ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Pathogenesis of antiphospholipid syndrome

Pathogenesis of antiphospholipid syndrome
Literature review current through: Jan 2024.
This topic last updated: Jan 02, 2024.

INTRODUCTION — Antiphospholipid syndrome (APS) is a systemic autoimmune disorder characterized by recurrent thrombosis and obstetric morbidity. Sometimes other manifestations such as thrombocytopenia and cardiac valve disease are present. Beyond clinical manifestations, classification criteria for APS require the persistent presence of antiphospholipid antibodies (aPL) as measured by at least one of three tests [1]:

Lupus anticoagulant (LA) testing using a method that detects aPL interference in phospholipid-dependent functional clotting assays such as the activated partial thromboplastin time (aPTT) or dilute Russell viper venom time.

An immunoassay for anti-beta2 glycoprotein I (anti-beta2GPI) antibodies (immunoglobulin [Ig]G or IgM).

An immunoassay for anticardiolipin (aCL) antibodies (IgG or IgM).

APS-related thrombosis may affect vascular beds of all sizes including both arterial and venous circuits. The deep veins of the lower extremities and the cerebral circulation are the most common sites of venous and arterial thrombosis, respectively [2]. Thrombosis may also occur in unusual sites including visceral arteries and veins and the cerebral venous sinuses.

In a minority of patients, microvascular thrombosis affects microvessels in the kidney, skin, lungs, heart, eye, and other organs. Approximately 1 percent of patients develop catastrophic APS (CAPS) [3,4]. (See "Catastrophic antiphospholipid syndrome (CAPS)", section on 'Definitions'.)

Obstetric complications of APS include recurrent early pregnancy losses, fetal demise beyond 10 weeks, and premature births associated with preeclampsia or placental insufficiency [1]. Other clinical findings such as thrombocytopenia, livedo reticularis/racemosa, skin ulcers, mitral and aortic valve damage, transient ischemic attacks, seizures, and premature cognitive decline may also occur in patients with APS, but are considered "non-criteria" manifestations [2].

It is generally accepted that aPL, particularly anti-beta2GPI antibodies, are central to the pathogenesis of APS. Multiple mechanisms by which aPL may induce thrombosis or obstetric morbidity have been identified and are discussed in this topic review. However, the extent to which the various distinct manifestations of APS are reflective of antibody heterogeneity or can be explained by a common central pathway remains unresolved.

For information on diagnosis, clinical manifestations, and treatment of APS and CAPS, as well as APS in pregnancy, refer to separate topic reviews:

Diagnosis – (See "Diagnosis of antiphospholipid syndrome".)

Clinical features – (See "Clinical manifestations of antiphospholipid syndrome".)

Kidney manifestations – (See "Antiphospholipid syndrome and the kidney".)

Management – (See "Management of antiphospholipid syndrome".)

CAPS diagnosis and management – (See "Catastrophic antiphospholipid syndrome (CAPS)".)

Pregnancy – (See "Antiphospholipid syndrome: Obstetric implications and management in pregnancy".)

ROLE OF ANTIPHOSPHOLIPID ANTIBODIES — Antiphospholipid antibodies (aPL) were originally thought to bind anionic phospholipids such as cardiolipin and phosphatidylserine (PS). However, it was subsequently recognized that the term "antiphospholipid" is a misnomer, since the best-characterized aPL are actually directed against specific phospholipid-binding proteins [5]. The most prominent of these proteins is beta2 glycoprotein I (beta2GPI). Anti-beta2GPI antibodies appear to be central to the pathogenesis of antiphospholipid syndrome (APS) [6,7]; however, other antigenic targets such as prothrombin (PT), PS/PT complexes, vimentin/cardiolipin complexes, annexin A2, and annexin A5 have been described [8-12].

Lupus anticoagulant — The so-called "lupus anticoagulant" (LA) effect is a functional property of a heterogenous group of antibodies, with its name derived from its original description in patients with systemic lupus erythematosus (SLE). Antibodies with LA activity interfere with clotting in in vitro assays and prolong phospholipid-dependent clotting times such as the activated partial thromboplastin time (aPTT). The first two patients with this finding also had a hemorrhagic diathesis [13]. This finding led to the initial mistaken impression that the antibodies had an anticoagulant effect, when in fact they are most commonly prothrombotic. LA activity is strongly associated with thrombosis and pregnancy loss [14-16]. (See "Clinical use of coagulation tests", section on 'Lupus anticoagulant tests'.)

Antibodies against beta2GPI or PT are the most common cause of an LA effect [17]. Overall, LA associated with anti-beta2GPI antibodies appears to confer greater thrombotic risk than does beta2GPI-independent LA (odds ratio [OR] 42.3, 95% CI 9.9-194.3, versus 1.6, 95% CI 0.8-3.9) [18]. Furthermore, the risk of thrombosis increases with the number of positive tests for aPL. In particular, patients with positive tests for LA, anticardiolipin (aCL) antibodies, and anti-beta2GPI antibodies (referred to as "triple positive" APS) have a higher risk of a first or recurrent thrombotic event than patients having only one or two positive tests [19-22]. (See "Management of antiphospholipid syndrome", section on 'Risk of a first thrombosis with aPL'.)

Anti-beta2 glycoprotein I antibodies — Beta2GPI is the best-defined antigenic target of pathogenic aPL. Anti-beta2GPI antibodies are associated with both thrombosis and pregnancy loss in individuals with APS [6,15]. Affinity-purified anti-beta2GPI antibodies also potentiate thrombosis in mice [23].

Beta2GPI is a circulating glycoprotein. The crystal structure has demonstrated a series of five "sushi" domains arranged like beads on a string in a "J"-shaped molecule that includes four complement control domains followed by a positively charged fifth domain that associates with phospholipids [24].

These beta2GPI domains exist in at least two configurations [25]:

Closed configuration – A closed/circular form circulates in plasma. In this form, domain 5 associates with domain 1, essentially hiding domain 1 from autoantibodies.

Open configuration – An open conformation is created when domain 5 binds to anionic phospholipids on the surface of various cells (endothelial cells, monocytes, platelets). This "opens" the molecule into a linear string, exposing domain 1 and allowing the binding of the aPL autoantibodies.

The presence of the closed configuration of beta2GPI that cannot be bound by autoantibodies in the circulation may explain the absence of circulating beta2GPI-containing immune complexes in patients with APS. Antibodies directed against beta2GPI appear to recognize beta2GPI only when it binds to anionic surfaces and assumes the open conformation, although the mechanisms underlying this conformational change are poorly understood [26].

Subsequent studies have suggested that domain 1-specific beta2GPI antibodies are the aPL most likely to cause thrombosis. IgG antibodies against the Gly40-Arg43 epitope in domain 1 (referred to as anti-beta2GPI-domain 1 antibodies) often cause the LA effect. (See 'Lupus anticoagulant' above.)

Anti-beta2GPI-domain 1 antibodies are more strongly associated with both thrombosis and obstetric morbidity than are antibodies directed against other regions of beta2GPI [27,28]. Anti-beta2GPI-domain 1 antibodies are also more likely to persist at 12 weeks [29] and to associate with triple positivity [30].

The clinical significance of IgA isotypes of anti-beta2GPI and aCL remains under investigation, and these antibodies are not included in the classification criteria for APS (see "Diagnosis of antiphospholipid syndrome", section on 'Antiphospholipid antibody testing' and "Diagnosis of antiphospholipid syndrome", section on 'Classification criteria'). IgA antibodies from patients with APS have been shown to induce thrombosis in a mouse model [31]. IgA anti-beta2GPI have also been associated with thrombotic events, particularly in individuals with SLE [32,33].

Non-criteria autoantibodies — Antibodies against other targets have been identified in patients with APS. These antibodies are not included in the classification criteria for APS (unless they act as an LA), and their role in APS pathogenesis is under study. (See "Diagnosis of antiphospholipid syndrome", section on 'Classification criteria'.)

