pathogenesis antifosfolip syndrome

23
About Us|News from UpToDate|Contact Us|Help Log in New Search Patient Info What's New Calculators SearchAll Topics Back to Search Results for " antiphospholipid syndrome " Pathogenesis of the antiphospholipid syndrome Find Print Email < div id="javascriptDisabled"> It seems to us that you have your JavaScript turned off on your browser. JavaScript is required in order for our site to behave correctly. Please enable your JavaScript to continue use our site.< /div> Pathogenesis of the antiphospholipid syndrome Authors Bonnie L Bermas, MD Peter H Schur, MD Section Editor David S Pisetsky, MD, PhD Deputy Editor Paul L Romain, MD Disclosures All topics are updated as new evidence becomes available and our peer review process is complete. antiphospho 0 0 USER_INPUT TO P_PULLDO W N

Upload: voicugabi

Post on 21-Jul-2016

2 views

Category:

Documents


0 download

DESCRIPTION

agdg

TRANSCRIPT

Page 1: Pathogenesis Antifosfolip Syndrome

About Us|News from UpToDate|Contact Us|HelpLog in

New Search

Patient Info

What's New

Calculators

SearchAll Topics

Back to Search Results for " antiphospholipid syndrome " Pathogenesis of the antiphospholipid syndrome

Find Print Email

< div id="javascriptDisabled"> It seems to us that you have your JavaScript turned off on your browser. JavaScript is required in order for our site to behave correctly. Please enable your JavaScript to continue use our site.< /div>

Pathogenesis of the antiphospholipid syndromeAuthorsBonnie L Bermas, MDPeter H Schur, MDSection EditorDavid S Pisetsky, MD, PhDDeputy EditorPaul L Romain, MDDisclosures

All topics are updated as new evidence becomes available and our peer review process is complete.Literature review current through: Nov 2012. |This topic last updated: Sep 11, 2012.

INTRODUCTION — The antiphospholipid antibody syndrome (APS) is defined by the presence of two

major components:

antiphospho

0 0 USER_INPUT TOP_PULLDOWN

Page 2: Pathogenesis Antifosfolip Syndrome

Presence in the plasma of at least one type of autoantibody known as an antiphospholipid

antibody (aPL).

The occurrence of at least one of the following clinical manifestations: venous or arterial

thromboses, or pregnancy morbidity.

Although the clinical manifestations of APS occur in other disease populations, in the APS they occur by

definition in the context of aPL. aPL, which are directed against plasma proteins bound to anionic

phospholipids, may be detected as:

Lupus anticoagulants

Anticardiolipin antibodies

Antibodies to ß2 glycoprotein-I

The full clinical significance of other autoantibodies, including those directed against prothrombin, annexin

V, phosphatidylserine, and phosphatidylinositol, remain unclear.

APS occurs either as a primary condition or in the setting of an underlying disease, particularly systemic

lupus erythematosus (SLE).

The pathogenesis of the APS will be reviewed here. The clinical manifestations, diagnosis, and treatment

of the APS are discussed separately. (See "Clinical manifestations of the antiphospholipid syndrome" and

"Diagnosis of the antiphospholipid syndrome" and "Treatment of the antiphospholipid syndrome" and

"Obstetrical manifestations of the antiphospholipid syndrome".)

ANTIPHOSPHOLIPID ANTIBODIES — The APS is characterized by antibodies directed against epitopes

on plasma proteins that are uncovered or generated by the binding of these proteins to phospholipids;

they do not bind directly to anionic phospholipids [1,2]. The mechanisms by which aPL cause thrombosis

are not completely understood.

There are a variety of tests that can detect aPL:

Anticardiolipin antibodies

Functional assays for the detection of aPL with lupus anticoagulant activity

Anti-ß2-glycoprotein-I antibodies

These tests vary in their sensitivity and specificity.

The standard types of aPL assays performed are enzyme-linked immunoassays (ELISAs) for

anticardiolipin antibodies (aCL) and antibodies to ß2-glycoprotein-I (ß2-GP-I), and coagulation tests

designed to detect lupus anticoagulants (LAs).

Anticardiolipin antibodies — aCL react with cardiolipin but may also react with phosphatidylserine,

phosphatidylinositol, phosphatidylglycerol, ß2-GP-I, prothrombin, or annexin V [3].

Page 3: Pathogenesis Antifosfolip Syndrome

The concordance between the presence of an LA and aCL is approximately 85 percent. In many cases,

however, LAs comprises a separate population of antibodies from aCL [1,4,5]. Thus, testing should be

performed for both LAs and aCL if APS is suspected on a clinical basis. (See "Diagnosis of the

antiphospholipid syndrome".)

Elevated levels of IgG aCL confer a greater risk of thrombosis than do other immunoglobulin isotypes [6].

However, IgM and IgA aCL isotypes may be associated with the APS [7].

False positive serologic test for syphilis — The biologic false positive test for syphilis (BFPTS)

phenomenon occurs because the syphilis antigen used in the VDRL and RPR tests is cardiolipin mixed

with cephaline and cholesterol. Examples of BFPTS are positive rapid plasma reagin (RPR) or Venereal

Disease Research Laboratory (VDRL) tests that are not confirmed by specific treponemal assays. These

tests for the diagnosis of syphilis have been replaced by specific antitreponemal tests in most

laboratories.

RPR and VDRL assays, if available, are not appropriate screening tests for aPL because of their low

sensitivities and specificities [8]. (See "Diagnostic testing for syphilis", section on 'Interpretation of

serologic testing'.)

Lupus anticoagulants — LAs are antibodies directed against plasma proteins such as ß2-GP-I,

prothrombin, or annexin V that are bound to anionic phospholipids [9-12]. The phrase "the lupus

anticoagulant" is a misnomer, for three reasons:

The presence of an LA is generally associated with a clotting tendency, not an anticoagulant

effect. (See "Clinical manifestations of the antiphospholipid syndrome", section on

'Thrombosis'.)

More than one antibody is associated with LA activity. As examples, both aCL and

antibodies to ß2-GP-I can have LA activity.

