Skip to main content

Advertisement

Log in

Lymphocyte Disturbances in Primary Antiphospholipid Syndrome and Application to Venous Thromboembolism Follow-Up

  • Published:
Clinical Reviews in Allergy & Immunology Aims and scope Submit manuscript

Abstract

Among patients with venous thromboembolism (VTE), the persistent detection of antiphospholipid (aPL) antibodies (Ab) represents an independent high risk factor for recurrence. However, oral anticoagulation vitamin K antagonist therapy, frequently used in these patients, is problematic in assessing and/or confirming a diagnosis of primary aPL syndrome (pAPS), suggesting use of alternative strategies. For this reason, and by analogy with other autoimmune diseases, a flow cytometer approach testing peripheral T cell subsets (CD3, CD4, and CD8), B cell subsets (B1, transitional, naive, and memory), and NK cells can be proposed. As an example and to validate the concept, pAPS patients selected from the monocentric VTE case–control EDITH’s cohort were selected during their follow-up. As suspected and in contrast to non-APS VTE patients, other autoimmune diseases, and controls, pAPS VTE patients displayed specific lymphocyte disturbances. Quantitative and qualitative modifications were related to total CD4+ T cell reduction, a lower CD4/CD8 ratio, and disturbance in B cell homeostasis with increased proportions of B1 cells, transitional B cells (CD24++CD38++), and naive B cells (IgD+CD27), while memory B cells (IgD+CD27+ and IgDCD27+) were reduced. Interestingly, the absolute number of CD4+ T cells positively correlated with IgG anti-cardiolipin Ab levels. Altogether, disturbances of T and B cell homeostasis characterized pAPS VTE patients during their follow-up. This suggests a means of profiling that could be used in addition to existing criteria to characterize them.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Harris EN, Gharavi AE, Boey ML, Patel BM, Mackworth-Young CG, Loizou S, Hughes GR (1983) Anticardiolipin antibodies: detection by radioimmunoassay and association with thrombosis in systemic lupus erythematosus. Lancet 2(8361):1211–1214

    Article  CAS  PubMed  Google Scholar 

  2. Miyakis S, Lockshin MD, Atsumi T, Branch DW, Brey RL, Cervera R, Derksen RH, De Groot PG, Koike T, Meroni PL, Reber G, Shoenfeld Y, Tincani A, Vlachoyiannopoulos PG, Krilis SA (2006) International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS). J Thromb Haemost 4(2):295–306. doi:10.1111/j.1538-7836.2006.01753.x

    Article  CAS  PubMed  Google Scholar 

  3. Brooks WH, Le Dantec C, Pers JO, Youinou P, Renaudineau Y (2010) Epigenetics and autoimmunity. J Autoimmun 34(3):J207–219. doi:10.1016/j.jaut.2009.12.006

    Article  CAS  PubMed  Google Scholar 

  4. Konsta OD, Le Dantec C, Charras A, Brooks WH, Arleevskaya MI, Bordron A, Renaudineau Y (2015) An in silico approach reveals associations between genetic and epigenetic factors within regulatory elements in B cells from primary Sjogren’s syndrome patients. Front Immunol 6:437. doi:10.3389/fimmu.2015.00437

    Article  PubMed  PubMed Central  Google Scholar 

  5. Canas F, Simonin L, Couturaud F, Renaudineau Y (2015) Annexin A2 autoantibodies in thrombosis and autoimmune diseases. Thromb Res 135(2):226–230. doi:10.1016/j.thromres.2014.11.034

    Article  CAS  PubMed  Google Scholar 

  6. Gomez-Puerta JA, Cervera R (2014) Diagnosis and classification of the antiphospholipid syndrome. J Autoimmun 48–49:20–25. doi:10.1016/j.jaut.2014.01.006

    Article  PubMed  Google Scholar 

  7. Sebastiani GD, Iuliano A, Cantarini L, Galeazzi M (2016) Genetic aspects of the antiphospholipid syndrome: an update. Autoimmun Rev 15:433–439. doi:10.1016/j.autrev.2016.01.005