Other phospholipid-binding proteins – aPL directed against a variety of other phospholipid-binding proteins and phospholipid/protein complexes have been detected in sera of patients with APS. These proteins include:

PT and PS/PT complexes [34-36]

Phosphatidylethanolamine (PE) [37]

Annexin A2 [8,9]

Annexin A5 [10]

Vimentin/cardiolipin complexes [11]

Endosomal lysobisphosphatidic acid (LBPA) associated with the endothelial receptor for protein C (endothelial protein C receptor [EPCR]) [38]

Some of these antibodies, particularly anti-PS/PT, have been associated with thrombosis and LA activity [39,40]. However, the clinical significance of most non-criteria aPL remains uncertain. Rigorous mechanistic studies are lacking.

Neutrophil extracellular traps – Neutrophil extracellular traps (NETs) are structures released from dying neutrophils that have antimicrobial properties. (See "An overview of the innate immune system", section on 'Neutrophil extracellular traps'.)

Anti-NET antibodies have been identified at higher levels in patients with aPLs and APS when compared with healthy controls [41]. In a cohort of patients with persistently elevated aPLs and/or APS, positive testing for anti-NET IgG was associated with white matter lesions in the brain, even after adjusting for other aPLs (odds ratio 11, 95% CI 1.9-62) [41]. The level of anti-NET IgM was inversely correlated with levels of complement components C3 and C4, and serum with high anti-NET IgM levels had higher C3d deposition on NETs compared with serum from controls, suggesting that anti-NET IgM may activate the complement cascade. (See 'Activation of vascular and immune cells' below and 'Complement activation' below.)

MECHANISMS OF THROMBOTIC ANTIPHOSPHOLIPID SYNDROME — The earliest identified prothrombotic effects of antiphospholipid antibodies (aPL) were inhibition of natural anticoagulant systems [42], along with other direct procoagulant and antifibrinolytic properties [43]. However, subsequent studies indicate that aPL-induced activation of several types of cells, resulting in multiple procoagulant and proinflammatory effects, and activation of complement are the major drivers of the vascular and obstetric complications of antiphospholipid syndrome (APS). The major pathogenic mechanisms are summarized in a table (table 1) and discussed further below.

Activation of vascular and immune cells — Activation of vascular and immune cells by aPL potentiates thrombosis through upregulation of surface adhesion molecules and release of proinflammatory cytokines and procoagulant substances including extracellular vesicles and other cellular remnants.

Cells that are activated in response to aPL include endothelial cells, monocytes, neutrophils, and platelets. The relative importance of each cell type in promoting thrombosis likely depends on the specific vascular bed.

Endothelial cells – aPL, particularly anti-beta2 glycoprotein I (anti-beta2GPI) antibodies, activate endothelial cells in vitro, leading to expression of tissue factor and adhesion molecules [44,45]. The antibodies simultaneously downregulate the expression of vasoprotective endothelial nitric oxide synthase (eNOS) [46].

Anti-beta2GPI antibodies recognize beta2GPI complexed with coreceptors such as annexin A2 [47] and apoER2 [46] to activate intracellular signaling through toll-like receptor (TLR)-like and Krüppel-like factor (KLF) transcription factor-mediated pathways [48-50]. In mice, blocking E-selectin, P-selectin, and endothelial integrin ligands (vascular cell adhesion molecule 1 [VCAM-1] and intercellular adhesion molecule 1 [ICAM-1]) protects against aPL-mediated thrombosis [51,52].

Monocytes – In vitro, aPL activate monocytes and cause them to express tissue factor [53-56] and other proinflammatory cytokines such as tumor necrosis factor (TNF)-alpha and interleukin (IL) 1 beta [57-59]. Monocyte tissue factor expression is increased in patients with systemic lupus erythematosus (SLE) and aPL [60] and in patients with primary thrombotic APS [61-63].

Monocytes from patients with APS also express higher levels of the proangiogenic cytokine vascular endothelial growth factor (VEGF) and its receptor Flt-1 [64], as well as proinflammatory mediators such as TLR8, CD14, and proteins associated with oxidative stress [65,66]. Several studies demonstrate increased levels of monocyte-derived microparticles in patients with APS [67,68], which may be an important source of tissue factor [69].

Neutrophils – Neutrophils contribute to thrombosis in arterial [70,71], venous [72,73], and microvascular beds [74,75]. aPL, particularly anti-beta2GPI antibodies [76-79], induce neutrophil tissue factor expression (similar to monocytes) [78] and the release of prothrombotic neutrophil extracellular traps (NETs) [79]. NETs are elongated extracellular "spiderwebs" composed of neutrophil-derived chromatin and microbicidal proteins; they can bind bacteria and other organisms, and they have a variety of immune functions [80,81]. (See "An overview of the innate immune system", section on 'Neutrophils'.)

NETs circulate at elevated levels in patients with APS [79]. NETs activate platelets and endothelial cells and ultimately form an integral part of venous [72,73,82,83] and arterial clots [70,84,85]. In mice, aPL-mediated large vein thrombosis is dependent on NETs [86,87] and neutrophil adhesion to the vessel wall [88,89]. Anti-NET antibodies are also elevated in patients with APS, and anti-NET IgM is inversely correlated with levels of complements C3 and C4, suggesting that anti-NET antibodies may activate the complement cascade [41]. (See 'Non-criteria autoantibodies' above and 'Complement activation' below.)

Platelets – While beta2GPI and anti-beta2GPI antibodies do not appear to bind to unstimulated platelets, under shear stress, anti-beta2GPI antibodies may trigger platelet activation through binding of beta2GPI to surface ApoER2 and GPIb receptors [90]. aPL also activate platelets in the presence of low levels of thrombin through a mitogen-activated protein kinase (MAP kinase)-dependent pathway [91]. In mouse models of APS, aPL-activated platelets may be preferentially required for fibrin generation in the expanding thrombus [92]. Increased platelet-leukocyte aggregates are detected in the blood of patients with APS, consistent with low-grade platelet activation [93]. The extent to which thrombocytopenia in APS is attributable to platelet activation (and subsequent removal) or to clearance via antiplatelet antibodies likely varies from patient to patient [94-96].

Interactions with coagulation and fibrinolytic systems — aPL appear to interfere with the activity of natural anticoagulant proteins including proteins S and C, as well as with the fibrinolytic system, which breaks down fibrin. (See "Overview of hemostasis", section on 'Control mechanisms and termination of clotting'.)

Protein C – Inhibition of anticoagulant activity in vitro was one of the earliest identified prothrombotic mechanisms in APS. aPL inhibit the activation of protein C and the ability of activated protein C to inactivate factors V and VIII (ie, activated protein C resistance) [42,97-100].

Antithrombin activity – aPL also reduce antithrombin (AT) activity by inhibiting the heparin binding required for full activation of AT [101]. aPL with activity against thrombin may further reduce the inactivation of thrombin by AT [102], while antibodies against activated factors X and IX also interfere with negative regulation by AT [103,104].

Fibrinolysis – aPL may inhibit fibrinolysis by neutralizing the ability of beta2GPI to stimulate tissue plasminogen activator (tPA)-mediated plasminogen activation and fibrinolysis [105]. In addition, inhibitory antibodies against tPA and other components of the fibrinolytic system are reported in patients with APS [106,107].

Interactions with procoagulant pathways – Tissue factor activity may be potentiated in APS via inhibition of tissue factor pathway inhibitor (TFPI) in patients with a positive lupus anticoagulant (LA) and/or anti-beta2GPI antibodies [108,109]. Anti-beta2GPI antibodies have also been reported to impair the ability of beta2GPI to inhibit von Willebrand factor (VWF)-dependent platelet aggregation [110]. Elevated levels of factor XI have been identified as a risk factor for thrombosis in the general population [111]. Patients with APS have higher levels of the active free thiol form of factor XI than age- and sex-matched controls [112].

Annexin A5 – Annexin A5 binds to phosphatidylserine (PS) to form a two-dimensional lattice, or "anticoagulant shield," over exposed phospholipids on cell surfaces [113]. The complex of beta2GPI and anti-beta2GPI disrupts this protective shield, exposing procoagulant PS and promoting thrombosis [114,115]. Interestingly, the antimalarial drug hydroxychloroquine has been reported to stabilize the annexin V shield in vitro [116,117] and has shown efficacy against aPL-mediated thrombosis in a murine model [118].