Only about 50 percent of individuals with an LA meet the American College of

Rheumatology criteria for the classification of SLE. (See "Clinical manifestations of the

antiphospholipid syndrome", section on 'Primary APS versus APS with SLE'.)

Because there is more than one LA and not all antibodies with APS are associated with anticoagulant

activity, the detection of LAs occurs through functional clotting tests, not through enzyme-linked

immunosorbent assays (ELISAs). LAs block in vitro assembly of the prothrombinase complex, resulting in

a prolongation of in vitro clotting assays such as the activated partial thromboplastin time (aPTT), the

dilute Russell viper venom time (dRVVT), the kaolin clotting time and, infrequently, the prothrombin time.

(See "Clinical use of coagulation tests" and "Diagnosis of the antiphospholipid syndrome", section on

'Lupus anticoagulant'.)

Prolongation of clotting times — Clotting factor deficiencies that lead to prolongation of coagulation

assays are reversed when the patient's plasma is diluted 1:1 with normal platelet-free plasma. Such a

Page 4: Pathogenesis Antifosfolip Syndrome

procedure is known as a mixing study. (See "Clinical use of coagulation tests", section on 'Mixing

studies'.)

In contrast, abnormalities of coagulation assays are not reversed when the cause is an LA [1,9]. However,

they can be reversed by incubation of the patient's plasma with a hexagonal phase phospholipid that

neutralizes the inhibitor [1,4,9].

Abnormal bleeding times — Many patients with LAs have prolonged bleeding times despite adequate

platelet counts [13,14]. Although these changes suggest impaired coagulation, patients with an LA have a

paradoxical increase in the frequency of arterial and venous thrombotic events [15,16].

Anti-ß2-glycoprotein-I antibodies — ß2-GP-I (apolipoprotein H) is a naturally occurring inhibitor of

coagulation and platelet aggregation [17]. The properties of this protein as a clotting inhibitor could

explain why neutralizing antibodies promote thrombosis. Consistent with this hypothesis is the

observation that aPL prolong the aPTT if added to normal plasma but not to plasma depleted of ß2-GP-I

[18]. ß2-GP-I binds to negatively-charged phospholipids such as phosphatidylserine and

phosphatidylinositol and inhibits both contact activation of the clotting cascade and the conversion of

prothrombin to thrombin [19,20].

Antibodies to ß2-GP-I are found in a large percentage of patients with primary or secondary APS [21].

Although antibodies to ß2-GP-I are usually found in association with other aPL, they are the sole aPL

detectable in approximately 11 percent of such patients [21]. (See "Diagnosis of the antiphospholipid

syndrome".)

Additional aPL — In addition to aCL, LAs, and antibodies to ß2-GP-I, other antibodies have been

reported in association with clinical features of the APS. These include antibodies to:

Prothrombin

Annexin V

Phosphatidylserine

Phosphatidylinositol

Phosphatidylethanolamine

In general, assays for these antibodies are not standardized for clinical use. Their interpretation and

relevance to clinical practice remains unclear. Among these additional aPL, the largest amount of

literature relates to antiprothrombin antibodies.

Antiprothrombin antibodies — Antiprothrombin antibodies have been described in association with both

clotting and hemorrhage [22]. The presence of prothrombin antibodies should be suspected when a

patient with known aPL develops a bleeding tendency. (See "Acquired inhibitors of coagulation", section

on 'Other coagulation factor inhibitors'.)

Page 5: Pathogenesis Antifosfolip Syndrome

Antiprothrombin antibodies have been described in 50 percent of patients with aPL, 34 percent of patients

with SLE, and 57 percent of patients with Sneddon's syndrome (livedo reticularis with stroke or transient

ischemic attacks) [23-25].

Antibodies directed against prothrombin may interfere with coagulation in conjunction with other aPL [23].

As an example, antibodies to a complex of phosphatidylserine and prothrombin appear to be more

significant in terms of the risk of clinical events than antibodies to prothrombin alone [26,27]. (See

"Acquired inhibitors of coagulation", section on 'Prothrombin inhibitors'.)

Antiprothrombin antibodies are also associated with an increased likelihood of recurrent venous

thromboembolic disease that is independent of the presence of aCL or LA. In one study of 236

consecutive patients with acute venous thrombotic events, the following observations were made [28]:

aPL were found in 85 (36 percent), of whom 24 (10 percent of the overall cohort) had

antiprothrombin antibodies.

A history of previous thromboembolism was identified in nearly one-quarter of the patients.

In a multivariate analysis, antiprothrombin antibodies were associated with previous

thromboembolism (odds ratio 3.3).

Future studies and further refinement of APS assays are likely to clarify the role of antibodies to

prothrombin, annexin V, phosphatidylserine, and other aPL targets.

PATHOGENESIS — The pathogenesis of the APS associated clinical manifestations appears to result

from a variety of aPL effects upon pathways of coagulation, including the procoagulant actions of these

antibodies upon protein C, annexin V, platelets, serum proteases, toll-like receptors, tissue factor, and via

impaired fibrinolysis [29-36]. In addition to heightening the risk of vascular thrombosis, aPL increase

vascular tone, thereby increasing the susceptibility to atherosclerosis, fetal loss, and neurological damage

[37].

The most commonly accepted explanation for the development of aPL is that they occur in susceptible

individuals following incidental exposure to infectious agents. However, aside from the milieu of rheumatic

diseases such as SLE, the conditions creating such susceptibility remain largely undefined.

Current thinking about the pathogenesis of the APS holds that once aPL are present, a "second-hit" is

required for the development of the full-blown syndrome [38]. Potential candidates for the delivery of such

a second hit are smoking, prolonged immobilization, pregnancy and the post-partum period, oral

contraceptive use, hormone replacement therapy, malignancy, nephrotic syndrome, hypertension, and

hyperlipidemia.

Clues to the pathogenesis of the APS derive from in vitro experiments, animal models, and clinical studies

in humans.

