CI-aPL induce venous thrombosis in vivo. SUMMARY Background There is general consensus that the antiphospholipid syndrome (APS) is caused by antiphospholipid antibodies (aPL) with antibodies against β2-glycoprotein-I being the most relevant. aPL that bind phospholipids in the absence of protein cofactors are generally considered pathogenetically irrelevant. We showed that cofactor-independent human monoclonal aPL isolated from APS patients induce proinflammatory and procoagulant cellular responses by activating endosomal NADPH-oxidase 2 (NOX2). Similar aPL were detected in all IgG fractions from APS patients analyzed. Objectives We aimed to clarify if cofactor-independent aPL can be thrombogenic in vivo and, if so, whether these effects are mediated via activation of NOX2. Methods Two cofactor-independent human monoclonal aPL, HL5B and RR7F, were tested in a mouse model of venous thrombosis. Genetically modified mice and in vitro assays were used to delineate the mechanisms underlying thrombus induction. Results HL5B and RR7F dramatically accelerate thrombus formation in this mouse model. Thrombus formation depends on tissue factor activation. It cannot be induced in NOX2-deficient mice. Bone marrow chimeras of C57BL/6J mice reconstituted with NOX2-deficient bone marrow showed that leukocyte activation plays a major role in thrombus formation. Neither TLR4 signaling nor platelet activation by our aPL is required for venous thrombus formation. Conclusions Cofactor-independent aPL can induce thrombosis in vivo. This effect is mainly mediated by leukocyte activation, which depends on the previously described signal transduction via endosomal NOX2. Because most APS patients have been shown to harbor aPL with similar activity, our data are of general relevance for the APS.
Collapse
Affiliation(s)
- D Manukyan
- Institute of Clinical Chemistry and Laboratory Medicine, Mainz, Germany
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - N Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, Mainz, Germany
| | - S Jäckel
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - K Luchmann
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - R Mönnikes
- Institute of Clinical Chemistry and Laboratory Medicine, Mainz, Germany
| | - K Kiouptsi
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - C Reinhardt
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - K Jurk
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - U Walter
- Center for Thrombosis and Haemostasis, University Medical Centre Mainz, Mainz, Germany
| | - K J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Mainz, Germany
| |
Collapse
|
20
|
Terrisse AD, Laurent PA, Garcia C, Gratacap MP, Vanhaesebroeck B, Sié P, Payrastre B. The class I phosphoinositide 3-kinases α and β control antiphospholipid antibodies-induced platelet activation. Thromb Haemost 2016; 115:1138-46. [PMID: 26818901 DOI: 10.1160/th15-08-0661] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/13/2016] [Indexed: 12/29/2022]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterised by the presence of antiphospholipid antibodies (aPL) associated with increased thrombotic risk and pregnancy morbidity. Although aPL are heterogeneous auto-antibodies, the major pathogenic target is the plasma protein β2-glycoprotein 1. The molecular mechanisms of platelet activation by aPL remain poorly understood. Here, we explored the role of the class IA phosphoinositide 3-kinase (PI3K) α and β isoforms in platelet activation by aPL. Compared to control IgG from healthy individuals, the IgG fraction isolated from patients with APS potentiates platelet aggregation induced by low dose of thrombin in vitro and increases platelet adhesion and thrombus growth on a collagen matrix under arterial shear rate through a mechanism involving glycoprotein Ib (GPIb) and Toll Like Receptor 2 (TLR-2). Using isoforms-selective pharmacological PI3K inhibitors and mice with megakaryocyte/platelet lineage-specific inactivation of class IA PI3K isoforms, we demonstrate a critical role of the PI3Kβ and PI3Kα isoforms in platelet activation induced by aPL. Our data show that aPL potentiate platelet activation through GPIbα and TLR-2 via a mechanism involving the class IA PI3Kα and β isoforms, which represent new potential therapeutic targets in the prevention or treatment of thrombotic events in patients with APS.
Collapse
Affiliation(s)
- Anne-Dominique Terrisse
- Anne-Dominique Terrisse, Inserm U1048, I2MC, 1 Avenue Jean Poulhés, BP 84225, 31432 Toulouse Cedex 04, France, Tel.: +33 5 3122 4150, Fax: +33 5 6132 5621, E-mail:
| | | | | | | | | | | | | |
Collapse
|
21
|
McDonnell T, Pericleous C, Laurine E, Tommasi R, Garza-Garcia A, Giles I, Ioannou Y, Rahman A. Development of a high yield expression and purification system for Domain I of Beta-2-glycoprotein I for the treatment of APS. BMC Biotechnol 2015; 15:104. [PMID: 26576675 PMCID: PMC4650279 DOI: 10.1186/s12896-015-0222-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 11/03/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In this paper we describe a novel method to achieve high yield bacterial expression of a small protein domain with considerable therapeutic potential; Domain I of Beta-2-glycoprotein I (β2GPI). β2GPI is intrinsic to the pathological progression of the Antiphospholipid Syndrome (APS). Patients develop autoantibodies targeting an epitope located on the N-terminal Domain I of β2GPI rendering this domain of interest as a possible therapeutic. RESULTS This new method of production of Domain I of β2GPI has increased the production yield by ~20 fold compared to previous methods in E.coli. This largely scalable, partially automated method produces 50-75 mg of pure, folded, active Domain I of β2GPI per litre of expression media. CONCLUSION The application of this method may enable production of Domain I on sufficient scale to allow its use as a therapeutic.