Complement activation — Complement consists of a system of over 50 proteins involved in innate immunity that link the inflammatory response to coagulation pathways [119]. In mice, thrombotic APS can be modeled by direct vessel injury or infusion of lipopolysaccharide followed by passive transfer of patient-derived aPL. In these models, aPL-augmented thrombosis is attenuated in the presence of complement inhibitors or by gene knockout to prevent expression of complement proteins such as C3, C5, and C6 [120-122]. The C5b-9 protein complex, also called the membrane attack complex (MAC), creates transmembrane channels (holes) in cellular membranes. (See "Complement pathways", section on 'Attack (the membrane attack complex)'.)

In clinical studies, higher levels of C5b-9 are reported in patients with APS and stroke [123]. Many patients with APS have hypocomplementemia (suggestive of complement consumption) [124] and/or elevated levels of complement activation products Bb and C3a [125]. A study using sera and purified anti-beta2GPI antibodies from patients with APS demonstrated that these could cause enhanced complement-mediated cell death via C5b-9 deposition on the cell surface [126]. The assay, referred to as the modified Ham test, measures complement-mediated cell lysis. Complement activation in the modified Ham test correlates with triple positivity (presence of three types of aPL, a marker of severe disease) and recurrent thrombosis.

Anecdotal reports of the efficacy of the terminal complement inhibitor eculizumab in treating refractory thrombosis in APS and catastrophic APS (CAPS) also support a role of complement in aPL-mediated thrombosis [127,128]. (See "Catastrophic antiphospholipid syndrome (CAPS)", section on 'Eculizumab'.)

Additional information about the role of complement in CAPS is presented separately. (See "Catastrophic antiphospholipid syndrome (CAPS)", section on 'Pathophysiology/mechanism of thrombosis'.)

Triggers of thrombosis — In animal models of APS, passive infusion of aPL does not cause thrombosis in the absence of mechanical or chemical vessel wall injury, disruption of blood flow, or infusion of an immune stimulant such as lipopolysaccharide. The "two-hit" model of APS proposes that aPL provide the first hit that induces a generalized procoagulant state by activating vascular cells and/or causing prothrombotic alterations in the coagulation and fibrinolytic systems. Subsequently, a second hit (often cryptic) in the form of a vascular injury or inflammatory stimulus tips the system toward thrombosis.

Although the precise initiating stimulus is not readily apparent in many cases of thrombotic APS, a precipitating factor such as infection, surgery, or pregnancy is identified most individuals with CAPS. (See "Catastrophic antiphospholipid syndrome (CAPS)", section on 'Additional risk factors'.)

Additional risk factors — The presence of additional risk factors for thrombosis may further increase thrombotic risk in patients with aPL [129-131]. These factors include:

SLE

Inherited thrombophilia

Cancer

Smoking

Pregnancy

Use of estrogen-containing oral contraceptives

Immobilization

Obesity

Hypertension

Hypercholesterolemia

In a large population-based case-control study, the odds ratio (OR) of ischemic stroke in females with LA was 43.1 (95% CI 12.2-152.0), increasing to 87 (95% CI 14.5-523.0) in those who smoked and to 201 (95% CI 1.9-242) in those who used estrogen-containing oral contraceptives [132].

In the same study, the risk of myocardial infarction increased from 5.3 (95% CI 1.4-20.8) to 33.7 (95% CI 6.0-189.0) in females who smoked and to 21.6 (95% CI 1.9-242.0) in those who used estrogen-containing oral contraceptives [132].

In another cohort of 122 patients with aPL, hypertriglyceridemia was associated with a 6.4-fold increase in the risk of thrombosis, hereditary thrombophilia with a 7.3-fold increase, and anticardiolipin (aCL) IgG with a titer >40 IgG phospholipid units (GPL) with a 2.8-fold increase [129].

MECHANISMS OF OBSTETRIC ANTIPHOSPHOLIPID SYNDROME — Historically, impairment of fetal-maternal exchange caused by intervillous thrombosis was assumed to be the main driver of pregnancy morbidity in antiphospholipid syndrome (APS). However, thrombosis is not reliably detected in the placentae of antiphospholipid antibody (aPL)-positive pregnancies [133]. Rather, clinical and experimental observations suggest that obstetric complications in APS are mediated primarily by trophoblast dysfunction and complement activation [134].

Early versus late-term pregnancy loss — Although related, the pathogeneses of first-trimester fetal loss and late pregnancy loss associated with APS are distinct:

Early pregnancy loss – First-trimester losses are typically attributed to direct inhibitory effects of aPL on trophoblast viability, proliferation, and invasion [135]. These derangements prevent proper anchorage of the placenta to maternal decidua, resulting in first-trimester miscarriage.

Late-term pregnancy loss – Late-term obstetric complications of APS, such as fetal growth restriction, preeclampsia, preterm birth, and fetal demise, are likely a consequence of both trophoblast dysfunction (leading to an inadequately developed placenta), as well as direct inflammatory effects mediated by aPL and complement.

Trophoblast dysfunction — Trophoblasts constitutively express beta2 glycoprotein I (beta2GPI) on their surface [136-139]. In vitro studies of the effects of aPL (and especially anti-beta2GPI) on trophoblast function have revealed defects in several normal processes:

Proliferation [140]

Invasiveness [138]

Secretion of human chorionic gonadotropin [138]

Production of angiogenic factors such as vascular endothelial growth factor (VEGF) [141]

Syncytialization (fusion) [142].

At the same time, studies using trophoblast cell lines demonstrate that aPL can cause the cells to take on a proinflammatory phenotype including activation of the NALP3 inflammasome [143] and interleukin (IL) 8 secretion [144].

These impaired cellular functions cause inadequate remodeling of uterine spiral arteries, thereby reducing maternal blood flow to the placenta and inducing hypoxic damage and reperfusion injury. A lack of well-developed vasculosyncytial membranes in the placenta of aPL-positive patients can further limit gas and nutrient exchange in the third trimester [133].

Complement activation and downstream mediators — As pregnancy progresses, it is hypothesized that recruitment of aPL to the placenta may trigger Fc-dependent inflammatory responses (activation of complement is the best characterized).

In murine "passive transfer" models, polyclonal IgG from patients with APS with high titers of aPL are administered to pregnant mice, resulting in fetal resorption and growth restriction. The seminal observations from these models were that mice deficient in complement component C3 were protected from fetal injury, and in wild-type mice, administration of C3 convertase inhibitors was protective [145].

Other mouse studies have suggested that downstream of complement, neutrophils are activated in C5a receptor-dependent fashion [146] with fetal damage further amplified by both tumor necrosis factor (TNF)-alpha [147] and tissue factor [148].

A study in a cohort of pregnant patients (487 with aPL or systemic lupus erythematosus [SLE] and 204 controls) found that circulating markers of complement activation (Bb and soluble C5b-9) were higher in the individuals with aPL or SLE, and adverse pregnancy outcomes correlated with higher levels of these circulating markers [149].

Notably, the efficacy of heparin in reducing pregnancy loss in individuals with APS is thought to be at least in part due to its ability to inhibit complement activation in the placenta [150].

PATHOGENESIS OF OTHER CLINICAL PRESENTATIONS

Catastrophic antiphospholipid syndrome — (See "Catastrophic antiphospholipid syndrome (CAPS)", section on 'Pathophysiology/mechanism of thrombosis'.)

Antiphospholipid syndrome vasculopathy — In addition to thrombosis, patients with APS may also develop chronic proliferative vascular lesions that contribute to progressive organ impairment [151]. As examples, small arteries in the kidney, leptomeninges, and lungs may show concentric cellular and fibrous intimal hyperplasia (neointima formation) reminiscent of hypertensive vascular disease, without evidence of thrombotic microangiopathy [151].

A study implicated the mammalian/mechanistic target of rapamycin (mTOR, a kinase that integrates a variety of signaling pathways to regulate cellular growth, proliferation, and survival) in the chronic proliferative vasculopathy of APS [152]. In individuals with aPL-associated nephropathy, the vascular endothelium of intrarenal vessels was found to display molecular markers consistent with activation of mTOR and downstream signaling [153]. Furthermore, patients with aPL-associated nephropathy who required transplantation and were receiving sirolimus (an mTOR inhibitor) had minimal recurrence of vascular lesions, which was in contrast to matched patients with aPL who were not receiving sirolimus [153].