    Article  CAS  PubMed  Google Scholar 

  8. Garcia-Carrasco M, Mendoza-Pinto C, Macias-Diaz S, Vazquez de Lara F, Etchegaray-Morales I, Galvez-Romero JL, Mendez-Martinez S, Cervera R (2015) The role of infectious diseases in the catastrophic antiphospholipid syndrome. Autoimmun Rev 14(11):1066–1071. doi:10.1016/j.autrev.2015.07.009

    Article  CAS  PubMed  Google Scholar 

  9. Garcia-Carrasco M, Mendoza-Pinto C, Macias Diaz S, Vera-Recabarren M, Vazquez de Lara L, Mendez Martinez S, Soto-Santillan P, Gonzalez-Ramirez R, Ruiz-Arguelles A (2015) P-glycoprotein in autoimmune rheumatic diseases. Autoimmun Rev 14(7):594–600. doi:10.1016/j.autrev.2015.02.006

    Article  CAS  PubMed  Google Scholar 

  10. Cervera R, Khamashta MA, Shoenfeld Y, Camps MT, Jacobsen S, Kiss E, Zeher MM, Tincani A, Kontopoulou-Griva I, Galeazzi M, Bellisai F, Meroni PL, Derksen RH, de Groot PG, Gromnica-Ihle E, Baleva M, Mosca M, Bombardieri S, Houssiau F, Gris JC, Quere I, Hachulla E, Vasconcelos C, Roch B, Fernandez-Nebro A, Piette JC, Espinosa G, Bucciarelli S, Pisoni CN, Bertolaccini ML, Boffa MC, Hughes GR (2009) Morbidity and mortality in the antiphospholipid syndrome during a 5-year period: a multicentre prospective study of 1000 patients. Ann Rheum Dis 68(9):1428–1432. doi:10.1136/ard.2008.093179

    Article  CAS  PubMed  Google Scholar 

  11. Cervera R, Serrano R, Pons-Estel GJ, Ceberio-Hualde L, Shoenfeld Y, de Ramon E, Buonaiuto V, Jacobsen S, Zeher MM, Tarr T, Tincani A, Taglietti M, Theodossiades G, Nomikou E, Galeazzi M, Bellisai F, Meroni PL, Derksen RH, de Groot PG, Baleva M, Mosca M, Bombardieri S, Houssiau F, Gris JC, Quere I, Hachulla E, Vasconcelos C, Fernandez-Nebro A, Haro M, Amoura Z, Miyara M, Tektonidou M, Espinosa G, Bertolaccini ML, Khamashta MA (2014) Morbidity and mortality in the antiphospholipid syndrome during a 10-year period: a multicentre prospective study of 1000 patients. Ann Rheum Dis. doi:10.1136/annrheumdis-2013-204838

    Google Scholar 

  12. Dlott JS, Roubey RA (2012) Drug-induced lupus anticoagulants and antiphospholipid antibodies. Curr Rheumatol Rep 14(1):71–78. doi:10.1007/s11926-011-0227-1

    Article  CAS  PubMed  Google Scholar 

  13. Kearon C, Gent M, Hirsh J, Weitz J, Kovacs MJ, Anderson DR, Turpie AG, Green D, Ginsberg JS, Wells P, MacKinnon B, Julian JA (1999) A comparison of three months of anticoagulation with extended anticoagulation for a first episode of idiopathic venous thromboembolism. N Engl J Med 340(12):901–907. doi:10.1056/NEJM199903253401201

    Article  CAS  PubMed  Google Scholar 

  14. Ruiz-Irastorza G, Crowther M, Branch W, Khamashta MA (2010) Antiphospholipid syndrome. Lancet 376(9751):1498–1509. doi:10.1016/S0140-6736(10)60709-X