Collapse
Affiliation(s)
- Thomas McDonnell
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK.
| | - Charis Pericleous
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK.
| | - Emmanuelle Laurine
- PolyTherics, Babraham Research Campus, Babraham, CB22 3AT, Cambridge, UK.
| | - Rita Tommasi
- PolyTherics, Babraham Research Campus, Babraham, CB22 3AT, Cambridge, UK.
| | - Acely Garza-Garcia
- Structural Biology, Medical Research Council National Institute for Medical Research, London, UK.
| | - Ian Giles
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK.
| | - Yiannis Ioannou
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK. .,Arthritis Research UK Centre for Adolescent Rheumatology, University College London, London, UK.
| | - Anisur Rahman
- Centre for Rheumatology, Division of Medicine, University College London, Rayne Institute, 5 University Street, London, WC1E 6JF, UK.
| |
Collapse
|
22
|
Hood DB, Snyder KR, Buckner TR, Hurley BL, Pitts KR, Lopez LR. Differential assay reactivity of immunglobulin A anti-ß 2 glycoprotein I antibodies: implications for the clinical interpretation of antiphospholipid antibody testing. Eur J Rheumatol 2015; 2:135-138. [PMID: 27708950 DOI: 10.5152/eurjrheum.2015.0012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/17/2015] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The routine measurement of IgA anticardiolipin (aCL) and IgA anti-β2 glycoprotein I (anti-β2 GPI) antibodies remain controversial despite several studies demonstrating an association with thromboembolic disease in patients with systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS). This controversy may be a contributing factor for the current under use of IgA antiphospholipid antibodies. We aimed to investigate the nature of discrepant IgA anti-β2 GPI reactivity to help define the diagnostic value of IgA antiphospholipid antibodies. MATERIAL AND METHODS Four sera selected from SLE/APS patients and positive for antiphospholipid antibodies but having discrepant IgA anti-β2 GPI reactivity on two commercial assays were studied. IgA antibodies were affinity purified to investigate anti-β2 GPI reactivity. Column wash through and eluent fractions were tested on both IgA anti-β2 GPI assays. Results were normalized to total protein. Assay conjugates and standards from the discrepant assays were interchanged. RESULTS The diseased samples were strongly positive in one assay [144-388 IgA antiphospholipid (APL) units] and negative or weakly positive in another assay (9.9-53 APL units). IgA eluents from IgA anti-β2 GPI positive samples reacted 10 times stronger on the reactive assay. When normalized to protein content, the eluents showed no cross-reactivity for IgG or IgM anti-β2 GPI antibodies, confirming IgA isotype specificity. Conjugate interchange confirmed that both assays bound IgA anti-β2 GPI antibodies, but the anti-IgA conjugate from the reactive assay was 4 times stronger, suggesting that its ability to detect IgA anti-β2 GPI antibodies was partially dependent on the anti-IgA conjugate and calibration. CONCLUSION These results confirm not only the presence of IgA anti-β2 GPI antibodies in the selected patient samples but also highlight an IgA conjugate issue for the unreactive assay, causing an underestimation of IgA anti-β2 GPI. This finding may assist in the ongoing standardization efforts of APS antibody testing. In addition, conclusions from published clinical studies may need to be revised as some assays may understate IgA significance.
Collapse
Affiliation(s)
- David B Hood
- Corgenix Medical Corporation, Broomfield, Colorado, USA
| | | | | | - Beth L Hurley
- Corgenix Medical Corporation, Broomfield, Colorado, USA
| | - Kelly R Pitts
- Corgenix Medical Corporation, Broomfield, Colorado, USA
| | - Luis R Lopez
- Corgenix Medical Corporation, Broomfield, Colorado, USA
| |
Collapse
|
23
|
Romay-Penabad Z, Carrera Marin AL, Willis R, Weston-Davies W, Machin S, Cohen H, Brasier A, Gonzalez EB. Complement C5-inhibitor rEV576 (coversin) ameliorates in-vivo effects of antiphospholipid antibodies. Lupus 2015; 23:1324-6. [PMID: 25228739 DOI: 10.1177/0961203314546022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Activation of the complement cascade is an important mechanism for antiphospholipid antibody-mediated thrombosis. We examined the effects of rEV576 (coversin), a recombinant protein inhibitor of complement factor 5 activation, on antiphospholipid antibody-mediated tissue factor up-regulation and thrombosis. Groups of C57BL/6J mice (n=5) received either IgG from a patient with antiphospholipid syndrome (APS) or control IgG from normal human serum (NHS). Each of these groups of mice had IgG administration preceded by either rEV576, or phosphate buffer control. For each of the four treatment groups, the size of induced thrombus, tissue factor activity in carotid homogenates, anticardiolipin and anti-β2glycoprotein I (anti-β2GPI) levels were measured 72 h after the first injection. Mice treated with IgG-APS had significantly higher titers of anticardiolipin antibodies and anti-β2GPI at thrombus induction compared with those treated with IgG-NHS. The IgG-APS/phosphate buffer treatment induced significantly larger thrombi and tissue factor activity compared with other groups. Mice treated with IgG-APS/rEV576 had significantly smaller thrombi and reduced tissue factor activity than those treated with IgG-APS/phosphate buffer. The data confirm involvement of complement activation in antiphospholipid antibody-mediated thrombogenesis and suggest that complement inhibition might ameliorate this effect.
Collapse
Affiliation(s)
| | | | - R Willis
- University of Texas Medical Branch, Galveston, USA
| | | | - S Machin
- University College London, London, UK
| | - H Cohen
- University College London, London, UK
| | - A Brasier
- University of Texas Medical Branch, Galveston, USA
| | - E B Gonzalez
- University of Texas Medical Branch, Galveston, USA
| |
Collapse
|
24
|
Kolyada A, Karageorgos I, Mahlawat P, Beglova N. An A1-A1 mutant with improved binding and inhibition of β2GPI/antibody complexes in antiphospholipid syndrome. FEBS J 2015; 282:864-73. [PMID: 25546421 DOI: 10.1111/febs.13185] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 12/01/2014] [Accepted: 12/24/2014] [Indexed: 11/29/2022]
Abstract
β2 glycoprotein I (β2GPI) is the most common antigen for autoimmune antibodies in antiphospholipid syndrome (APS). Thrombosis is a clinical feature of APS. We created a molecule (A1-A1) that consists of two identical β2GPI-binding modules from ApoE receptor 2 (ApoER2). A1-A1 binds to β2GPI/antibody complexes, preventing their association with ApoER2 and anionic phospholipids, and reducing thrombus size in the mouse model of APS. Here, we describe a mutant of A1-A1 (mA1-A1ND) with improved affinity for β2GPI. mA1-A1ND inhibits the binding of β2GPI to cardiolipin in the presence of anti-β2GPI antibodies, and inhibits the binding to phospholipids in plasma samples of APS patients, affecting the clotting time. Reduction of the clotting time demonstrates the presence of soluble β2GPI/antibody complexes in patients' plasma. These complexes either already exist in patients' plasma or form rapidly in the proximity to phospholipids. All members of the low-density lipoprotein receptor family bind β2GPI. Modeling studies of A1 in a complex with domain V of β2GPI (β2GPI-DV) revealed two possible modes of interaction of a ligand-binding module from lipoprotein receptors with β2GPI-DV. In both orientations, the ligand-binding module interferes with binding of β2GPI to anionic phospholipids; however, it interacts with two different but overlapping sets of lysine residues in β2GPI-DV, depending on the orientation.