Page 6: Pathogenesis Antifosfolip Syndrome

In vitro experiments — Multiple in vitro experiments suggest that antibodies to ß2-GP-I play an

important role in the thrombotic events associated with APS. The range of findings is summarized by the

following points:

ß2-GP-I helps maintain adequate plasma levels of free protein S [39]. Antibodies to ß2-GP-I

may cause thrombosis by interfering with the activity of protein S. (See "Protein S

deficiency", section on 'Physiology of protein S'.)

Other aPL may have similar effects on protein C and other coagulation factors through

binding to these proteins or receptors on endothelial cell surfaces [40-43]. (See "Protein C

deficiency", section on 'Physiology of protein C'.)

Specific T cells in APS patients respond to stimulation with ß2-GP-I [44]. Depletion of these

T cells significantly reduces anti-ß2-GP-I antibody production.

Complexes of ß2-GP-I and aCL bind to platelet phospholipids, upregulating coagulation and

stimulating platelet aggregation [45]. Antibody binding to ß2-GP-I attached to the platelet

glycoprotein Ib-IX-V receptor may lead to platelet activation [46].

Antibodies to ß2-GP-I may cross-react with antigenic determinants on endothelial cell CD40

[47]. CD40 ligation in vitro causes endothelial cell activation characterized in part by

increased surface expression of adhesion molecules [48].

APL may lead to platelet activation and facilitate the adherence of platelets to endothelium

[40,49]. This effect may be enhanced by the presence of antibodies to both aCL, ß2-GP-I,

and phosphatidylserine/prothrombin complexes [50], as well as by the presence of thrombin

[51].

APL may activate vascular endothelium, resulting in increased binding of monocytes and

activated platelets and the expression of tissue factor, chemokines, and growth factors [52-

55].

Animal models — The following data from animal models that are relevant to the pathogenesis of the

APS include:

Findings in a mouse model of APS challenge the dogma that this syndrome is entirely a

noninflammatory, thrombotic disease, and provide evidence that complement activation is

critical for APS pregnancy complications [56,57].

In the same mouse model, aPL-induced fetal loss may be inhibited by treatment with

heparin [58], and some evidence in humans suggests that similar findings apply to patients

with the APS [59]. (See 'Clinical studies' below.)

The infusion of human or murine aCL or immunization with a monoclonal human aCL

prolongs the PTT and increases the rate of fetal loss in mice [60,61].

Data from a mouse model suggest that antibodies directed against an antigenic hexapeptide

derived from ß2-GP-I may cross-react with bacterial proteins containing the same motif (ie,

possible molecular mimicry) [62]. Passive transfer of purified antibodies to the hexapeptide

Page 7: Pathogenesis Antifosfolip Syndrome

was associated with decreased platelets counts, fetal resorption, and prolongation of the

activated partial tissue thromboplastin time.

In other mouse experiments, antibodies to a 15 amino acid peptide derived from ß2-GP-I

transfer disease [63]. Some data indicate that one ß2-GP-I domain (domain I) plays a role in

human disease [64].

Clinical studies — Clinical studies in humans are consistent with findings from in vitro and animal model

experiments about the importance of complement activation and antibodies to ß2-GP-I in disease

pathogenesis:

Patients with primary APS had lower levels of CH50, C3 and C4 than controls; they also had

increased levels of complement fragments indicative of complement activation (ie, C3a and

C4a) [65].

When immunohistochemical analyses were performed on placentas from 47 patients with

aPL and 23 healthy controls, there was evidence of increased complement deposition within

the trophoblast cytoplasm, trophoblastic cell and basement membrane, and extravillous

trophoblasts of placentas from aPL patients [59]. These findings are consistent with murine

studies implicating complement as a critical factor in fetal tissue injury [56-58].

However, it remains uncertain whether complement activation causes a hypercoagulable state, or, results

from it.

Antibodies to ß2-GP-I correlate better with the development of APS than do aCL or LAs

alone [1,66-70]. As an example, in a study of 100 patients with an LA, 12 of the 14 who

developed a thromboembolic event had prolongations of their dilute Russell viper venom

time (dRVVT) assay, a coagulation test that is highly sensitive for antibodies to ß2-GP-I [70].

Antibodies to ß2-GP-I were found in 35 of 39 lupus patients with features of APS compared

with only two of 55 patients without APS [66,67]. As noted above, patients with syphilis and

other infections that often lead to aCL production do not form anti-ß2-GP-I antibodies, which

may explain why they do not become hypercoagulable [40,66,67].

Patients with antibodies to ß2-GP-I but without either LA or aCL can display the clinical

features of APS, including venous and/or arterial thromboses [21,71].

APS patients express greater quantities of tissue factor, the major initiator of coagulation in

vivo, on the surfaces of monocytes. In one report, enhanced monocyte expression of tissue

factor was observed in patients with primary APS and a history of thrombosis, but not in

patients with aPL without thrombosis or in patients with thrombosis but without aPL [72].

Monocytes from patients with thromboembolic phenomena also appear to over-express

potentially procoagulant proteins including: annexin I and II, protein disulfide isomerase,

Nedd8, RhoA, and heat shock protein-60 on their surfaces; in contrast, monocytes from

patients with recurrent abortions reportedly over-express fibrinogen and hemoglobin [73].

Page 8: Pathogenesis Antifosfolip Syndrome

Interaction with other antibodies — APL may also exert their effects through complex interactions with

other antibodies. As examples, patients with aPL may also have antibodies directed against

heparin/heparan sulfate, platelet-activating factor, tissue-type plasminogen activator, annexins (2, IV, and

V), thromboplastin, oxidized low density lipoproteins, thrombomodulin, kininogen, and coagulation factors

VII, VIIa, and XII [23,24,37,74-87].

Microparticles — Microparticles are fragments of cell surface membranes that are released from

apoptotic, activated, and damaged cells. The plasma concentration of microparticles that are derived from

endothelial cells is increased among patients with APS when compared to people with aPL without

thrombotic events and to healthy controls [88-90]. (See "Endothelial dysfunction", section on

'Microparticles'.)

GENETIC PREDISPOSITION — Although genetic studies of the APS are in their infancy, genetic risk

factors such as concurrence of coagulation factor mutations heighten the risk of aPL-associated

thrombosis. In different studies, factor V Leiden and the prothrombin gene mutation and activated protein

C resistance were associated with an increased risk of venous thromboembolism in patients with aPL

[91,92]. (See appropriate topic reviews on the different hypercoagulable states).