    Article  CAS  PubMed  Google Scholar 

  15. Bertolaccini ML, Amengual O, Andreoli L, Atsumi T, Chighizola CB, Forastiero R, de Groot P, Lakos G, Lambert M, Meroni P, Ortel TL, Petri M, Rahman A, Roubey R, Sciascia S, Snyder M, Tebo AE, Tincani A, Willis R (2014) 14th International Congress on Antiphospholipid Antibodies Task Force. Report on antiphospholipid syndrome laboratory diagnostics and trends. Autoimmun Rev 13(9):917–930. doi:10.1016/j.autrev.2014.05.001

    Article  PubMed  Google Scholar 

  16. Reynaud Q, Lega JC, Mismetti P, Chapelle C, Wahl D, Cathebras P, Laporte S (2014) Risk of venous and arterial thrombosis according to type of antiphospholipid antibodies in adults without systemic lupus erythematosus: a systematic review and meta-analysis. Autoimmun Rev 13(6):595–608. doi:10.1016/j.autrev.2013.11.004

    Article  CAS  PubMed  Google Scholar 

  17. Youinou P, Renaudineau Y (2004) The antiphospholipid syndrome as a model for B cell-induced autoimmune diseases. Thromb Res 114(5–6):363–369. doi:10.1016/j.thromres.2004.06.019

    Article  CAS  PubMed  Google Scholar 

  18. Carbone J, Gallego A, Lanio N, Navarro J, Orera M, Aguaron A, Fernandez-Cruz E, Sarmiento E (2009) Quantitative abnormalities of peripheral blood distinct T, B, and natural killer cell subsets and clinical findings in obstetric antiphospholipid syndrome. J Rheumatol 36(6):1217–1225. doi:10.3899/jrheum.081079

    Article  CAS  PubMed  Google Scholar 

  19. Cornec D, Saraux A, Pers JO, Jousse-Joulin S, Marhadour T, Roguedas-Contios AM, Genestet S, Renaudineau Y, Devauchelle-Pensec V (2014) Diagnostic accuracy of blood B-cell subset profiling and autoimmunity markers in Sjogren’s syndrome. Arthritis Res Ther 16(1):R15. doi:10.1186/ar4442

    Article  PubMed  PubMed Central  Google Scholar 

  20. Hajas A, Barath S, Szodoray P, Nakken B, Gogolak P, Szekanecz Z, Zold E, Zeher M, Szegedi G, Bodolay E (2013) Derailed B cell homeostasis in patients with mixed connective tissue disease. Hum Immunol 74(7):833–841. doi:10.1016/j.humimm.2013.04.007

    Article  CAS  PubMed  Google Scholar 

  21. Le Coz C, Joublin A, Pasquali JL, Korganow AS, Dumortier H, Monneaux F (2013) Circulating TFH subset distribution is strongly affected in lupus patients with an active disease. PLoS One 8(9):e75319. doi:10.1371/journal.pone.0075319

    Article  PubMed  PubMed Central  Google Scholar 

  22. Thiel J, Salzer U, Hassler F, Effelsberg NM, Hentze C, Sic H, Bartsch M, Miehle N, Peter HH, Warnatz K, Schlesier M, Voll RE, Venhoff N (2013) B cell homeostasis is disturbed by immunosuppressive therapies in patients with ANCA-associated vasculitides. Autoimmunity 46(7):429–438. doi:10.3109/08916934.2013.798652

    Article  CAS  PubMed  Google Scholar 

  23. Cornec D, Saraux A, Cochener B, Pers JO, Jousse-Joulin S, Renaudineau Y, Marhadour T, Devauchelle-Pensec V (2014) Level of agreement between 2002 American-European Consensus Group and 2012 American College of Rheumatology classification criteria for Sjogren’s syndrome and reasons for discrepancies. Arthritis Res Ther 16(2):R74. doi:10.1186/ar4514

    Article  PubMed  PubMed Central  Google Scholar 

  24. Gazeau P, Devauchelle-Pensec V, Pochard P, Pers JO, Saraux A, Renaudineau Y, Cornec D (2016) Abatacept efficacy in rheumatoid arthritis is dependent upon baseline blood B-cell levels. Rheumatology (Oxford) 55:1138–1140. doi:10.1093/rheumatology/kev417