Collapse
Affiliation(s)
- Alexey Kolyada
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
25
|
Platelets are required for enhanced activation of the endothelium and fibrinogen in a mouse thrombosis model of APS. Blood 2014; 124:611-22. [PMID: 24825863 DOI: 10.1182/blood-2014-02-554980] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Antiphospholipid syndrome (APS) is defined by thrombosis, fetal loss, and the presence of antiphospholipid antibodies, including anti-β2-glycoprotein-1 autoantibodies (anti-β2GP1) that have a direct role in the pathogenesis of thrombosis in vivo. The cellular targets of the anti-β2GP1 autoantibody/β2GP1 complex in vivo were studied using a laser-induced thrombosis model of APS in a live mouse and human anti-β2GP1 autoantibodies affinity-purified from APS patients. Cell binding of fluorescently labeled β2GP1 and anti-β2GP1 autoantibodies revealed their colocalization on the platelet thrombus but not the endothelium. Anti-β2GP1 autoantibodies enhanced platelet activation, monitored by calcium mobilization, and endothelial activation, monitored by intercellular adhesion molecule-1 expression. When eptifibatide was infused to block platelet thrombus formation, enhanced fibrin generation and endothelial cell activation were eliminated. Thus, the anti-β2GP1 autoantibody/β2GP1 complex binds to the thrombus, enhancing platelet activation, and platelet secretion leads to enhanced endothelium activation and fibrin generation. These results lead to a paradigm shift away from the concept that binding of the anti-β2GP1 autoantibody/β2GP1 complex activates both endothelial cells and platelets toward one in which activation of platelets in response to anti-β2GP1 autoantibody/β2GP1 complex binding leads to subsequent enhanced endothelium activation and fibrin generation.
Collapse
|
26
|
Pons I, Espinosa G, Cervera R. [Efficacy and safety of rituximab in the treatment of primary antiphospholipid syndrome: analysis of 24 cases from the bibliography review]. Med Clin (Barc) 2014; 144:97-104. [PMID: 24698717 DOI: 10.1016/j.medcli.2014.01.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 01/22/2014] [Accepted: 01/23/2014] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND OBJECTIVE Antiphospholipid syndrome (APS) is characterized by the presence of antiphospholipid antibodies (aPL) and thrombotic and/or obstetric manifestations. Patients without another associated autoimmune disease are considered to have primary APS. Some patients develop thrombosis recurrence despite anticoagulant treatment and some clinical features do not respond to standard therapy. Rituximab may be an alternative in these cases. We review the published scientific evidence on the use of rituximab in the treatment of primary APS. PATIENTS AND METHODS Description of a case and review of the literature with descriptive analysis of the demographic, clinical, and immunologic features, treatment and outcome of patients. RESULTS We identified 24 patients (15 women [62.5%]), with a mean age of 37.0 ± 13.4 years. The reasons for the use of rituximab were thrombocytopenia (41.7%), skin involvement (33.3%), neurologic and heart valve involvement (12.5%), hemolytic anemia (8.3%) and pulmonary and renal involvement (4.2%). Lupus anticoagulant was present in 72.7% of the cases, the IgG and IgM isotypes of anticardiolipin antibodies in 75 and 50%, respectively, and the anti-β2GPI (IgG e IgM) antibodies in 80% of patients. Thirteen (54.1%) patients received 2 doses of 1,000 mg of rituximab fortnightly, 10 (41.7%) 4 doses of 375 mg/m(2) weekly and one (4.2%) 8 doses of 375 mg/m(2) weekly. Eleven (45.8%) patients presented a complete clinical response, 7 (29.2%) a partial response and 6 (25%) did not respond to rituximab. Four patients with clinical improvement presented with aPL titer decrease and in one patient, aPL levels did not change. In one patient without clinical response, aPL remained positive. A clinical-immunologic dissociation existed in 2 additional cases. CONCLUSIONS The results obtained suggest a possible potential benefit of rituximab in the treatment of some clinical manifestations of primary APS such as thrombocytopenia, skin and heart valve involvement.
Collapse
Affiliation(s)
- Isaac Pons
- Unidad de Enfermedades Autoinmunes Sistémicas, Servicio de Medicina Interna, Hospital d'Igualada, Igualada, Barcelona, España
| | - Gerard Espinosa
- Servicio de Enfermedades Autoinmunes Sistémicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, España.
| | - Ricard Cervera
- Servicio de Enfermedades Autoinmunes Sistémicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, España
| |
Collapse
|
27
|
Murthy V, Willis R, Romay-Penabad Z, Ruiz-Limón P, Martínez-Martínez LA, Jatwani S, Jajoria P, Seif A, Alarcón GS, Papalardo E, Liu J, Vilá LM, McGwin G, McNearney TA, Maganti R, Sunkureddi P, Parekh T, Tarantino M, Akhter E, Fang H, Gonzalez EB, Binder WR, Norman GL, Shums Z, Teodorescu M, Reveille JD, Petri M, Pierangeli SS. Value of isolated IgA anti-β2 -glycoprotein I positivity in the diagnosis of the antiphospholipid syndrome. ACTA ACUST UNITED AC 2014; 65:3186-93. [PMID: 23983008 DOI: 10.1002/art.38131] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 08/06/2013] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To examine the prevalence of isolated IgA anti-β2 -glycoprotein I (anti-β2 GPI) positivity and the association of these antibodies, and a subgroup that bind specifically to domain IV/V of β2 GPI, with clinical manifestations of the antiphospholipid syndrome (APS) in 3 patient groups and to evaluate the pathogenicity of IgA anti-β2 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-β2 GPI titers and binding to domain IV/V of β2 GPI were examined by enzyme-linked immunosorbent assay (ELISA). CD1 mice were inoculated with purified IgA anti-β2 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-β2 GPI isotype, and 57 patients were positive exclusively for IgA anti-β2 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-β2 GPI-positive serum samples reacted with domain IV/V of anti-β2 GPI, and 77% of those had clinical features of APS. Isolated IgA anti-β2 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-β2 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-β2 GPI antibodies induced significantly larger thrombi and higher TF levels compared to controls. CONCLUSION Isolated IgA anti-β2 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-β2 GPI antibodies directed to domain IV/V of β2 GPI represent an important subgroup of clinically relevant antiphospholipids.