Several other types of studies have implicated genetic factors in the pathogenesis of the APS. The

following are illustrative:

Relatives of probands with aPL are more likely to have aPL. In a series of 23 patients with

aCL, for example, 29 of their 87 relatives (33 percent) also had aCL [93].

There is a strong association of aPL with HLA-DR7 in Canadian, German, Italian, and

Mexican patients, and with HLA-DQ7 in American and Spanish patients [94].

Weaker associations have been noted between various polymorphisms of the

immunoglobulin receptor (Fcgamma RIIA) and platelet glycoproteins [95,96].

The inheritance of certain polymorphisms of the ß2-GP-I gene and of the HLA-DMA and

HLA-DMB genes may also enhance the risk of aPL [97,98].

An increased incidence of arterial thrombosis may be associated with certain platelet

glycoprotein polymorphisms [96].

Genetic associations between two genes known to be associated with the risk of SLE (ie,

STAT4 gene and BLK gene) were noted in a study of 133 patients with primary APS.

However, two other SLE-associated genes were not associated with APS (ie, BANK1 gene

and IRF5 gene) [99].

PREVALENCE IN DIFFERENT CONDITIONS — In addition to their occurrence in primary APS, aPL are

found in many other clinical settings. Their clinical significance varies widely in the conditions outlined

below.

Healthy individuals — A minority of healthy individuals have aPL. In most cases, the aPL are not

present upon retesting. However, in some individuals, the presence of aPL is associated with an

Page 9: Pathogenesis Antifosfolip Syndrome

increased risk of developing the APS. (See "Diagnosis of the antiphospholipid syndrome", section on

'Initial laboratory testing'.)

Autoimmune and rheumatic diseases — The most important rheumatic disease associated with aPL is

SLE. aPL occur in a substantial proportion of patients with SLE [100-103]:

Approximately 31 percent of patients have an LA

23 to 47 percent have an aCL

20 percent have antibodies to ß2-GP-I

Conversely, approximately 50 percent of patients with an LA have SLE [5,102].

Both LAs and aCL have also been found in patients with a variety of other autoimmune and rheumatic

diseases (eg, scleroderma, psoriatic arthritis) but, in the absence of clinical events associated with the

APS, their significance is not clear [102,104].

Infections — APL have also been noted in patients with infections. These are usually IgM aCL, which

may occasionally result in thrombotic events [102,105]. Furthermore, these antibodies usually do not have

anti-ß2-GP-I antibody activity [106,107].

The infections that have been associated with aPL include [104,106-114]:

Bacterial infections — Bacterial septicemia, leptospirosis, syphilis, Lyme disease

(borreliosis), tuberculosis, leprosy, infective endocarditis, post-streptococcal rheumatic fever,

and Klebsiella infections.

Viral infections — Hepatitis A, B, and C, mumps, HIV, HTLV-I, cytomegalovirus, varicella-

zoster, Epstein-Barr virus, adenovirus parvovirus, and rubella. Several earlier studies had

reported an association between infection with hepatitis C virus and aPL [109-111].

However, more recent studies suggest no link between the two disorders [112]. As a result,

the correlation between hepatitis C virus infection and aPL, if present, is weak and may not

have underlying pathogenic significance.

Parasitic infections — Malaria, Pneumocystis jirovecii, and visceral leishmaniasis (also

known as kala-azar).

Medications — A number of medications have been associated with aPL. These include phenothiazines

(chlorpromazine), phenytoin, hydralazine, procainamide, quinidine, quinine, dilantin, ethosuximide, alpha

interferon, amoxicillin, chlorothiazide, oral contraceptives, and propranolol [104,105,115]. The aPL are

usually transient, often of the IgM isotype, and rarely associated with thrombosis. The mechanism of

drug-induced aPL is not known.

Neoplasms — Associations with malignant neoplasms have been reported including solid tumors of the

lung, colon, cervix, prostate, kidney, ovary, breast, and bone; with Hodgkin's and non-Hodgkin

lymphomas; and with myelofibrosis, polycythemia vera, myeloid and lymphocytic leukemias [104].

Page 10: Pathogenesis Antifosfolip Syndrome

Other associations — APL have been noted in association with immune thrombocytopenia, sickle cell

anemia, pernicious anemia, diabetes mellitus, inflammatory bowel disease, dialysis, and Klinefelter

syndrome [104].

SUMMARY

The antiphospholipid antibody syndrome (APS) is defined by two major components:

Presence in the plasma of at least one type of autoantibody known as an antiphospholipid

antibody (aPL).

The occurrence of at least one clinical feature: either venous or arterial thromboses, or

pregnancy morbidity.

Although the clinical manifestations of APS occur in other disease populations, in the APS they occur by

definition in the context of aPL. aPL, which are directed against plasma proteins bound to anionic

phospholipids, may be detected as:

Lupus anticoagulants

Anticardiolipin antibodies

Antibodies to ß2 glycoprotein-I

In addition to aCL, LAs, and antibodies to ß2-GP-I, other antibodies have been reported to be associated

with thrombosis. Their relevance in the APS remains uncertain. Antiprothrombin antibodies may be

associated with a bleeding tendency as well as thrombosis. (See 'Additional aPL' above.)

The pathogenesis of the APS associated clinical manifestations appears to result from a variety of aPL

effects upon pathways of coagulation, including the procoagulant actions of these antibodies upon protein

C, annexin V, platelets, and fibrinolysis. (See 'Pathogenesis' above.)

The most commonly accepted explanation for the development of aPL is that they occur in susceptible

individuals following incidental exposure to infectious agents. However, aside from the milieu of rheumatic

diseases such as SLE, the conditions creating such susceptibility remain largely undefined.

Current thinking about the pathogenesis of the APS holds that once aPL are present, a "second-hit" is

required for the development of the full-blown syndrome.

Multiple in vitro experiments suggest that antibodies to ß2-GP-I play an important role in the APS.

Findings in a mouse model of APS provide evidence that complement activation is critical for APS

pregnancy complications.