COVID-19 AND ANTIPHOSPHOLIPID ANTIBODIES — Similar to antiphospholipid syndrome (APS), coronavirus disease 2019 (COVID-19) is associated with a higher-than-expected incidence of thrombosis in arterial, microcirculatory, and venous vascular beds [154,155]. Mechanistic studies of COVID-19-associated thrombosis demonstrate some similarities with the pathophysiology of APS, including evidence for hyperactivation of neutrophils [156], endothelial cells [157], platelets [158], and the complement system [159]. (See "COVID-19: Hypercoagulability", section on 'Pathogenesis'.)

Reports from early in the pandemic found antiphospholipid antibodies (aPL) at high titers in a small number of patients with COVID-19 who experienced macrovascular thrombotic events [160]. In a subsequent series of 56 patients hospitalized with COVID-19, lupus anticoagulant (LA) was detected in 25 (44.6 percent), 5 of whom (8.9 percent) also had either anticardiolipin (aCL) or anti-beta2 glycoprotein I (anti-beta2GPI) antibodies [161].

Studies in COVID-19 patients have detected both traditional aPL (such as IgG and IgM isotypes of aCL) and various "non-criteria" aPL (including IgG and IgM isotypes of anti-phosphatidylserine/prothrombin [anti-PS/PT] as well as IgA isotypes of aCL and anti-beta2GPI). There is significant heterogeneity across studies in terms of both overall prevalence of aPL (some as high as 50 percent) and which particular aPL species are most commonly detected [162-164]. It is not known whether these are transient aPL, as have been reported in viral infections previously [165], or persistent aPL more likely to be associated with long-term thrombotic risk. (See "Diagnosis of antiphospholipid syndrome", section on 'Transient antiphospholipid antibodies'.)

Most studies have not found a clear association of aPL with macrovascular thrombotic events in COVID-19. Furthermore, functional assays such as LA must be interpreted with caution in critically ill patients due to potential confounding by high levels of C-reactive protein, an acute phase reactant that can also prolong in vitro clotting times and lead to false-positive tests [166].

Despite these caveats, the relationship between aPL and COVID-19 is an emerging area deserving of further research. There is some evidence that IgG fractions isolated from the serum of patients with COVID-19 with high titers of aPL have prothrombotic properties in vitro and in mice [164]. Future studies are required to determine persistence of these antibodies and identify mechanistic connections that can further clarify whether aPL in patients with COVID-19 are similar to aPL seen in patients with APS.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Antiphospholipid syndrome".)

SUMMARY

Role of antiphospholipid antibodies – Antiphospholipid antibodies (aPL), particularly anti-beta2 glycoprotein I (anti-beta2GPI) antibodies, are diverse autoantibodies central to the pathogenesis of antiphospholipid syndrome (APS). aPL directed against other antigenic targets such as prothrombin (PT), phosphatidylserine/prothrombin (PS/PT) complexes, vimentin/cardiolipin complexes, annexin A2, annexin A5, and neutrophil extracellular traps (NETs) have been detected in the sera of patients with APS. Their roles in pathogenesis are under study. (See 'Role of antiphospholipid antibodies' above.)

Mechanisms of thrombosis – aPL-induced cellular activation resulting in multiple procoagulant and proinflammatory effects, as well as activation of complement, are the major drivers of the thrombotic complications of APS (table 1). (See 'Mechanisms of thrombotic antiphospholipid syndrome' above.)

The presence of additional risk factors such as systemic lupus erythematosus (SLE), inherited thrombophilia, cancer, smoking, pregnancy, and the use of estrogen-containing oral contraceptives further increases thrombotic risk in patients with aPL. (See 'Additional risk factors' above.)

Mechanisms of obstetric APS – Clinical and experimental observations suggest that the pathophysiology of obstetric complications in APS is driven by trophoblast dysfunction and complement activation. (See 'Mechanisms of obstetric antiphospholipid syndrome' above.)

Although related, the pathogenesis of first-trimester fetal loss and late pregnancy morbidity associated with aPL are distinct:

Early pregnancy loss – First-trimester losses are typically attributed to direct inhibitory effects of aPL on trophoblast viability, proliferation, and invasion. These derangements prevent proper anchorage of the placenta to maternal decidua, resulting in first-trimester miscarriage.

Late-term pregnancy loss – Late-term obstetric complications of APS, such as fetal growth restriction, preeclampsia, preterm birth, and fetal demise, are likely a consequence of both trophoblast dysfunction (leading to an inadequately developed placenta), as well as direct inflammatory effects mediated by aPL and complement.

Other clinical presentations – Progress has also been made in understanding the drivers of some other less common manifestations of APS:

Catastrophic antiphospholipid syndrome (CAPS) – (See 'Catastrophic antiphospholipid syndrome' above.)

Antiphospholipid vasculopathy – In addition to thrombosis, patients with APS may also demonstrate chronic proliferative vascular lesions that contribute to progressive organ impairment. (See 'Antiphospholipid syndrome vasculopathy' above.)

COVID-19 and antiphospholipid antibodies (aPL) – Most studies have not found a clear association of aPL with macrovascular thrombotic events in patients with coronavirus disease 2019 (COVID-19). Functional assays such as lupus anticoagulant (LA) must be interpreted with caution in critically ill patients because of potential confounding by transient aPL positivity due to infection and by high levels of C-reactive protein, which can cause the LA phenomenon. Future mechanistic studies will be most illuminating if they define the specificity and function of aPL in COVID-19 infection in comparison to those in traditional APS. (See 'COVID-19 and antiphospholipid antibodies' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Bonnie Bermas, MD, who contributed to an earlier version of this topic review.