    Article  Google Scholar 

  25. Cornec D, Saraux A, Jousse-Joulin S, Pers JO, Boisrame-Gastrin S, Renaudineau Y, Gauvin Y, Roguedas-Contios AM, Genestet S, Chastaing M, Cochener B, Devauchelle-Pensec V (2015) The differential diagnosis of dry eyes, dry mouth, and parotidomegaly: a comprehensive review. Clin Rev Allergy Immunol 49:278–287. doi:10.1007/s12016-014-8431-1

    Article  PubMed  Google Scholar 

  26. Nouel A, Segalen I, Jamin C, Doucet L, Caillard S, Renaudineau Y, Pers JO, Le Meur Y, Hillion S (2014) B cells display an abnormal distribution and an impaired suppressive function in patients with chronic antibody-mediated rejection. Kidney Int 85(3):590–599. doi:10.1038/ki.2013.457

    Article  CAS  PubMed  Google Scholar 

  27. Le Dantec C, Alonso R, Fali T, Montero E, Devauchelle V, Saraux A, Pers JO, Renaudineau Y (2013) Rationale for treating primary Sjogren’s syndrome patients with an anti-CD6 monoclonal antibody (Itolizumab). Immunol Res 56(2–3):341–347. doi:10.1007/s12026-013-8423-x

    Article  CAS  PubMed  Google Scholar 

  28. Shirota Y, Yarboro C, Fischer R, Pham TH, Lipsky P, Illei GG (2013) Impact of anti-interleukin-6 receptor blockade on circulating T and B cell subsets in patients with systemic lupus erythematosus. Ann Rheum Dis 72(1):118–128. doi:10.1136/annrheumdis-2012-201310

    Article  CAS  PubMed  Google Scholar 

  29. Maldonado A, Mueller YM, Thomas P, Bojczuk P, O’Connors C, Katsikis PD (2003) Decreased effector memory CD45RA+ CD62L CD8+ T cells and increased central memory CD45RA CD62L+ CD8+ T cells in peripheral blood of rheumatoid arthritis patients. Arthritis Res Ther 5(2):R91–96

    Article  PubMed  PubMed Central  Google Scholar 

  30. Sudzius G, Mieliauskaite D, Siaurys A, Viliene R, Butrimiene I, Characiejus D, Dumalakiene I (2015) Distribution of peripheral lymphocyte populations in primary Sjogren’s syndrome patients. J Immunol Res 2015:854706. doi:10.1155/2015/854706

    Article  PubMed  PubMed Central  Google Scholar 

  31. Almeida I, Silva SV, Fonseca AR, Silva I, Vasconcelos C, Lima M (2015) T and NK cell phenotypic abnormalities in systemic sclerosis: a cohort study and a comprehensive literature review. Clin Rev Allergy Immunol 49(3):347–369. doi:10.1007/s12016-015-8505-8

    Article  CAS  PubMed  Google Scholar 

  32. Mellergard J, Edstrom M, Jenmalm MC, Dahle C, Vrethem M, Ernerudh J (2013) Increased B cell and cytotoxic NK cell proportions and increased T cell responsiveness in blood of natalizumab-treated multiple sclerosis patients. PLoS One 8(12):e81685. doi:10.1371/journal.pone.0081685

    Article  PubMed  PubMed Central  Google Scholar 

  33. Abdulahad WH, van der Geld YM, Stegeman CA, Kallenberg CG (2006) Persistent expansion of CD4+ effector memory T cells in Wegener’s granulomatosis. Kidney Int 70(5):938–947. doi:10.1038/sj.ki.5001670

    Article  CAS  PubMed  Google Scholar 

  34. KDIGO Board Members (2012) Kidney Int Suppl (2011) 2(1):3. doi:10.1038/kisup.2012.3

    Article  Google Scholar 

  35. Fali T, Le Dantec C, Thabet Y, Jousse S, Hanrotel C, Youinou P, Brooks WH, Perl A, Renaudineau Y (2014) DNA methylation modulates HRES1/p28 expression in B cells from patients with lupus. Autoimmunity 47(4):265–271. doi:10.3109/08916934.2013.826207