Use of UpToDate is subject to the Subscription and License Agreement.

REFERENCES

1. Roubey RA. Immunology of the antiphospholipid antibody syndrome. Arthritis Rheum 1996; 39:1444.

Page 11: Pathogenesis Antifosfolip Syndrome

2. Ichikawa K, Khamashta MA, Koike T, et al. beta 2-Glycoprotein I reactivity of monoclonal anticardiolipin antibodies from patients with the antiphospholipid syndrome. Arthritis Rheum 1994; 37:1453.

3. Harris EN, Gharavi AE, Boey ML, et al. Anticardiolipin antibodies: detection by radioimmunoassay and association with thrombosis in systemic lupus erythematosus. Lancet 1983; 2:1211.

4. Ginsberg JS, Wells PS, Brill-Edwards P, et al. Antiphospholipid antibodies and venous thromboembolism. Blood 1995; 86:3685.

5. Triplett DA, Brandt JT, Musgrave KA, Orr CA. The relationship between lupus anticoagulants and antibodies to phospholipid. JAMA 1988; 259:550.

6. Sammaritano LR, Ng S, Sobel R, et al. Anticardiolipin IgG subclasses: association of IgG2 with arterial and/or venous thrombosis. Arthritis Rheum 1997; 40:1998.

7. Gallart T, Benito C, Reverter JC, et al. True anti-anionic phospholipid immunoglobulin M antibodies can exert lupus anticoagulant activity. Br J Haematol 2002; 116:875.

8. Sammaritano LR, Gharavi AE, Lockshin MD. Antiphospholipid antibody syndrome: immunologic and clinical aspects. Semin Arthritis Rheum 1990; 20:81.

9. Santoro SA. Antiphospholipid antibodies and thrombotic predisposition: underlying pathogenetic mechanisms. Blood 1994; 83:2389.

10. Roubey RA. Autoantibodies to phospholipid-binding plasma proteins: a new view of lupus anticoagulants and other "antiphospholipid" autoantibodies. Blood 1994; 84:2854.

11. Rauch J. Lupus anticoagulant antibodies: recognition of phospholipid-binding protein complexes. Lupus 1998; 7 Suppl 2:S29.

12. Passam F, Krilis S. Laboratory tests for the antiphospholipid syndrome: current concepts. Pathology 2004; 36:129.

13. Urbanus RT, de Laat HB, de Groot PG, Derksen RH. Prolonged bleeding time and lupus anticoagulant: a second paradox in the antiphospholipid syndrome. Arthritis Rheum 2004; 50:3605.

14. Orlando E, Cortelazzo S, Marchetti M, et al. Prolonged bleeding time in patients with lupus anticoagulant. Thromb Haemost 1992; 68:495.

15. Asherson RA, Khamashta MA, Ordi-Ros J, et al. The "primary" antiphospholipid syndrome: major clinical and serological features. Medicine (Baltimore) 1989; 68:366.

16. Mackworth-Young CG, Loizou S, Walport MJ. Antiphospholipid antibodies and disease. Q J Med 1989; 72:767.

17. Harris EN, Gharavi A, Asherson RA, et al. Antiphospholipid antibodies--middle aged but robust. J Rheumatol 1994; 21:978.

18. Roubey RA, Pratt CW, Buyon JP, Winfield JB. Lupus anticoagulant activity of autoimmune antiphospholipid antibodies is dependent upon beta 2-glycoprotein I. J Clin Invest 1992; 90:1100.

19. Schousboe I. beta 2-Glycoprotein I: a plasma inhibitor of the contact activation of the intrinsic blood coagulation pathway. Blood 1985; 66:1086.

20. Nimpf J, Bevers EM, Bomans PH, et al. Prothrombinase activity of human platelets is inhibited by beta 2-glycoprotein-I. Biochim Biophys Acta 1986; 884:142.

21. Day HM, Thiagarajan P, Ahn C, et al. Autoantibodies to beta2-glycoprotein I in systemic lupus erythematosus and primary antiphospholipid antibody syndrome: clinical correlations in comparison with other antiphospholipid antibody tests. J Rheumatol 1998; 25:667.

22. Galli M. Should we include anti-prothrombin antibodies in the screening for the antiphospholipid syndrome? J Autoimmun 2000; 15:101.

23. Galli M. Non beta 2-glycoprotein I cofactors for antiphospholipid antibodies. Lupus 1996; 5:388.24. Puurunen M, Vaarala O, Julkunen H, et al. Antibodies to phospholipid-binding plasma proteins and

occurrence of thrombosis in patients with systemic lupus erythematosus. Clin Immunol Immunopathol 1996; 80:16.

25. Kalashnikova LA, Korczyn AD, Shavit S, et al. Antibodies to prothrombin in patients with Sneddon's syndrome. Neurology 1999; 53:223.

26. Amengual O, Atsumi T, Koike T. Specificities, properties, and clinical significance of antiprothrombin antibodies. Arthritis Rheum 2003; 48:886.

27. Bertolaccini ML, Atsumi T, Koike T, et al. Antiprothrombin antibodies detected in two different assay systems. Prevalence and clinical significance in systemic lupus erythematosus. Thromb Haemost 2005; 93:289.

Page 12: Pathogenesis Antifosfolip Syndrome

28. Zanon E, Saggiorato G, Ramon R, et al. Anti-prothrombin antibodies as a potential risk factor of recurrent venous thromboembolism. Thromb Haemost 2004; 91:255.

29. Forastiero R, Martinuzzo M. Prothrombotic mechanisms based on the impairment of fibrinolysis in the antiphospholipid syndrome. Lupus 2008; 17:872.

30. Urbanus RT, Derksen RH, de Groot PG. Platelets and the antiphospholipid syndrome. Lupus 2008; 17:888.

31. Chen PP, Giles I. Antibodies to serine proteases in the antiphospholipid syndrome. Curr Rheumatol Rep 2010; 12:45.

32. Rand JH, Wu XX, Quinn AS, Taatjes DJ. Resistance to annexin A5 anticoagulant activity: a thrombogenic mechanism for the antiphospholipid syndrome. Lupus 2008; 17:922.