Collapse
|
28
|
WANG TING, ZHOU HONG, XIE HONGXIANG, MU YUAN, XU YA, LIU JINGJING, ZHANG XIAOLEI. Epigallocatechin-3-gallate inhibits TF and TNF-α expression induced by the anti-β2GPI/β2GPI complex in human THP-1 cells. Int J Mol Med 2014; 33:994-1002. [DOI: 10.3892/ijmm.2014.1635] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 01/10/2014] [Indexed: 11/06/2022] Open
|
29
|
B-Cell Targeted Therapies in Autoimmune Cytopenias and Thrombosis. MILESTONES IN DRUG THERAPY 2014. [PMCID: PMC7123699 DOI: 10.1007/978-3-0348-0706-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ever since the advent of Rituximab and subsequently the emergence of other compounds targeting B cells, a cornucopia of medical applications have been found for this family of compounds. After their establishment as standard of care in many conditions such as rituximab in lymphoma and rheumatoid arthritis, they have been progressively found to aid in the treatment of many other conditions. This area constituted a fertile area of research in the past 12 years. Physicians have investigated the B-cell depleting agents use in cases of autoimmune hematologic cytopenias such as immune thrombocytopenia, Evans syndrome, cold and warm autoimmune hemolytic anemia, and other thrombophilic disorders such as the antiphospholipid syndrome and thrombocytopenic purpura. This chapter presents a historical perspective reviewing the various studies that have been published in this field. In addition, it offers a current assessment of the evidence regarding the use of B-cell depleting agents in the aforementioned conditions.
Collapse
|
30
|
Abstract
AIM To investigate the effects of affinity-purified rabbit anti-β2GP1, and anti-β2GP1 purified from patients with systemic lupus erythematosus (SLE), on adenosine diphosphate (ADP)-induced aggregation. METHODS Whole blood was collected and processed to obtain platelet poor plasma (PPP) from normal controls (n = 15) and SLE patients (n = 15). Using PPP, anti-β2GP1 titres were determined using an ELISA and IgG fractions isolated using a HiTrap protein G column. Anti-β2GP1 was purified from two SLE patients using purified β2GP1 coupled to a HiTrap NHS-activated HP column. RESULTS The effect of rabbit and human derived anti-β2GP1 (0-100 μg/mL), on ADP (2.5, 5 μM) induced platelet aggregation were investigated using light transmission aggregometry. Rabbit anti-β2GP1 significantly inhibited all parameters of 5 μM ADP-induced platelet aggregation; %Max (p = 0.028), %AUC (p = 0.014) and slope (p < 0.001). In contrast, anti-β2GP1 purified from SLE patients significantly enhanced the %Max (p = 0.031) and %AUC (p = 0.007) in a concentration dependent manner, but inhibited the slope (p < 0.05) of 5 μM ADP-induced platelet aggregation. CONCLUSION Our data suggest anti-β2GP1 purified from different species have variable effects on in vitro platelet aggregation. The disparity between rabbit and human anti-β2GP1 may be due to the heterogeneous nature of anti-β2GP1, varying avidity or different antibody binding specificities between species.
Collapse
|
31
|
Antiphospholipid syndrome: From pathogenesis to novel immunomodulatory therapies. Autoimmun Rev 2013; 12:752-7. [DOI: 10.1016/j.autrev.2012.12.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 12/19/2012] [Indexed: 02/02/2023]
|
32
|
Velayuthaprabhu S, Matsubayashi H, Sugi T, Nakamura M, Ohnishi Y, Ogura T, Archunan G. Expression of Apoptosis in Placenta of Experimental Antiphospholipid Syndrome Mouse. Am J Reprod Immunol 2013; 69:486-94. [DOI: 10.1111/aji.12084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 01/03/2013] [Indexed: 01/06/2023] Open
Affiliation(s)
| | | | | | | | - Yasuyuki Ohnishi
- Central Institute for Experimental Animals; Kawasaki; Kanagawa; Japan
| | - Tomoyuki Ogura
- Central Institute for Experimental Animals; Kawasaki; Kanagawa; Japan
| | - Govindaraju Archunan
- Department of Aniaml Science; Bharathidasan University; Tiruchirappalli; Tamil nadu; India
| |
Collapse
|
33
|
Khattri S, Zandman-Goddard G, Peeva E. B-cell directed therapies in antiphospholipid antibody syndrome — New directions based on murine and human data. Autoimmun Rev 2012; 11:717-22. [DOI: 10.1016/j.autrev.2011.12.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 12/22/2011] [Indexed: 12/31/2022]
|
34
|
Ioannou Y. The Michael Mason prize: Pathogenic antiphospholipid antibodies, stressed out antigens and the deployment of decoys. Rheumatology (Oxford) 2011; 51:32-6. [DOI: 10.1093/rheumatology/ker353] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
35
|
|
36
|
Blank M, Baraam L, Eisenstein M, Fridkin M, Dardik R, Heldman Y, Katchalski-Katzir E, Shoenfeld Y. β2-Glycoprotein-I based peptide regulate endothelial-cells tissue-factor expression via negative regulation of pGSK3β expression and reduces experimental-antiphospholipid-syndrome. J Autoimmun 2011; 37:8-17. [PMID: 21524885 DOI: 10.1016/j.jaut.2011.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2011] [Revised: 02/16/2011] [Accepted: 02/17/2011] [Indexed: 11/26/2022]
Abstract
Antiphospholipid syndrome (APS) is characterized by thromboembolic phenomena and recurrent fetal loss associated with elevated circulating anti-phospholipid/beta2glycoprotein-I(β2GPI)-binding-antibodies(Abs). Individual APS patients harbor diverse clusters of circulating anti-β2GPI Abs, targeting different epitopes on the β2GPI molecule. Our novel approach was to construct a peptide composed of β2GPI-ECs-binding-site (phospholipids-membrane), named "EMBI". EMBI exert dual activities: a) At first EMBI prevented β2GPI ECs binding, thus reduced by 89% the binding of β2GPI/anti-β2GPI to the cells in comparison with 9.3% inhibition by EMBI scrambled form (scEMBI). b) Longer exposure of ECs to EMBI resulted in intracellular EMBI penetration which did not prevent β2GPI/anti-β2GPI binding to HUVEC. Surprisingly, β2GPI/anti-β2GPI did not activate ECs harboring EMBI, illustrated by prevention of E-selectin and tissue factor (TF) expression. The inhibition of TF mRNA transcription was illustrated by quantitative RT-PCR. EMBI decreased the expression of phosphorylated JNK1/2, p38, HSP27 and enhanced phosphorylation of glycogen synthase kinase-3β (pGSK3β). Knocking down the GSK3β expression by siRNA-GSK3β, reduced the TF expression by β2GPI/anti-β2GPI-exposed-HUVEC. In-vivo, EMBI significantly decreased the percentage of fetal loss in naïve mice infused with anti-β2GPI Abs, p<0.04. Thus, the dual activity of EMBI may introduce EMBI as a potential novel candidate peptide, to treat patients with APS.