  1. Miyakis S, Lockshin MD, Atsumi T, et al. International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS). J Thromb Haemost 2006; 4:295.
  2. Cervera R, Serrano R, Pons-Estel GJ, et al. Morbidity and mortality in the antiphospholipid syndrome during a 10-year period: a multicentre prospective study of 1000 patients. Ann Rheum Dis 2015; 74:1011.
  3. Cervera R, Espinosa G. Update on the catastrophic antiphospholipid syndrome and the "CAPS Registry". Semin Thromb Hemost 2012; 38:333.
  4. Asherson RA, Cervera R, de Groot PG, et al. Catastrophic antiphospholipid syndrome: international consensus statement on classification criteria and treatment guidelines. Lupus 2003; 12:530.
  5. Bevers EM, Galli M, Barbui T, et al. Lupus anticoagulant IgG's (LA) are not directed to phospholipids only, but to a complex of lipid-bound human prothrombin. Thromb Haemost 1991; 66:629.
  6. Galli M, Borrelli G, Jacobsen EM, et al. Clinical significance of different antiphospholipid antibodies in the WAPS (warfarin in the antiphospholipid syndrome) study. Blood 2007; 110:1178.
  7. McNeil HP, Simpson RJ, Chesterman CN, Krilis SA. Anti-phospholipid antibodies are directed against a complex antigen that includes a lipid-binding inhibitor of coagulation: beta 2-glycoprotein I (apolipoprotein H). Proc Natl Acad Sci U S A 1990; 87:4120.
  8. Salle V, Mazière JC, Smail A, et al. Anti-annexin II antibodies in systemic autoimmune diseases and antiphospholipid syndrome. J Clin Immunol 2008; 28:291.
  9. Cesarman-Maus G, Ríos-Luna NP, Deora AB, et al. Autoantibodies against the fibrinolytic receptor, annexin 2, in antiphospholipid syndrome. Blood 2006; 107:4375.
  10. Bećarević M. The IgG and IgM isotypes of anti-annexin A5 antibodies: relevance for primary antiphospholipid syndrome. J Thromb Thrombolysis 2016; 42:552.
  11. Ortona E, Capozzi A, Colasanti T, et al. Vimentin/cardiolipin complex as a new antigenic target of the antiphospholipid syndrome. Blood 2010; 116:2960.
  12. Žigon P, Čučnik S, Ambrožič A, et al. Antibodies to phosphatidylserine/prothrombin complex as an additional diagnostic marker of APS? Lupus 2012; 21:790.
  13. Jayakody Arachchillage D, Greaves M. The chequered history of the antiphospholipid syndrome. Br J Haematol 2014; 165:609.
  14. Galli M, Luciani D, Bertolini G, Barbui T. Lupus anticoagulants are stronger risk factors for thrombosis than anticardiolipin antibodies in the antiphospholipid syndrome: a systematic review of the literature. Blood 2003; 101:1827.
  15. Galli M, Luciani D, Bertolini G, Barbui T. Anti-beta 2-glycoprotein I, antiprothrombin antibodies, and the risk of thrombosis in the antiphospholipid syndrome. Blood 2003; 102:2717.
  16. Opatrny L, David M, Kahn SR, et al. Association between antiphospholipid antibodies and recurrent fetal loss in women without autoimmune disease: a metaanalysis. J Rheumatol 2006; 33:2214.
  17. De Laat B, Derksen RH, Reber G, et al. An international multicentre-laboratory evaluation of a new assay to detect specifically lupus anticoagulants dependent on the presence of anti-beta2-glycoprotein autoantibodies. J Thromb Haemost 2011; 9:149.
  18. de Laat HB, Derksen RH, Urbanus RT, et al. beta2-glycoprotein I-dependent lupus anticoagulant highly correlates with thrombosis in the antiphospholipid syndrome. Blood 2004; 104:3598.
  19. Pengo V, Ruffatti A, Legnani C, et al. Clinical course of high-risk patients diagnosed with antiphospholipid syndrome. J Thromb Haemost 2010; 8:237.
  20. Pengo V, Ruffatti A, Legnani C, et al. Incidence of a first thromboembolic event in asymptomatic carriers of high-risk antiphospholipid antibody profile: a multicenter prospective study. Blood 2011; 118:4714.
  21. Lee EY, Lee CK, Lee TH, et al. Does the anti-beta2-glycoprotein I antibody provide additional information in patients with thrombosis? Thromb Res 2003; 111:29.
  22. Mustonen P, Lehtonen KV, Javela K, Puurunen M. Persistent antiphospholipid antibody (aPL) in asymptomatic carriers as a risk factor for future thrombotic events: a nationwide prospective study. Lupus 2014; 23:1468.
  23. Arad A, Proulle V, Furie RA, et al. β₂-Glycoprotein-1 autoantibodies from patients with antiphospholipid syndrome are sufficient to potentiate arterial thrombus formation in a mouse model. Blood 2011; 117:3453.
  24. de Groot PG, Meijers JC. β(2) -Glycoprotein I: evolution, structure and function. J Thromb Haemost 2011; 9:1275.
  25. Agar C, van Os GM, Mörgelin M, et al. Beta2-glycoprotein I can exist in 2 conformations: implications for our understanding of the antiphospholipid syndrome. Blood 2010; 116:1336.
  26. de Laat B, Derksen RH, van Lummel M, et al. Pathogenic anti-beta2-glycoprotein I antibodies recognize domain I of beta2-glycoprotein I only after a conformational change. Blood 2006; 107:1916.
  27. de Laat B, Derksen RH, Urbanus RT, de Groot PG. IgG antibodies that recognize epitope Gly40-Arg43 in domain I of beta 2-glycoprotein I cause LAC, and their presence correlates strongly with thrombosis. Blood 2005; 105:1540.
  28. de Laat B, Pengo V, Pabinger I, et al. The association between circulating antibodies against domain I of beta2-glycoprotein I and thrombosis: an international multicenter study. J Thromb Haemost 2009; 7:1767.
  29. Pengo V, Ruffatti A, Tonello M, et al. Antiphospholipid syndrome: antibodies to Domain 1 of β2-glycoprotein 1 correctly classify patients at risk. J Thromb Haemost 2015; 13:782.
  30. De Craemer AS, Musial J, Devreese KM. Role of anti-domain 1-β2 glycoprotein I antibodies in the diagnosis and risk stratification of antiphospholipid syndrome. J Thromb Haemost 2016; 14:1779.
  31. Pierangeli SS, Liu XW, Barker JH, et al. Induction of thrombosis in a mouse model by IgG, IgM and IgA immunoglobulins from patients with the antiphospholipid syndrome. Thromb Haemost 1995; 74:1361.
  32. Mehrani T, Petri M. Association of IgA Anti-beta2 glycoprotein I with clinical and laboratory manifestations of systemic lupus erythematosus. J Rheumatol 2011; 38:64.
  33. Sweiss NJ, Bo R, Kapadia R, et al. IgA anti-beta2-glycoprotein I autoantibodies are associated with an increased risk of thromboembolic events in patients with systemic lupus erythematosus. PLoS One 2010; 5:e12280.
  34. Bertolaccini ML, Sciascia S, Murru V, et al. Prevalence of antibodies to prothrombin in solid phase (aPT) and to phosphatidylserine-prothrombin complex (aPS/PT) in patients with and without lupus anticoagulant. Thromb Haemost 2013; 109:207.
  35. Fleck RA, Rapaport SI, Rao LV. Anti-prothrombin antibodies and the lupus anticoagulant. Blood 1988; 72:512.
  36. Sciascia S, Khamashta MA, Bertolaccini ML. New tests to detect antiphospholipid antibodies: antiprothrombin (aPT) and anti-phosphatidylserine/prothrombin (aPS/PT) antibodies. Curr Rheumatol Rep 2014; 16:415.
  37. Sanmarco M, Bardin N. The contribution of antiphosphatidylethanolamine antibodies in the diagnosis of the antiphospholipid syndrome. Lupus 2012; 21:727.
  38. Müller-Calleja N, Hollerbach A, Royce J, et al. Lipid presentation by the protein C receptor links coagulation with autoimmunity. Science 2021; 371.
  39. Sciascia S, Murru V, Sanna G, et al. Clinical accuracy for diagnosis of antiphospholipid syndrome in systemic lupus erythematosus: evaluation of 23 possible combinations of antiphospholipid antibody specificities. J Thromb Haemost 2012; 10:2512.
  40. Sciascia S, Sanna G, Murru V, et al. Anti-prothrombin (aPT) and anti-phosphatidylserine/prothrombin (aPS/PT) antibodies and the risk of thrombosis in the antiphospholipid syndrome. A systematic review. Thromb Haemost 2014; 111:354.
  41. Zuo Y, Navaz S, Tsodikov A, et al. Anti-Neutrophil Extracellular Trap Antibodies in Antiphospholipid Antibody-Positive Patients: Results From the Antiphospholipid Syndrome Alliance for Clinical Trials and InternatiOnal Networking Clinical Database and Repository. Arthritis Rheumatol 2023; 75:1407.