33. Raschi E, Borghi MO, Grossi C, et al. Toll-like receptors: another player in the pathogenesis of the anti-phospholipid syndrome. Lupus 2008; 17:937.

34. Cockrell E, Espinola RG, McCrae KR. Annexin A2: biology and relevance to the antiphospholipid syndrome. Lupus 2008; 17:943.

35. Kinev AV, Roubey RA. Tissue factor in the antiphospholipid syndrome. Lupus 2008; 17:952.36. Bu C, Gao L, Xie W, et al. beta2-glycoprotein i is a cofactor for tissue plasminogen activator-

mediated plasminogen activation. Arthritis Rheum 2009; 60:559.37. Mackworth-Young CG. Antiphospholipid syndrome: multiple mechanisms. Clin Exp Immunol 2004;

136:393.38. Erkan D, Lockshin MD. What is antiphospholipid syndrome? Curr Rheumatol Rep 2004; 6:451.39. Merrill JT, Zhang HW, Shen C, et al. Enhancement of protein S anticoagulant function by beta2-

glycoprotein I, a major target antigen of antiphospholipid antibodies: beta2-glycoprotein I interferes with binding of protein S to its plasma inhibitor, C4b-binding protein. Thromb Haemost 1999; 81:748.

40. Shapiro SS. The lupus anticoagulant/antiphospholipid syndrome. Annu Rev Med 1996; 47:533.41. Male C, Mitchell L, Julian J, et al. Acquired activated protein C resistance is associated with lupus

anticoagulants and thrombotic events in pediatric patients with systemic lupus erythematosus. Blood 2001; 97:844.

42. Galli M, Willems GM, Rosing J, et al. Anti-prothrombin IgG from patients with anti-phospholipid antibodies inhibits the inactivation of factor Va by activated protein C. Br J Haematol 2005; 129:240.

43. Hurtado V, Montes R, Gris JC, et al. Autoantibodies against EPCR are found in antiphospholipid syndrome and are a risk factor for fetal death. Blood 2004; 104:1369.

44. Yoshida K, Arai T, Kaburaki J, et al. Restricted T-cell receptor beta-chain usage by T cells autoreactive to beta(2)-glycoprotein I in patients with antiphospholipid syndrome. Blood 2002; 99:2499.

45. De Jong A, Ziboh V, Robbins D. Antiphospholipid antibodies and platelets. Curr Rheumatol Rep 2000; 2:238.

46. Shi T, Giannakopoulos B, Yan X, et al. Anti-beta2-glycoprotein I antibodies in complex with beta2-glycoprotein I can activate platelets in a dysregulated manner via glycoprotein Ib-IX-V. Arthritis Rheum 2006; 54:2558.

47. Vlachoyiannopoulos PG, Mavragani CP, Bourazopoulou E, et al. Anti-CD40 antibodies in antiphospholipid syndrome and systemic lupus erythematosus. Thromb Haemost 2004; 92:1303.

48. Yellin MJ, Brett J, Baum D, et al. Functional interactions of T cells with endothelial cells: the role of CD40L-CD40-mediated signals. J Exp Med 1995; 182:1857.

49. Silver RK, Mullen TA, Caplan MS, et al. Inducible platelet adherence to human umbilical vein endothelium by anticardiolipin antibody-positive sera. Am J Obstet Gynecol 1995; 173:702.

50. Nojima J, Kuratsune H, Suehisa E, et al. Strong correlation between the prevalence of cerebral infarction and the presence of anti-cardiolipin/beta2-glycoprotein I and anti-phosphatidylserine/prothrombin antibodies--Co-existence of these antibodies enhances ADP-induced platelet activation in vitro. Thromb Haemost 2004; 91:967.

51. 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.

52. Simantov R, LaSala JM, Lo SK, et al. Activation of cultured vascular endothelial cells by antiphospholipid antibodies. J Clin Invest 1995; 96:2211.

Page 13: Pathogenesis Antifosfolip Syndrome

53. Del Papa N, Guidali L, Spatola L, et al. Relationship between anti-phospholipid and anti-endothelial cell antibodies III: beta 2 glycoprotein I mediates the antibody binding to endothelial membranes and induces the expression of adhesion molecules. Clin Exp Rheumatol 1995; 13:179.

54. 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.

55. Hamid C, Norgate K, D'Cruz DP, et al. Anti-beta2GPI-antibody-induced endothelial cell gene expression profiling reveals induction of novel pro-inflammatory genes potentially involved in primary antiphospholipid syndrome. Ann Rheum Dis 2007; 66:1000.

56. 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.

57. Salmon JE, Girardi G, Holers VM. Activation of complement mediates antiphospholipid antibody-induced pregnancy loss. Lupus 2003; 12:535.

58. Girardi G, Redecha P, Salmon JE. Heparin prevents antiphospholipid antibody-induced fetal loss by inhibiting complement activation. Nat Med 2004; 10:1222.

59. Shamonki JM, Salmon JE, Hyjek E, Baergen RN. Excessive complement activation is associated with placental injury in patients with antiphospholipid antibodies. Am J Obstet Gynecol 2007; 196:167.e1.

60. Blank M, Cohen J, Toder V, Shoenfeld Y. Induction of anti-phospholipid syndrome in naive mice with mouse lupus monoclonal and human polyclonal anti-cardiolipin antibodies. Proc Natl Acad Sci U S A 1991; 88:3069.

61. Bakimer R, Fishman P, Blank M, et al. Induction of primary antiphospholipid syndrome in mice by immunization with a human monoclonal anticardiolipin antibody (H-3). J Clin Invest 1992; 89:1558.

62. Blank M, Krause I, Fridkin M, et al. Bacterial induction of autoantibodies to beta2-glycoprotein-I accounts for the infectious etiology of antiphospholipid syndrome. J Clin Invest 2002; 109:797.

63. Gharavi AE, Wilson W, Pierangeli S. The molecular basis of antiphospholipid syndrome. Lupus 2003; 12:579.

64. Giles IP, Isenberg DA, Latchman DS, Rahman A. How do antiphospholipid antibodies bind beta2-glycoprotein I? Arthritis Rheum 2003; 48:2111.