Collapse
Affiliation(s)
- Miri Blank
- Zabludowicz Center for Autoimmune Diseases, Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | | | | | | | | | | | | | | |
Collapse
|
37
|
β₂-Glycoprotein-1 autoantibodies from patients with antiphospholipid syndrome are sufficient to potentiate arterial thrombus formation in a mouse model. Blood 2011; 117:3453-9. [PMID: 21245481 DOI: 10.1182/blood-2010-08-300715] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antiphospholipid syndrome is characterized by thrombosis, recurrent fetal loss, and the presence of the lupus anticoagulant, anticardiolipin antibodies, or anti-β(2)-glycoprotein-1 (anti-β(2)-GP1) antibodies. Although anti-β(2)-GP1 antibodies have been documented as a biomarker for diagnosis of antiphospholipid syndrome, their direct role in the pathogenesis of thrombosis is unknown. We have demonstrated using intravital microscopy that anti-β(2)-GP1 autoantibodies purified from the sera of patients with antiphospholipid syndrome complicated by thrombosis greatly amplify thrombus size after laser-induced vessel wall injury in live mice. Anti-β(2)-GP1 autoantibodies from 3 patients with antiphospholipid syndrome were affinity-purified using human β(2)-GP1 bound to agarose. The effects of purified anti-β(2)-GP1 IgG autoantibodies, of anti-β(2)-GP1-depleted IgG, and of IgG from normal human sera on thrombus formation were measured in mice after arterial injury in the cremaster muscle. Before injury, purified anti-β(2)-GP1 IgG autoantibodies, anti-β(2)-GP1 antibody-depleted IgG, or IgG from normal human sera were infused. Increasing amounts of purified anti-β(2)-GP1 autoantibodies increased thrombus size in a dose-dependent manner, whereas neither anti-β(2)-GP1 antibody-depleted IgG nor IgG from normal serum affected thrombus size. These results indicate that anti-β(2)-GP1 IgG autoantibodies in antiphospholipid syndrome patient sera are not only a marker of antiphospholipid syndrome but are directly involved in the pathogenesis of thrombosis.
Collapse
|
38
|
Saleh T, Matta F, Alali F, Stein PD. Venous thromboembolism with chronic liver disease. Am J Med 2011; 124:64-8. [PMID: 21187186 DOI: 10.1016/j.amjmed.2010.08.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 06/24/2010] [Accepted: 08/06/2010] [Indexed: 12/21/2022]
Abstract
BACKGROUND patients with chronic liver disease have both antithrombotic and prothrombotic coagulation abnormalities. Published data conflict on whether patients with chronic liver disease have a high or low prevalence of venous thromboembolism. METHODS the number of patients discharged from hospitals throughout the US with a diagnostic code for chronic alcoholic and chronic nonalcoholic liver disease from 1979 through 2006 was obtained from the National Hospital Discharge Survey. We compared prevalences of venous thromboembolism among patients with chronic alcoholic liver disease and chronic nonalcoholic liver disease. RESULTS among 4,927,000 hospitalized patients with chronic alcoholic liver disease from 1979-2006, the prevalence of venous thromboembolism was 0.6%, compared with 0.9% among 4,565,000 hospitalized patients with chronic nonalcoholic liver disease. CONCLUSION the prevalence of venous thromboembolism in hospitalized patients with chronic liver disease, both alcoholic and nonalcoholic, was low. The prevalence of venous thromboembolism was higher in those with chronic non-alcoholic liver disease, but the difference was small and of no clinical consequence. Based on the literature, both showed a lower prevalence of venous thromboembolism than in hospitalized patients with most other medical diseases. It may be that both chronic alcoholic liver disease and chronic nonalcoholic liver disease have protective antithrombotic mechanisms, although the mechanisms differ.