  42. Marciniak E, Romond EH. Impaired catalytic function of activated protein C: a new in vitro manifestation of lupus anticoagulant. Blood 1989; 74:2426.
  43. Garcia D, Erkan D. Diagnosis and Management of the Antiphospholipid Syndrome. N Engl J Med 2018; 378:2010.
  44. Simantov R, LaSala JM, Lo SK, et al. Activation of cultured vascular endothelial cells by antiphospholipid antibodies. J Clin Invest 1995; 96:2211.
  45. Del Papa N, Guidali L, Sala A, et al. Endothelial cells as target for antiphospholipid antibodies. Human polyclonal and monoclonal anti-beta 2-glycoprotein I antibodies react in vitro with endothelial cells through adherent beta 2-glycoprotein I and induce endothelial activation. Arthritis Rheum 1997; 40:551.
  46. Ramesh S, Morrell CN, Tarango C, et al. Antiphospholipid antibodies promote leukocyte-endothelial cell adhesion and thrombosis in mice by antagonizing eNOS via β2GPI and apoER2. J Clin Invest 2011; 121:120.
  47. Allen KL, Fonseca FV, Betapudi V, et al. A novel pathway for human endothelial cell activation by antiphospholipid/anti-β2 glycoprotein I antibodies. Blood 2012; 119:884.
  48. Dunoyer-Geindre S, de Moerloose P, Galve-de Rochemonteix B, et al. NFkappaB is an essential intermediate in the activation of endothelial cells by anti-beta(2)-glycoprotein 1 antibodies. Thromb Haemost 2002; 88:851.
  49. Vega-Ostertag M, Casper K, Swerlick R, et al. Involvement of p38 MAPK in the up-regulation of tissue factor on endothelial cells by antiphospholipid antibodies. Arthritis Rheum 2005; 52:1545.
  50. Allen KL, Hamik A, Jain MK, McCrae KR. Endothelial cell activation by antiphospholipid antibodies is modulated by Kruppel-like transcription factors. Blood 2011; 117:6383.
  51. Pierangeli SS, Espinola RG, Liu X, Harris EN. Thrombogenic effects of antiphospholipid antibodies are mediated by intercellular cell adhesion molecule-1, vascular cell adhesion molecule-1, and P-selectin. Circ Res 2001; 88:245.
  52. Espinola RG, Liu X, Colden-Stanfield M, et al. E-Selectin mediates pathogenic effects of antiphospholipid antibodies. J Thromb Haemost 2003; 1:843.
  53. Kornberg A, Blank M, Kaufman S, Shoenfeld Y. Induction of tissue factor-like activity in monocytes by anti-cardiolipin antibodies. J Immunol 1994; 153:1328.
  54. Amengual O, Atsumi T, Khamashta MA, Hughes GR. The role of the tissue factor pathway in the hypercoagulable state in patients with the antiphospholipid syndrome. Thromb Haemost 1998; 79:276.
  55. Reverter JC, Tàssies D, Font J, et al. Effects of human monoclonal anticardiolipin antibodies on platelet function and on tissue factor expression on monocytes. Arthritis Rheum 1998; 41:1420.
  56. Zhou H, Wolberg AS, Roubey RA. Characterization of monocyte tissue factor activity induced by IgG antiphospholipid antibodies and inhibition by dilazep. Blood 2004; 104:2353.
  57. Sorice M, Longo A, Capozzi A, et al. Anti-beta2-glycoprotein I antibodies induce monocyte release of tumor necrosis factor alpha and tissue factor by signal transduction pathways involving lipid rafts. Arthritis Rheum 2007; 56:2687.
  58. Xie H, Zhou H, Wang H, et al. Anti-β(2)GPI/β(2)GPI induced TF and TNF-α expression in monocytes involving both TLR4/MyD88 and TLR4/TRIF signaling pathways. Mol Immunol 2013; 53:246.
  59. Müller-Calleja N, Köhler A, Siebald B, et al. Cofactor-independent antiphospholipid antibodies activate the NLRP3-inflammasome via endosomal NADPH-oxidase: implications for the antiphospholipid syndrome. Thromb Haemost 2015; 113:1071.
  60. Martini F, Farsi A, Gori AM, et al. Antiphospholipid antibodies (aPL) increase the potential monocyte procoagulant activity in patients with systemic lupus erythematosus. Lupus 1996; 5:206.
  61. Cuadrado MJ, López-Pedrera C, Khamashta MA, et al. Thrombosis in primary antiphospholipid syndrome: a pivotal role for monocyte tissue factor expression. Arthritis Rheum 1997; 40:834.
  62. Dobado-Berrios PM, López-Pedrera C, Velasco F, et al. Increased levels of tissue factor mRNA in mononuclear blood cells of patients with primary antiphospholipid syndrome. Thromb Haemost 1999; 82:1578.
  63. López-Pedrera C, Buendía P, Cuadrado MJ, et al. Antiphospholipid antibodies from patients with the antiphospholipid syndrome induce monocyte tissue factor expression through the simultaneous activation of NF-kappaB/Rel proteins via the p38 mitogen-activated protein kinase pathway, and of the MEK-1/ERK pathway. Arthritis Rheum 2006; 54:301.
  64. Cuadrado MJ, Buendía P, Velasco F, et al. Vascular endothelial growth factor expression in monocytes from patients with primary antiphospholipid syndrome. J Thromb Haemost 2006; 4:2461.
  65. Bernales I, Fullaondo A, Marín-Vidalled MJ, et al. Innate immune response gene expression profiles characterize primary antiphospholipid syndrome. Genes Immun 2008; 9:38.
  66. Perez-Sanchez C, Barbarroja N, Messineo S, et al. Gene profiling reveals specific molecular pathways in the pathogenesis of atherosclerosis and cardiovascular disease in antiphospholipid syndrome, systemic lupus erythematosus and antiphospholipid syndrome with lupus. Ann Rheum Dis 2015; 74:1441.
  67. Nagahama M, Nomura S, Kanazawa S, et al. Significance of anti-oxidized LDL antibody and monocyte-derived microparticles in anti-phospholipid antibody syndrome. Autoimmunity 2003; 36:125.
  68. Vikerfors A, Mobarrez F, Bremme K, et al. Studies of microparticles in patients with the antiphospholipid syndrome (APS). Lupus 2012; 21:802.
  69. Chaturvedi S, Cockrell E, Espinola R, et al. Circulating microparticles in patients with antiphospholipid antibodies: characterization and associations. Thromb Res 2015; 135:102.
  70. Borissoff JI, Joosen IA, Versteylen MO, et al. Elevated levels of circulating DNA and chromatin are independently associated with severe coronary atherosclerosis and a prothrombotic state. Arterioscler Thromb Vasc Biol 2013; 33:2032.
  71. de Boer OJ, Li X, Teeling P, et al. Neutrophils, neutrophil extracellular traps and interleukin-17 associate with the organisation of thrombi in acute myocardial infarction. Thromb Haemost 2013; 109:290.
  72. Fuchs TA, Brill A, Duerschmied D, et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A 2010; 107:15880.
  73. von Brühl ML, Stark K, Steinhart A, et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 2012; 209:819.
  74. Mikes B, Sinkovits G, Farkas P, et al. Elevated plasma neutrophil elastase concentration is associated with disease activity in patients with thrombotic thrombocytopenic purpura. Thromb Res 2014; 133:616.
  75. Jiménez-Alcázar M, Napirei M, Panda R, et al. Impaired DNase1-mediated degradation of neutrophil extracellular traps is associated with acute thrombotic microangiopathies. J Thromb Haemost 2015; 13:732.
  76. Arvieux J, Jacob MC, Roussel B, et al. Neutrophil activation by anti-beta 2 glycoprotein I monoclonal antibodies via Fc gamma receptor II. J Leukoc Biol 1995; 57:387.
  77. Gladigau G, Haselmayer P, Scharrer I, et al. A role for Toll-like receptor mediated signals in neutrophils in the pathogenesis of the anti-phospholipid syndrome. PLoS One 2012; 7:e42176.
  78. Ritis K, Doumas M, Mastellos D, et al. A novel C5a receptor-tissue factor cross-talk in neutrophils links innate immunity to coagulation pathways. J Immunol 2006; 177:4794.
  79. Yalavarthi S, Gould TJ, Rao AN, et al. Release of neutrophil extracellular traps by neutrophils stimulated with antiphospholipid antibodies: a newly identified mechanism of thrombosis in the antiphospholipid syndrome. Arthritis Rheumatol 2015; 67:2990.
  80. Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science 2004; 303:1532.
  81. Branzk N, Lubojemska A, Hardison SE, et al. Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens. Nat Immunol 2014; 15:1017.