65. Oku K, Atsumi T, Bohgaki M, et al. Complement activation in patients with primary antiphospholipid syndrome. Ann Rheum Dis 2009; 68:1030.

66. Cabral AR, Cabiedes J, Alarcón-Segovia D. Antibodies to phospholipid-free beta 2-glycoprotein-I in patients with primary antiphospholipid syndrome. J Rheumatol 1995; 22:1894.

67. Cabiedes J, Cabral AR, Alarcón-Segovia D. Clinical manifestations of the antiphospholipid syndrome in patients with systemic lupus erythematosus associate more strongly with anti-beta 2-glycoprotein-I than with antiphospholipid antibodies. J Rheumatol 1995; 22:1899.

68. Tsutsumi A, Matsuura E, Ichikawa K, et al. Antibodies to beta 2-glycoprotein I and clinical manifestations in patients with systemic lupus erythematosus. Arthritis Rheum 1996; 39:1466.

69. Roubey RA, Maldonado MA, Byrd SN. Comparison of an enzyme-linked immunosorbent assay for antibodies to beta 2-glycoprotein I and a conventional anticardiolipin immunoassay. Arthritis Rheum 1996; 39:1606.

70. Galli M, Finazzi G, Norbis F, et al. The risk of thrombosis in patients with lupus anticoagulants is predicted by their specific coagulation profile. Thromb Haemost 1999; 81:695.

71. Cabral AR, Amigo MC, Cabiedes J, Alarcon-Segovia D. The antiphospholipid/cofactor syndromes: a primary variant with antibodies to beta 2-glycoprotein-I but no antibodies detectable in standard antiphospholipid assays. Am J Med 1996; 101:472.

72. 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.

73. López-Pedrera C, Cuadrado MJ, Herández V, et al. Proteomic analysis in monocytes of antiphospholipid syndrome patients: deregulation of proteins related to the development of thrombosis. Arthritis Rheum 2008; 58:2835.

74. Jones DW, MacKie IJ, Gallimore MJ, Winter M. Antibodies to factor XII and recurrent fetal loss in patients with the anti-phospholipid syndrome. Br J Haematol 2001; 113:550.

75. Bidot CJ, Jy W, Horstman LL, et al. Factor VII/VIIa: a new antigen in the anti-phospholipid antibody syndrome. Br J Haematol 2003; 120:618.

76. 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.

Page 14: Pathogenesis Antifosfolip Syndrome

77. Vaarala O. Antiphospholipid antibodies and atherosclerosis. Lupus 1996; 5:442.78. Wu R, Nityanand S, Berglund L, et al. Antibodies against cardiolipin and oxidatively modified LDL in

50-year-old men predict myocardial infarction. Arterioscler Thromb Vasc Biol 1997; 17:3159.79. Rand JH, Wu XX, Lapinski R, et al. Detection of antibody-mediated reduction of annexin A5

anticoagulant activity in plasmas of patients with the antiphospholipid syndrome. Blood 2004; 104:2783.

80. Gromnica-Ihle E, Schössler W. Antiphospholipid syndrome. Int Arch Allergy Immunol 2000; 123:67.81. Cesarman-Maus G, Ríos-Luna NP, Deora AB, et al. Autoantibodies against the fibrinolytic receptor,

annexin 2, in antiphospholipid syndrome. Blood 2006; 107:4375.82. Satoh A, Suzuki K, Takayama E, et al. Detection of anti-annexin IV and V antibodies in patients with

antiphospholipid syndrome and systemic lupus erythematosus. J Rheumatol 1999; 26:1715.83. Song KS, Park YS, Kim HK. Prevalence of anti-protein S antibodies in patients with systemic lupus

erythematosus. Arthritis Rheum 2000; 43:557.84. Bertolaccini ML, Sanna G, Ralhan S, et al. Antibodies directed to protein S in patients with systemic

lupus erythematosus: prevalence and clinical significance. Thromb Haemost 2003; 90:636.85. Muñoz-Rodríguez FJ, Reverter JC, Font J, et al. Prevalence and clinical significance of

antiprothrombin antibodies in patients with systemic lupus erythematosus or with primary antiphospholipid syndrome. Haematologica 2000; 85:632.

86. Tektonidou MG, Petrovas CA, Ioannidis JP, et al. Clinical importance of antibodies against platelet activating factor in antiphospholipid syndrome manifestations. Eur J Clin Invest 2000; 30:646.

87. 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.

88. Dignat-George F, Camoin-Jau L, Sabatier F, et al. Endothelial microparticles: a potential contribution to the thrombotic complications of the antiphospholipid syndrome. Thromb Haemost 2004; 91:667.

89. Ambrozic A, Bozic B, Kveder T, et al. Budding, vesiculation and permeabilization of phospholipid membranes-evidence for a feasible physiologic role of beta2-glycoprotein I and pathogenic actions of anti-beta2-glycoprotein I antibodies. Biochim Biophys Acta 2005; 1740:38.

90. Morel O, Jesel L, Freyssinet JM, Toti F. Elevated levels of procoagulant microparticles in a patient with myocardial infarction, antiphospholipid antibodies and multifocal cardiac thrombosis. Thromb J 2005; 3:15.

91. Brouwer JL, Bijl M, Veeger NJ, et al. The contribution of inherited and acquired thrombophilic defects, alone or combined with antiphospholipid antibodies, to venous and arterial thromboembolism in patients with systemic lupus erythematosus. Blood 2004; 104:143.

92. Nojima J, Kuratsune H, Suehisa E, et al. Acquired activated protein C resistance is associated with the co-existence of anti-prothrombin antibodies and lupus anticoagulant activity in patients with systemic lupus erythematosus. Br J Haematol 2002; 118:577.

93. Goldberg SN, Conti-Kelly AM, Greco TP. A family study of anticardiolipin antibodies and associated clinical conditions. Am J Med 1995; 99:473.