Collapse
Affiliation(s)
- Tarek Saleh
- Department of Internal Medicine, St. Joseph Mercy Oakland Hospital, Pontiac, Mich., USA
| | | | | | | |
Collapse
|
39
|
Diaz JA, Hawley AE, Alvarado CM, Berguer AM, Baker NK, Wrobleski SK, Wakefield TW, Lucchesi BR, Myers DD. Thrombogenesis with continuous blood flow in the inferior vena cava. A novel mouse model. Thromb Haemost 2010; 104:366-75. [PMID: 20589322 DOI: 10.1160/th09-09-0672] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 03/16/2010] [Indexed: 11/05/2022]
Abstract
Several rodent models have been used to study deep venous thrombosis (DVT). However, a model that generates consistent venous thrombi in the presence of continuous blood flow, to evaluate therapeutic agents for DVT, is not available. Mice used in the present study were wild-type C57BL/6 (WT), plasminogen activator inhibitor-1 (PAI-1) knock out (KO) and Delta Cytoplasmic Tail (DCT). An electrolytic inferior vena cava (IVC) model (EIM) was used. A 25G stainless-steel needle, attached to a silver coated copper wire electrode (anode), was inserted into the exposed caudal IVC. Another electrode (cathode) was placed subcutaneously. A current of 250 muAmps over 15 minutes was applied. Ultrasound imaging was used to demonstrate the presence of IVC blood flow. Analyses included measurement of plasma soluble P-selectin (sP-Sel), thrombus weight (TW), vein wall morphometrics, P-selectin and Von Willebrand factor (vWF) staining, transmission electron microscopy (TEM), scanning electron microscopy (SEM); and the effect of enoxaparin on TW was evaluated. A current of 250 muAmps over 15 minutes consistently promoted thrombus formation in the IVC. Plasma sP-Sel was decreased in PAI-1 KO and increased in DCT vs. WT (WT/PAI-1: p=0.003, WT/DCT: p=0.0002). Endothelial activation was demonstrated by SEM, TEM, P-selectin and vWF immunohistochemistry and confirmed by inflammatory cell counts. Ultrasound imaging demonstrated thrombus formation in the presence of blood flow. Enoxaparin significantly reduced the thrombus size by 61% in this model. This EIM closely mimics clinical venous disease and can be used to study endothelial cell activation, leukocyte migration, thrombogenesis and therapeutic applications in the presence of blood flow.
Collapse
Affiliation(s)
- José A Diaz
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, Michigan 48109-0654, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Antiphospholipid antibody-mediated reproductive failure in antiphospholipid syndrome. Clin Rev Allergy Immunol 2010; 38:141-7. [PMID: 19562524 DOI: 10.1007/s12016-009-8146-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The association of elevated titers of circulating antiphospholipid (anti-PL) Abs in antiphospholipid syndrome (APS) and reproductive failure is well established in the literature. The clinical features include recurrent abortions at various stages, including implantation, placentation in the first trimester, miscarriages in the second and third trimesters, intrauterine growth retardation, preeclampsia with placental insufficiency and growth restrictions, arterial and venous thrombosis, and possibly also infertility. APS-mediated recurrent pregnancy loss and other features of reproductive failure might result from diverse autoimmune factors, inflammation, involving different mechanisms, which encompass pathogenic anti-PL Abs. Herein, we discuss the association of anti-PL Abs with reproductive failure with special emphasis on antiphospholipid autoantibodies characterizing APS. This association is evident from either human studies or murine models.
Collapse
|
41
|
Mode of Interaction between β2GPI and Lipoprotein Receptors Suggests Mutually Exclusive Binding of β2GPI to the Receptors and Anionic Phospholipids. Structure 2010; 18:366-76. [DOI: 10.1016/j.str.2009.12.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 11/30/2009] [Accepted: 12/29/2009] [Indexed: 12/20/2022]
|
42
|
Infections and autoimmunity--friends or foes? Trends Immunol 2009; 30:409-14. [PMID: 19643667 DOI: 10.1016/j.it.2009.05.005] [Citation(s) in RCA: 276] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2009] [Revised: 05/02/2009] [Accepted: 05/07/2009] [Indexed: 01/08/2023]
Abstract
Autoimmunity can be triggered by many environmental factors, among which infectious agents are pivotal. Here, we summarize current knowledge of the relationship between infection and autoimmunity. An autoimmune disease can be induced or triggered by infectious agents, which can also determine its clinical manifestations. Most infectious agents, such as viruses, bacteria and parasites, can induce autoimmunity via different mechanisms. In many cases, it is not a single infection but rather the 'burden of infections' from childhood that is responsible for the induction of autoimmunity. The development of an autoimmune disease after infection tends to occur in genetically susceptible individuals. By contrast, some infections can protect individuals from specific autoimmune diseases (i.e. the hygiene hypothesis).
Collapse
|
43
|
Ioannou Y, Romay-Penabad Z, Pericleous C, Giles I, Papalardo E, Vargas G, Shilagard T, Latchman DS, Isenberg DA, Rahman A, Pierangeli S. In vivo inhibition of antiphospholipid antibody-induced pathogenicity utilizing the antigenic target peptide domain I of beta2-glycoprotein I: proof of concept. J Thromb Haemost 2009; 7:833-42. [PMID: 19220729 DOI: 10.1111/j.1538-7836.2009.03316.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
SUMMARY OBJECTIVES In the antiphospholipid syndrome (APS), the immunodominant epitope for the majority of circulating pathogenic antiphospholipid antibodies (aPLs) is the N-terminal domain I (DI) of beta(2)-glycoprotein I. We have previously shown that recombinant DI inhibits the binding of aPLs in fluid phase to immobilized native antigen, and that this inhibition is greater with the DI(D8S/D9G) mutant and absent with the DI(R39S) mutant. Hence, we hypothesized that DI and DI(D8S/D9G) would inhibit aPL-induced pathogenicity in vivo. METHODS C57BL/6 mice (n = 5, each group) were injected with purified IgG derived from APS patients (IgG-APS, 500 microg) or IgG from normal healthy serum (IgG-NHS) and either recombinant DI, DI(R39S), DI(D8S/D9G), or an irrelevant control peptide (at 10-40 microg). Outcome variables measured were femoral vein thrombus dynamics in treated and control groups following standardized vessel injury, expression of vascular cell adhesion molecule-1 (VCAM-1) on the aortic endothelial surface, and tissue factor (TF) activity in murine macrophages. RESULTS IgG-APS significantly increased thrombus size as compared with IgG-NHS. The IgG-APS thrombus enhancement effect was abolished in mice pretreated with recombinant DI (P CONCLUSION Our findings in this proof-of-concept study support the development of recombinant DI or the novel variant DI(D8S/D9G) as a potential future therapeutic agent for APS.