  82. Brill A, Fuchs TA, Savchenko AS, et al. Neutrophil extracellular traps promote deep vein thrombosis in mice. J Thromb Haemost 2012; 10:136.
  83. Massberg S, Grahl L, von Bruehl ML, et al. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med 2010; 16:887.
  84. Knight JS, Luo W, O'Dell AA, et al. Peptidylarginine deiminase inhibition reduces vascular damage and modulates innate immune responses in murine models of atherosclerosis. Circ Res 2014; 114:947.
  85. Farkas ÁZ, Farkas VJ, Gubucz I, et al. Neutrophil extracellular traps in thrombi retrieved during interventional treatment of ischemic arterial diseases. Thromb Res 2019; 175:46.
  86. Meng H, Yalavarthi S, Kanthi Y, et al. In Vivo Role of Neutrophil Extracellular Traps in Antiphospholipid Antibody-Mediated Venous Thrombosis. Arthritis Rheumatol 2017; 69:655.
  87. Ali RA, Gandhi AA, Meng H, et al. Adenosine receptor agonism protects against NETosis and thrombosis in antiphospholipid syndrome. Nat Commun 2019; 10:1916.
  88. Knight JS, Meng H, Coit P, et al. Activated signature of antiphospholipid syndrome neutrophils reveals potential therapeutic target. JCI Insight 2017; 2.
  89. Sule G, Kelley WJ, Gockman K, et al. Increased Adhesive Potential of Antiphospholipid Syndrome Neutrophils Mediated by β2 Integrin Mac-1. Arthritis Rheumatol 2020; 72:114.
  90. Pennings MT, Derksen RH, van Lummel M, et al. Platelet adhesion to dimeric beta-glycoprotein I under conditions of flow is mediated by at least two receptors: glycoprotein Ibalpha and apolipoprotein E receptor 2'. J Thromb Haemost 2007; 5:369.
  91. Vega-Ostertag M, Harris EN, Pierangeli SS. Intracellular events in platelet activation induced by antiphospholipid antibodies in the presence of low doses of thrombin. Arthritis Rheum 2004; 50:2911.
  92. Proulle V, Furie RA, Merrill-Skoloff G, et al. Platelets are required for enhanced activation of the endothelium and fibrinogen in a mouse thrombosis model of APS. Blood 2014; 124:611.
  93. Joseph JE, Harrison P, Mackie IJ, et al. Increased circulating platelet-leucocyte complexes and platelet activation in patients with antiphospholipid syndrome, systemic lupus erythematosus and rheumatoid arthritis. Br J Haematol 2001; 115:451.
  94. Cuadrado MJ, Mujic F, Muñoz E, et al. Thrombocytopenia in the antiphospholipid syndrome. Ann Rheum Dis 1997; 56:194.
  95. Diz-Küçükkaya R, Hacihanefioğlu A, Yenerel M, et al. Antiphospholipid antibodies and antiphospholipid syndrome in patients presenting with immune thrombocytopenic purpura: a prospective cohort study. Blood 2001; 98:1760.
  96. Fanelli A, Bergamini C, Rapi S, et al. Flow cytometric detection of circulating activated platelets in primary antiphospholipid syndrome. Correlation with thrombocytopenia and anticardiolipin antibodies. Lupus 1997; 6:261.
  97. Wahl D, Membre A, Perret-Guillaume C, et al. Mechanisms of antiphospholipid-induced thrombosis: effects on the protein C system. Curr Rheumatol Rep 2009; 11:77.
  98. Rossetto V, Spiezia L, Franz F, et al. The role of antiphospholipid antibodies toward the protein C/protein S system in venous thromboembolic disease. Am J Hematol 2009; 84:594.
  99. Oosting JD, Derksen RH, Bobbink IW, et al. Antiphospholipid antibodies directed against a combination of phospholipids with prothrombin, protein C, or protein S: an explanation for their pathogenic mechanism? Blood 1993; 81:2618.
  100. Borrell M, Sala N, de Castellarnau C, et al. Immunoglobulin fractions isolated from patients with antiphospholipid antibodies prevent the inactivation of factor Va by activated protein C on human endothelial cells. Thromb Haemost 1992; 68:268.
  101. Shibata S, Harpel PC, Gharavi A, et al. Autoantibodies to heparin from patients with antiphospholipid antibody syndrome inhibit formation of antithrombin III-thrombin complexes. Blood 1994; 83:2532.
  102. Hwang KK, Grossman JM, Visvanathan S, et al. Identification of anti-thrombin antibodies in the antiphospholipid syndrome that interfere with the inactivation of thrombin by antithrombin. J Immunol 2001; 167:7192.
  103. Yang YH, Chien D, Wu M, et al. Novel autoantibodies against the activated coagulation factor IX (FIXa) in the antiphospholipid syndrome that interpose the FIXa regulation by antithrombin. J Immunol 2009; 182:1674.
  104. Yang YH, Hwang KK, FitzGerald J, et al. Antibodies against the activated coagulation factor X (FXa) in the antiphospholipid syndrome that interfere with the FXa inactivation by antithrombin. J Immunol 2006; 177:8219.
  105. Bu C, Gao L, Xie W, et al. beta2-glycoprotein i is a cofactor for tissue plasminogen activator-mediated plasminogen activation. Arthritis Rheum 2009; 60:559.
  106. Cugno M, Cabibbe M, Galli M, et al. Antibodies to tissue-type plasminogen activator (tPA) in patients with antiphospholipid syndrome: evidence of interaction between the antibodies and the catalytic domain of tPA in 2 patients. Blood 2004; 103:2121.
  107. Yang CD, Hwang KK, Yan W, et al. Identification of anti-plasmin antibodies in the antiphospholipid syndrome that inhibit degradation of fibrin. J Immunol 2004; 172:5765.
  108. Liestøl S, Sandset PM, Jacobsen EM, et al. Decreased anticoagulant response to tissue factor pathway inhibitor type 1 in plasmas from patients with lupus anticoagulants. Br J Haematol 2007; 136:131.
  109. Adams M, Breckler L, Stevens P, et al. Anti-tissue factor pathway inhibitor activity in subjects with antiphospholipid syndrome is associated with increased thrombin generation. Haematologica 2004; 89:985.
  110. Hulstein JJ, Lenting PJ, de Laat B, et al. beta2-Glycoprotein I inhibits von Willebrand factor dependent platelet adhesion and aggregation. Blood 2007; 110:1483.
  111. Meijers JC, Tekelenburg WL, Bouma BN, et al. High levels of coagulation factor XI as a risk factor for venous thrombosis. N Engl J Med 2000; 342:696.
  112. Giannakopoulos B, Gao L, Qi M, et al. Factor XI is a substrate for oxidoreductases: enhanced activation of reduced FXI and its role in antiphospholipid syndrome thrombosis. J Autoimmun 2012; 39:121.
  113. Irman S, Miha S, Igor M, et al. In vitro model of annexin A5 crystallization on natural phospholipid bilayers observed by atomic force microscopy. Autoimmunity 2009; 42:414.
  114. Rand JH, Wu XX, Quinn AS, et al. Human monoclonal antiphospholipid antibodies disrupt the annexin A5 anticoagulant crystal shield on phospholipid bilayers: evidence from atomic force microscopy and functional assay. Am J Pathol 2003; 163:1193.
  115. Rand JH, Wu XX, Quinn AS, et al. Hydroxychloroquine directly reduces the binding of antiphospholipid antibody-beta2-glycoprotein I complexes to phospholipid bilayers. Blood 2008; 112:1687.
  116. Rand JH, Wu XX, Quinn AS, et al. Hydroxychloroquine protects the annexin A5 anticoagulant shield from disruption by antiphospholipid antibodies: evidence for a novel effect for an old antimalarial drug. Blood 2010; 115:2292.
  117. Wu XX, Guller S, Rand JH. Hydroxychloroquine reduces binding of antiphospholipid antibodies to syncytiotrophoblasts and restores annexin A5 expression. Am J Obstet Gynecol 2011; 205:576.e7.
  118. Edwards MH, Pierangeli S, Liu X, et al. Hydroxychloroquine reverses thrombogenic properties of antiphospholipid antibodies in mice. Circulation 1997; 96:4380.
  119. Foley JH, Conway EM. Cross Talk Pathways Between Coagulation and Inflammation. Circ Res 2016; 118:1392.
  120. Carrera-Marín A, Romay-Penabad Z, Papalardo E, et al. C6 knock-out mice are protected from thrombophilia mediated by antiphospholipid antibodies. Lupus 2012; 21:1497.
  121. Fischetti F, Durigutto P, Pellis V, et al. Thrombus formation induced by antibodies to beta2-glycoprotein I is complement dependent and requires a priming factor. Blood 2005; 106:2340.