    Article  CAS  PubMed  Google Scholar 

  36. Le Dantec C, Vallet S, Brooks WH, Renaudineau Y (2015) Human endogenous retrovirus group E and its involvement in diseases. Viruses 7(3):1238–1257. doi:10.3390/v7031238

    Article  PubMed  PubMed Central  Google Scholar 

  37. Taher TE, Muhammad HA, Bariller E, Flores-Borja F, Renaudineau Y, Isenberg DA, Mageed RA (2013) B-lymphocyte signalling abnormalities and lupus immunopathology. Int Rev Immunol 32(4):428–444. doi:10.3109/08830185.2013.788648

    Article  CAS  PubMed  Google Scholar 

  38. Taher TE, Muhammad HA, Rahim A, Flores-Borja F, Renaudineau Y, Isenberg DA, Mageed RA (2013) Aberrant B-lymphocyte responses in lupus: inherent or induced and potential therapeutic targets. Eur J Clin Investig 43(8):866–880. doi:10.1111/eci.12111

    Article  CAS  Google Scholar 

  39. Garaud S, Youinou P, Renaudineau Y (2011) DNA methylation and B-cell autoreactivity. Adv Exp Med Biol 711:50–60

    Article  CAS  PubMed  Google Scholar 

  40. Garaud S, Le Dantec C, Jousse-Joulin S, Hanrotel-Saliou C, Saraux A, Mageed RA, Youinou P, Renaudineau Y (2009) IL-6 modulates CD5 expression in B cells from patients with lupus by regulating DNA methylation. J Immunol 182(9):5623–5632. doi:10.4049/jimmunol.0802412

    Article  CAS  PubMed  Google Scholar 

  41. Renaudineau Y, JO P, Youinou P (2013) Lymphocytotoxic autoantibodies, vol Chapter 63. Elsevier edn

  42. Oger E, Lacut K, Le Gal G, Couturaud F, Guenet D, Abalain JH, Roguedas AM, Mottier D (2006) Hyperhomocysteinemia and low B vitamin levels are independently associated with venous thromboembolism: results from the EDITH Study: a hospital-based case-control study. J Thromb Haemost 4(4):793–799. doi:10.1111/j.1538-7836.2006.01856.x

    Article  CAS  PubMed  Google Scholar 

  43. Tan EM, Cohen AS, Fries JF, Masi AT, McShane DJ, Rothfield NF, Schaller JG, Talal N, Winchester RJ (1982) The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 25(11):1271–1277

    Article  CAS  PubMed  Google Scholar 

  44. Vitali C, Bombardieri S, Jonsson R, Moutsopoulos HM, Alexander EL, Carsons SE, Daniels TE, Fox PC, Fox RI, Kassan SS, Pillemer SR, Talal N, Weisman MH, European Study Group on Classification Criteria for Sjogren’s Syndrome (2002) Classification criteria for Sjogren’s syndrome: a revised version of the European criteria proposed by the American-European Consensus Group. Ann Rheum Dis 61(6):554–558

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yu C, Gershwin ME, Chang C (2014) Diagnostic criteria for systemic lupus erythematosus: a critical review. J Autoimmun 48–49:10–13. doi:10.1016/j.jaut.2014.01.004

    Article  PubMed  Google Scholar 

  46. Kourilovitch M, Galarza-Maldonado C, Ortiz-Prado E (2014) Diagnosis and classification of rheumatoid arthritis. J Autoimmun 48–49:26–30. doi:10.1016/j.jaut.2014.01.027

    Article  PubMed  Google Scholar 

  47. Goules AV, Tzioufas AG, Moutsopoulos HM (2014) Classification criteria of Sjogren’s syndrome. J Autoimmun 48–49:42–45. doi:10.1016/j.jaut.2014.01.013