94. Schur PH. Genetics of systemic lupus erythematosus. Lupus 1995; 4:425.95. Karassa FB, Bijl M, Davies KA, et al. Role of the Fcgamma receptor IIA polymorphism in the

antiphospholipid syndrome: an international meta-analysis. Arthritis Rheum 2003; 48:1930.96. Jiménez S, Tàssies D, Espinosa G, et al. Double heterozygosity polymorphisms for platelet

glycoproteins Ia/IIa and IIb/IIIa increases arterial thrombosis and arteriosclerosis in patients with the antiphospholipid syndrome or with systemic lupus erythematosus. Ann Rheum Dis 2008; 67:835.

97. Hirose N, Williams R, Alberts AR, et al. A role for the polymorphism at position 247 of the beta2-glycoprotein I gene in the generation of anti-beta2-glycoprotein I antibodies in the antiphospholipid syndrome. Arthritis Rheum 1999; 42:1655.

98. Sanchez ML, Katsumata K, Atsumi T, et al. Association of HLA-DM polymorphism with the production of antiphospholipid antibodies. Ann Rheum Dis 2004; 63:1645.

99. Yin H, Borghi MO, Delgado-Vega AM, et al. Association of STAT4 and BLK, but not BANK1 or IRF5, with primary antiphospholipid syndrome. Arthritis Rheum 2009; 60:2468.

100. Love PE, Santoro SA. Antiphospholipid antibodies: anticardiolipin and the lupus anticoagulant in systemic lupus erythematosus (SLE) and in non-SLE disorders. Prevalence and clinical significance. Ann Intern Med 1990; 112:682.

Page 15: Pathogenesis Antifosfolip Syndrome

101. Abu-Shakra M, Gladman DD, Urowitz MB, Farewell V. Anticardiolipin antibodies in systemic lupus erythematosus: clinical and laboratory correlations. Am J Med 1995; 99:624.

102. McNeil HP, Chesterman CN, Krilis SA. Immunology and clinical importance of antiphospholipid antibodies. Adv Immunol 1991; 49:193.

103. Sebastiani GD, Galeazzi M, Tincani A, et al. Anticardiolipin and anti-beta2GPI antibodies in a large series of European patients with systemic lupus erythematosus. Prevalence and clinical associations. European Concerted Action on the Immunogenetics of SLE. Scand J Rheumatol 1999; 28:344.

104. Cervera R, Asherson RA. Clinical and epidemiological aspects in the antiphospholipid syndrome. Immunobiology 2003; 207:5.

105. Triplett DA. Many faces of lupus anticoagulants. Lupus 1998; 7 Suppl 2:S18.106. McNally T, Purdy G, Mackie IJ, et al. The use of an anti-beta 2-glycoprotein-I assay for

discrimination between anticardiolipin antibodies associated with infection and increased risk of thrombosis. Br J Haematol 1995; 91:471.

107. Santiago M, Martinelli R, Ko A, et al. Anti-beta2 glycoprotein I and anticardiolipin antibodies in leptospirosis, syphilis and Kala-azar. Clin Exp Rheumatol 2001; 19:425.

108. García Moncó JC, Wheeler CM, Benach JL, et al. Reactivity of neuroborreliosis patients (Lyme disease) to cardiolipin and gangliosides. J Neurol Sci 1993; 117:206.

109. Prieto J, Yuste JR, Beloqui O, et al. Anticardiolipin antibodies in chronic hepatitis C: implication of hepatitis C virus as the cause of the antiphospholipid syndrome. Hepatology 1996; 23:199.

110. Matsuda J, Saitoh N, Gotoh M, et al. High prevalence of anti-phospholipid antibodies and anti-thyroglobulin antibody in patients with hepatitis C virus infection treated with interferon-alpha. Am J Gastroenterol 1995; 90:1138.

111. Leroy V, Arvieux J, Jacob MC, et al. Prevalence and significance of anticardiolipin, anti-beta2 glycoprotein I and anti-prothrombin antibodies in chronic hepatitis C. Br J Haematol 1998; 101:468.

112. Muñoz-Rodríguez FJ, Tàssies D, Font J, et al. Prevalence of hepatitis C virus infection in patients with antiphospholipid syndrome. J Hepatol 1999; 30:770.

113. Uthman IW, Gharavi AE. Viral infections and antiphospholipid antibodies. Semin Arthritis Rheum 2002; 31:256.

114. Von Landenberg P, Lehmann HW, Knöll A, et al. Antiphospholipid antibodies in pediatric and adult patients with rheumatic disease are associated with parvovirus B19 infection. Arthritis Rheum 2003; 48:1939.

115. Merrill JT, Shen C, Gugnani M, et al. High prevalence of antiphospholipid antibodies in patients taking procainamide. J Rheumatol 1997; 24:1083.

Topic 4676 Version 5.0

© 2012 UpToDate, Inc. All rights reserved. |Subscription and License Agreement|Release: 20.11 - C20.44 Licensed to: Dr Carol Davila Univ|Support Tag: [ecapp1005p.utd.com-46.243.119.65-5C976263A1-499488.14]

TOPIC OUTLINE

SUMMARY

INTRODUCTION ANTIPHOSPHOLIPID ANTIBODIES Anticardiolipin antibodies - False positive serologic test for syphilis Lupus anticoagulants - Prolongation of clotting times - Abnormal bleeding times Anti-ß2-glycoprotein-I antibodies Additional aPL - Antiprothrombin antibodies PATHOGENESIS In vitro experiments

Page 16: Pathogenesis Antifosfolip Syndrome

Animal models Clinical studies Interaction with other antibodies Microparticles GENETIC PREDISPOSITION PREVALENCE IN DIFFERENT CONDITIONS Healthy individuals Autoimmune and rheumatic diseases Infections Medications Neoplasms Other associations SUMMARY REFERENCES

RELATED TOPICS

Acquired inhibitors of coagulation Clinical manifestations of the antiphospholipid syndrome Clinical use of coagulation tests Diagnosis of the antiphospholipid syndrome Diagnostic testing for syphilis Endothelial dysfunction Obstetrical manifestations of the antiphospholipid syndrome Protein C deficiency Protein S deficiency Treatment of the antiphospholipid syndrome

Topic Feedbackclose