Collapse
Affiliation(s)
- Y Ioannou
- Centre for Rheumatology Research, Windeyer Building, University College London, London, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Vlachoyiannopoulos PG, Samarkos M, Sikara M, Tsiligros P. Antiphospholipid antibodies: laboratory and pathogenetic aspects. Crit Rev Clin Lab Sci 2008; 44:271-338. [PMID: 17453920 DOI: 10.1080/10408360601079549] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Antiphospholipid antibodies (aPL) constitute a heterogeneous group of autoantibodies that share the ability to bind phospholipids (PL) alone, protein-PL complexes, or PL-binding proteins. They have been detected in isolation, in association with autoimmune diseases such as systemic lupus erythematosus (SLE), and during the course of different infections. aPL have been associated with an array of clinical manifestations in virtually every organ, although deep vein and arterial thrombosis as well as pregnancy morbidity are predominant. The co-occurrence of these clinical findings with aPL constitutes the so-called antiphospholipid syndrome (APS). aPL can be detected by immunological methods [e.g., anticardiolipin antibodies (aCL)] or by functional methods that exploit the effect of aPL on blood coagulation [lupus anticoagulant (LA)]. Since aPL are heterogeneous, numerous immunological and coagulation assays have been developed. These assays have not been fully standardized, and, therefore, problems such as high interlaboratory variation are relatively frequent. Recently, recommendations have been published regarding LA and aCL testing. Not all aPL are pathogenic. However, when they are not associated with infections, they have a role in the pathogenesis of APS. Clinical and experimental data have shown that aPL exert their pathogenic activity by interfering with the function of coagulation factors, such as thrombin and factors X, XI and XII, and with the function of anticoagulant proteins of the protein C system. In addition, aPL interaction with platelets and endothelial cells induces a pro-adhesive activated phenotype.
Collapse
|
45
|
Yasuda S, Bohgaki M, Atsumi T, Koike T. Pathogenesis of antiphospholipid antibodies: impairment of fibrinolysis and monocyte activation via the p38 mitogen-activated protein kinase pathway. Immunobiology 2005; 210:775-80. [PMID: 16325497 DOI: 10.1016/j.imbio.2005.10.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2005] [Accepted: 08/30/2005] [Indexed: 11/30/2022]
Abstract
Antiphospholipid syndrome (APS) is characterized by recurrent thrombosis or pregnancy morbidity associated with antiphospholipid antibodies (aPL). Impaired fibrinolysis is a contributing factor for the development of thrombosis, and the effect of aPL in the fibrinolytic system has been investigated. Impaired release of tPA and enhanced release of PAI-1 after endothelial activation is reported in patients with APS. Elevated Lipoprotein (a) levels have been found in APS, which results in inhibition of fibrinolytic activity. Phospholipid-bound beta(2)-glycoprotein I (beta(2)GPI) is a major autoantigen for aPLs. beta(2)GPI exerts both anti-coagulant and pro-coagulant properties mainly by interacting with other phospholipid-binding proteins such as coagulation factors and protein C. Dramatic increase in the affinity of beta(2)GPI to the cell surface is induced by binding of pathogenic anti-beta(2)GPI antibodies, which may modify the physiological function of beta(2)GPI and may affect the coagulation/fibrinolysis balance on the cell surface. Using chromogenic assays for measuring fibrinolytic activity, we demonstrated that addition of monoclonal anticardiolipin antibody (aCL) decreases the activity of extrinsic/intrinsic fibrinolysis. Significantly lower activity of intrinsic fibrinolysis was also demonstrated in the euglobulin fractions from APS patients. Endothelial cells and monocytes are activated by aPLs in vitro, resulting in production of tissue factor (TF), a major initiator of the coagulation system. Recently, aPLs are reported to induce thrombocytes to produce thromboxane. The importance of apoE receptor 2 on platelets for the binding of artificially dimerized beta(2)GPI was suggested. By investigating aPL-inducible genes in peripheral blood mononuclear cells, we found that the mitogen-activated protein kinase (MAPK) pathway was up-regulated. Using a monocyte cell line, phosphorylation of p38 MAPK, NF-kappaB translocation to the nuclear fraction, and up-regulated TF mRNA expression were demonstrated after treatment with monoclonal aCL. These phenomena were observed only in the presence of beta(2)GPI. Moreover, a specific p38 MAPK inihibitor SB203580 decreased aCL/beta(2)GPI-induced TF mRNA expression. Thus, aCL/beta(2)GPI plays dual roles in the pathogenesis of APS, firstly by deranging the fibrinolytic system and secondly by activating monocytes, endothelial cells and thrombocytes to produce TF or thromboxane.
Collapse
Affiliation(s)
- Shinsuke Yasuda
- Department of Medicine II, Hokkaido University School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8648, Japan.
| | | | | | | |
Collapse
|
46
|
Shapiro SS. Lupus anticoagulants and anticardiolipin antibodies: personal reminiscences, a little history, and some random thoughts. J Thromb Haemost 2005; 3:831-3. [PMID: 15869569 DOI: 10.1111/j.1538-7836.2005.01392.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- S S Shapiro
- Department of Physiology, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
47
|
Yasuda S, Atsumi T, Ieko M, Koike T. Beta2-glycoprotein I, anti-beta2-glycoprotein I, and fibrinolysis. Thromb Res 2005; 114:461-5. [PMID: 15507279 DOI: 10.1016/j.thromres.2004.07.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2004] [Revised: 07/25/2004] [Accepted: 07/29/2004] [Indexed: 11/24/2022]
Abstract
Beta2-glycoprotein-I (beta2GPI) is a phospholipid-binding plasma protein that consists of five homologous domains. Domain V is distinguished from others by bearing a positively charged lysine cluster and hydrophobic extra C-terminal loop. Beta2GPI has been known as a natural anticoagulant regulator. Beta2GPI exerts anticoagulant activity by inhibition of phospholipid-dependent coagulation reactions such as prothrombinase, tenase, and factor XII activation. It also binds factor XI and inhibits its activation. On the other hand, beta2GPI inhibits anticoagulant activity of activated protein C. According to the data from knockout mice, beta2GPI may contribute to thrombin generation in vivo. Phospholipid-bound beta2GPI is one of the major target antigens for antiphospholipid antibodies present in patients with antiphospholipid syndrome (APS). Binding of pathogenic anti-beta2GPI antibodies increases the affinity of beta2GPI to the cell surface and disrupts the coagulation/fibrinolysis balance on the cell surface. These pathogenic antibodies activate endothelial cells via signal transduction events in the presence of beta2GPI. Impaired fibrinolysis has been reported in patients with APS. Using a newly developed chromogenic assay, we demonstrated lower activity of intrinsic fibrinolysis in euglobulin fractions from APS patients. Addition of monoclonal anti-beta2GPI antibodies with beta2GPI also decreased fibrinolytic activity in this assay system. beta2GPI is proteolytically cleaved by plasmin in domain V (nicked beta2GPI) and becomes unable to bind to phospholipids, reducing antigenicity against antiphospholipid antibodies. This cleavage occurs in patients with increased fibrinolysis turnover. Nicked beta2GPI binds to plasminogen and suppresses plasmin generation in the presence of fibrin, plasminogen, and tissue plasminogen activator (tPA). Thus, nicked beta2GPI plays a role in the extrinsic fibrinolysis via a negative feedback pathway loop.