    Article  PubMed  Google Scholar 

  48. Brandt JT, Triplett DA, Alving B, Scharrer I (1995) Criteria for the diagnosis of lupus anticoagulants: an update. On behalf of the Subcommittee on Lupus Anticoagulant/Antiphospholipid Antibody of the Scientific and Standardisation Committee of the ISTH. Thromb Haemost 74(4):1185–1190

    CAS  PubMed  Google Scholar 

  49. Renaudineau Y, Dalekos GN, Gueguen P, Zachou K, Youinou P (2008) Anti-alpha-actinin antibodies cross-react with anti-ssDNA antibodies in active autoimmune hepatitis. Clin Rev Allergy Immunol 34(3):321–325. doi:10.1007/s12016-007-8050-1

    Article  CAS  PubMed  Google Scholar 

  50. Pregnolato F, Chighizola CB, Encabo S, Shums Z, Norman GL, Tripodi A, Chantarangkul V, Bertero T, De Micheli V, Borghi MO, Meroni PL (2013) Anti-phosphatidylserine/prothrombin antibodies: an additional diagnostic marker for APS? Immunol Res 56(2-3):432–438. doi:10.1007/s12026-013-8421-z

    Article  CAS  PubMed  Google Scholar 

  51. Beauvillard D, Ségalen I, Le Meur Y, Leroyer C, Renaudineau Y, Youinou P (2011) Auto-anticorps anti-membrane basale glomérulaire et syndrome de Goodpasture. Immuno-analyse & Biologie Spécialisée 26(2):60–67. doi:10.1016/j.immbio.2010.10.002

    Article  Google Scholar 

  52. Renaudineau Y, Croquefer S, Jousse S, Renaudineau E, Devauchelle V, Gueguen P, Hanrotel C, Gilburd B, Saraux A, Shoenfeld Y, Putterman C, Youinou P (2006) Association of alpha-actinin-binding anti-double-stranded DNA antibodies with lupus nephritis. Arthritis Rheum 54(8):2523–2532. doi:10.1002/art.22015

    Article  CAS  PubMed  Google Scholar 

  53. Reber G, Arvieux J, Comby E, Degenne D, de Moerloose P, Sanmarco M, Potron G (1995) Multicenter evaluation of nine commercial kits for the quantitation of anticardiolipin antibodies. The Working Group on Methodologies in Haemostasis from the GEHT (Groupe d’Etudes sur l’Hemostase et la Thrombose). Thromb Haemost 73(3):444–452

    CAS  PubMed  Google Scholar 

  54. Arvieux J, Renaudineau Y, Mane I, Perraut R, Krilis SA, Youinou P (2002) Distinguishing features of anti-beta2 glycoprotein I antibodies between patients with leprosy and the antiphospholipid syndrome. Thromb Haemost 87(4):599–605

    CAS  PubMed  Google Scholar 

  55. Renaudineau Y, Grunebaum E, Krause I, Praprotnik S, Revelen R, Youinou P, Blanks M, Gilburd B, Sherer Y, Luderschmidt C, Eldor A, Weksler B, Gershwin EM, Shoenfeld Y (2001) Anti-endothelial cell antibodies (AECA) in systemic sclerosis—increased sensitivity using different endothelial cell substrates and association with other autoantibodies. Autoimmunity 33(3):171–179

    Article  CAS  PubMed  Google Scholar 

  56. George J, Aron A, Levy Y, Gilburd B, Ben-David A, Renaudineau Y, Zonana-Nachach A, Youinou P, Harats D, Shoenfeld Y (1999) Anti-cardiolipin, anti-endothelial-cell and anti-malondialdehyde-LDL antibodies in uremic patients undergoing hemodialysis: relationship with vascular access thrombosis and thromboembolic events. Hum Antibodies 9(2):125–131

    CAS  PubMed  Google Scholar 

  57. Kornberg A, Renaudineau Y, Blank M, Youinou P, Shoenfeld Y (2000) Anti-beta 2-glycoprotein I antibodies and anti-endothelial cell antibodies induce tissue factor in endothelial cells. Isr Med Assoc J 2(Suppl):27–31