  122. Pierangeli SS, Girardi G, Vega-Ostertag M, et al. Requirement of activation of complement C3 and C5 for antiphospholipid antibody-mediated thrombophilia. Arthritis Rheum 2005; 52:2120.
  123. Davis WD, Brey RL. Antiphospholipid antibodies and complement activation in patients with cerebral ischemia. Clin Exp Rheumatol 1992; 10:455.
  124. Oku K, Atsumi T, Bohgaki M, et al. Complement activation in patients with primary antiphospholipid syndrome. Ann Rheum Dis 2009; 68:1030.
  125. Breen KA, Seed P, Parmar K, et al. Complement activation in patients with isolated antiphospholipid antibodies or primary antiphospholipid syndrome. Thromb Haemost 2012; 107:423.
  126. Chaturvedi S, Braunstein EM, Yuan X, et al. Complement activity and complement regulatory gene mutations are associated with thrombosis in APS and CAPS. Blood 2020; 135:239.
  127. Meroni PL, Macor P, Durigutto P, et al. Complement activation in antiphospholipid syndrome and its inhibition to prevent rethrombosis after arterial surgery. Blood 2016; 127:365.
  128. Yelnik CM, Miranda S, Mékinian A, et al. Patients with refractory catastrophic antiphospholipid syndrome respond inconsistently to eculizumab. Blood 2020; 136:2473.
  129. Danowski A, de Azevedo MN, de Souza Papi JA, Petri M. Determinants of risk for venous and arterial thrombosis in primary antiphospholipid syndrome and in antiphospholipid syndrome with systemic lupus erythematosus. J Rheumatol 2009; 36:1195.
  130. Matyja-Bednarczyk A, Swadźba J, Iwaniec T, et al. Risk factors for arterial thrombosis in antiphospholipid syndrome. Thromb Res 2014; 133:173.
  131. Erkan D, Harrison MJ, Levy R, et al. Aspirin for primary thrombosis prevention in the antiphospholipid syndrome: a randomized, double-blind, placebo-controlled trial in asymptomatic antiphospholipid antibody-positive individuals. Arthritis Rheum 2007; 56:2382.
  132. Urbanus RT, Siegerink B, Roest M, et al. Antiphospholipid antibodies and risk of myocardial infarction and ischaemic stroke in young women in the RATIO study: a case-control study. Lancet Neurol 2009; 8:998.
  133. Viall CA, Chamley LW. Histopathology in the placentae of women with antiphospholipid antibodies: A systematic review of the literature. Autoimmun Rev 2015; 14:446.
  134. Tedesco F, Borghi MO, Gerosa M, et al. Pathogenic Role of Complement in Antiphospholipid Syndrome and Therapeutic Implications. Front Immunol 2018; 9:1388.
  135. Tong M, Viall CA, Chamley LW. Antiphospholipid antibodies and the placenta: a systematic review of their in vitro effects and modulation by treatment. Hum Reprod Update 2015; 21:97.
  136. La Rosa L, Meroni PL, Tincani A, et al. Beta 2 glycoprotein I and placental anticoagulant protein I in placentae from patients with antiphospholipid syndrome. J Rheumatol 1994; 21:1684.
  137. Chamley LW, Allen JL, Johnson PM. Synthesis of beta2 glycoprotein 1 by the human placenta. Placenta 1997; 18:403.
  138. Di Simone N, Meroni PL, de Papa N, et al. Antiphospholipid antibodies affect trophoblast gonadotropin secretion and invasiveness by binding directly and through adhered beta2-glycoprotein I. Arthritis Rheum 2000; 43:140.
  139. Di Simone N, Raschi E, Testoni C, et al. Pathogenic role of anti-beta 2-glycoprotein I antibodies in antiphospholipid associated fetal loss: characterisation of beta 2-glycoprotein I binding to trophoblast cells and functional effects of anti-beta 2-glycoprotein I antibodies in vitro. Ann Rheum Dis 2005; 64:462.
  140. Chamley LW, Duncalf AM, Mitchell MD, Johnson PM. Action of anticardiolipin and antibodies to beta2-glycoprotein-I on trophoblast proliferation as a mechanism for fetal death. Lancet 1998; 352:1037.
  141. Carroll TY, Mulla MJ, Han CS, et al. Modulation of trophoblast angiogenic factor secretion by antiphospholipid antibodies is not reversed by heparin. Am J Reprod Immunol 2011; 66:286.
  142. Marchetti T, Ruffatti A, Wuillemin C, et al. Hydroxychloroquine restores trophoblast fusion affected by antiphospholipid antibodies. J Thromb Haemost 2014; 12:910.
  143. Mulla MJ, Salmon JE, Chamley LW, et al. A role for uric acid and the Nalp3 inflammasome in antiphospholipid antibody-induced IL-1β production by human first trimester trophoblast. PLoS One 2013; 8:e65237.
  144. Gysler SM, Mulla MJ, Guerra M, et al. Antiphospholipid antibody-induced miR-146a-3p drives trophoblast interleukin-8 secretion through activation of Toll-like receptor 8. Mol Hum Reprod 2016; 22:465.
  145. Holers VM, Girardi G, Mo L, et al. Complement C3 activation is required for antiphospholipid antibody-induced fetal loss. J Exp Med 2002; 195:211.
  146. Girardi G, Berman J, Redecha P, et al. Complement C5a receptors and neutrophils mediate fetal injury in the antiphospholipid syndrome. J Clin Invest 2003; 112:1644.
  147. Berman J, Girardi G, Salmon JE. TNF-alpha is a critical effector and a target for therapy in antiphospholipid antibody-induced pregnancy loss. J Immunol 2005; 174:485.
  148. Redecha P, Tilley R, Tencati M, et al. Tissue factor: a link between C5a and neutrophil activation in antiphospholipid antibody induced fetal injury. Blood 2007; 110:2423.
  149. Kim MY, Guerra MM, Kaplowitz E, et al. Complement activation predicts adverse pregnancy outcome in patients with systemic lupus erythematosus and/or antiphospholipid antibodies. Ann Rheum Dis 2018; 77:549.
  150. Girardi G, Redecha P, Salmon JE. Heparin prevents antiphospholipid antibody-induced fetal loss by inhibiting complement activation. Nat Med 2004; 10:1222.
  151. Hughson MD, McCarty GA, Brumback RA. Spectrum of vascular pathology affecting patients with the antiphospholipid syndrome. Hum Pathol 1995; 26:716.
  152. Manning BD, Cantley LC. AKT/PKB signaling: navigating downstream. Cell 2007; 129:1261.
  153. Canaud G, Terzi F. Inhibition of the mTORC pathway in the antiphospholipid syndrome. N Engl J Med 2014; 371:1554.
  154. Connors JM, Levy JH. COVID-19 and its implications for thrombosis and anticoagulation. Blood 2020; 135:2033.
  155. Lodigiani C, Iapichino G, Carenzo L, et al. Venous and arterial thromboembolic complications in COVID-19 patients admitted to an academic hospital in Milan, Italy. Thromb Res 2020; 191:9.
  156. Zuo Y, Yalavarthi S, Shi H, et al. Neutrophil extracellular traps in COVID-19. JCI Insight 2020; 5.
  157. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020; 395:1417.
  158. Manne BK, Denorme F, Middleton EA, et al. Platelet gene expression and function in patients with COVID-19. Blood 2020; 136:1317.
  159. Magro C, Mulvey JJ, Berlin D, et al. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: A report of five cases. Transl Res 2020; 220:1.
  160. Zhang Y, Xiao M, Zhang S, et al. Coagulopathy and Antiphospholipid Antibodies in Patients with Covid-19. N Engl J Med 2020; 382:e38.
  161. Harzallah I, Debliquis A, Drénou B. Lupus anticoagulant is frequent in patients with Covid-19. J Thromb Haemost 2020; 18:2064.
  162. Xiao M, Zhang Y, Zhang S, et al. Antiphospholipid Antibodies in Critically Ill Patients With COVID-19. Arthritis Rheumatol 2020; 72:1998.
  163. Devreese KMJ, Linskens EA, Benoit D, Peperstraete H. Antiphospholipid antibodies in patients with COVID-19: A relevant observation? J Thromb Haemost 2020; 18:2191.
  164. Zuo Y, Estes SK, Ali RA, et al. Prothrombotic autoantibodies in serum from patients hospitalized with COVID-19. Sci Transl Med 2020; 12.
  165. Abdel-Wahab N, Talathi S, Lopez-Olivo MA, Suarez-Almazor ME. Risk of developing antiphospholipid antibodies following viral infection: a systematic review and meta-analysis. Lupus 2018; 27:572.
  166. Chaturvedi S, Braunstein EM, Brodsky RA. Antiphospholipid syndrome: Complement activation, complement gene mutations, and therapeutic implications. J Thromb Haemost 2021; 19:607.
Topic 4676 Version 28.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