Collapse
Affiliation(s)
- Shinsuke Yasuda
- Department of Medicine II, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo 060-8638, Japan.
| | | | | | | |
Collapse
|
48
|
Abstract
We herein review evidence that the phospholipid-binding protein beta2 glycoprotein-1 (beta2GPI) is a causative autoantigen in APS. Recent work suggests that the molecular regions in beta2GPI that facilitate autoimmunization are those that promote binding to negatively charged phospholipids by means of strong positive (anionic) charge and hydrophobicity. Although many common infections can cause antiphospholipid antibodies to be produced in humans, such postinfectious aPL are rarely associated with thromboses or pregnancy morbidity, the central features of antiphospholipid syndrome (APS). We propose that the causes of APS include those infectious agents that mimic the above molecular domains in beta2GPI. In people who are susceptible to APS, tolerance to self-beta2GPI and phospholipids is likely to be broken by foreign bacterial or viral proteins that contain such beta2GPI-like epitopes.
Collapse
Affiliation(s)
- A E Gharavi
- Department of Medicine, Morehouse School of Medicine, Atlanta, GA 30310-1495, USA.
| | | | | |
Collapse
|
49
|
Yasuda S. [Pathogenic roles of anti-beta2-GPI antibody in patients with antiphospholipid syndrome]. NIHON RINSHO MEN'EKI GAKKAI KAISHI = JAPANESE JOURNAL OF CLINICAL IMMUNOLOGY 2004; 27:373-8. [PMID: 15678890 DOI: 10.2177/jsci.27.373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Antiphospholipid syndrome (APS) is characterized by arterial and/or venous thrombosis and pregnancy morbidity in the presence of antiphospholipid antibodies (aPL). Beta2-glycoprotein I (beta2-GPI) and prothrombin are representative autoantigens, the former more extensively investigated. Anti-beta2-GPI antibodies are not only markers of APS, but also are considered to be pathogenic. Possible roles of anti-beta2-GPI antibodies are, 1) enhancement the binding of beta2-GPI to anionic phospholipid and inhibition of protein C activation/activated protein C, 2) to form anti-beta2-GPI antibody-beta2-GPI-oxidized LDL complex and to promote uptake by sub-endothelial macrophage, resulting in atherosclerosis, 3) to dimerize beta2-GPI on the surface of platelets and to activate platelets via apoE receptor 2 and subsequent signal transduction, 4) stimulation of monocytes via p38 MAP kinase pathway and induction of tissue factor production. In pregnancy morbidity, activation of complement cascade plays an important role. These findings may provide a novel target in the management of APS.
Collapse
Affiliation(s)
- Shinsuke Yasuda
- Department of Medicine II, Hokkaido University Graduate School of Medicine
| |
Collapse
|
50
|
Ferrara DE, Swerlick R, Casper K, Meroni PL, Vega-Ostertag ME, Harris EN, Pierangeli SS. Fluvastatin inhibits up-regulation of tissue factor expression by antiphospholipid antibodies on endothelial cells. J Thromb Haemost 2004; 2:1558-63. [PMID: 15333031 DOI: 10.1111/j.1538-7836.2004.00896.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Mechanisms of thrombosis induced by antiphospholipid (aPL) antibodies include up-regulation of tissue factor (TF) expression on endothelial cells (ECs). Statins have been shown to reduce levels of TF induced by tumor necrosis factor (TNF-alpha) and lipopolysaccharide (LPS) on ECs. In a recent study, fluvastatin inhibited thrombogenic and proinflammatory properties of aPL antibodies in in vivo models. The aim of this study was to determine whether fluvastatin has an effect on aPL-induced expression of TF on ECs. METHODS IgGs were purified from four patients with APS (IgG-APS) and from control sera (IgG-NHS). Cultured human umbilical vein endothelial cells (HUVEC) were treated with IgG-APS or IgG-NHS or with medium alone or with phorbol myristate acetate (PMA), as a positive control. In some experiments, cells were pretreated with fluvastatin (2.5, 5 or 10 micro m) with and without mevalonate (100 micro m). TF expression on HUVECs was measured by ELISA. RESULTS PMA and the four IgG-APS preparations increased the expression of TF on EC significantly (4.9-, 2.4-, 4.2-, 3.5- and 3.1-fold, respectively), in a dose-dependent fashion. Fluvastatin (10 micro m) inhibited the effects of PMA and the four IgG-APS on TF expression by 70, 47, 65, 22 and 68%, respectively, and this effect was dose-dependent. Mevalonate (100 micro m) completely abrogated the inhibitory effects of fluvastatin on TF expression induced by aPL. CONCLUSION Because of the suggested pathogenic role of aPL on induction of TF on ECs, our data provide a rationale for using statins as a therapeutic tool in treatment of thrombosis in APS.
Collapse
Affiliation(s)
- D E Ferrara
- Antiphospholipid Standardization Laboratory, Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310-1495, USA
| | | | | | | | | | | | | |
Collapse
|