    PubMed  Google Scholar 

  58. Croquefer S, Renaudineau Y, Jousse S, Gueguen P, Ansart S, Saraux A, Youinou P (2005) The ananti-alpha-actinin test completes ananti-DNA determination in systemic lupus erythematosus. Ann N Y Acad Sci 1050:170–175. doi:10.1196/annals.1313.017

    Article  CAS  PubMed  Google Scholar 

  59. Konsta OD, Le Dantec C, Charras A, Cornec D, Kapsogeorgou EK, Tzioufas AG, Pers JO, Renaudineau Y (2016) Defective DNA methylation in salivary gland epithelial acini from patients with Sjogren’s syndrome is associated with SSB gene expression, anti-SSB/LA detection, and lymphocyte infiltration. J Autoimmun 68:30–38. doi:10.1016/j.jaut.2015.12.002

    Article  CAS  PubMed  Google Scholar 

  60. Seret G, Canas F, Pougnet-Di Costanzo L, Hanrotel-Saliou C, Jousse-Joulin S, Le Meur Y, Saraux A, Valeri A, Putterman C, Youinou P, Rojas-Villarraga A, Anaya JM, Renaudineau Y (2015) Anti-alpha-actinin antibodies are part of the anti-cell membrane antibody spectrum that characterize patients with lupus nephritis. J Autoimmun 61:54–61. doi:10.1016/j.jaut.2015.05.009

    Article  CAS  PubMed  Google Scholar 

  61. Capaldo C, Carvajal Alegria G, Cornec D, Jousse-Joulin S, Devauchelle-Pensec V, Renaudineau Y (2016) The active immunological profile in patients with primary Sjogren’s syndrome is restricted to typically encountered autoantibodies. Clin Exp Rheumatol (in press)

  62. Agmon-Levin N, Shapira Y, Selmi C, Barzilai O, Ram M, Szyper-Kravitz M, Sella S, Katz BS, Youinou P, Renaudineau Y, Larida B, Invernizzi P, Gershwin ME, Shoenfeld Y (2010) A comprehensive evaluation of serum autoantibodies in primary biliary cirrhosis. J Autoimmun 34(1):55–58. doi:10.1016/j.jaut.2009.08.009

    Article  CAS  PubMed  Google Scholar 

  63. Pengo V, Banzato A, Denas G, Jose SP, Bison E, Hoxha A, Ruffatti A (2013) Correct laboratory approach to APS diagnosis and monitoring. Autoimmun Rev 12(8):832–834. doi:10.1016/j.autrev.2012.11.008

    Article  CAS  PubMed  Google Scholar 

  64. Ames PR, Tommasino C, Fossati G, Matsuura E, Margarita A, Saulino A, Lopez L, Scenna G, Brancaccio V (2005) Lymphocyte subpopulations and intima media thickness in primary antiphospholipd syndrome. Lupus 14(10):809–813

    Article  CAS  PubMed  Google Scholar 

  65. Arai T, Yoshida K, Kaburaki J, Inoko H, Ikeda Y, Kawakami Y, Kuwana M (2001) Autoreactive CD4(+) T-cell clones to beta2-glycoprotein I in patients with antiphospholipid syndrome: preferential recognition of the major phospholipid-binding site. Blood 98(6):1889–1896

    Article  CAS  PubMed  Google Scholar 

  66. Dal Ben ER, do Prado CH, Baptista TS, Bauer ME, Staub HL (2013) Decreased levels of circulating CD4+CD25+Foxp3+ regulatory T cells in patients with primary antiphospholipid syndrome. J Clin Immunol 33(4):876–879. doi:10.1007/s10875-012-9857-y

    Article  CAS  PubMed  Google Scholar 

  67. Tomer Y, Blank M, Shoenfeld Y (1994) Suppression of experimental antiphospholipid syndrome and systemic lupus erythematosus in mice by anti-CD4 monoclonal antibodies. Arthritis Rheum 37(8):1236–1244

    Article  CAS  PubMed  Google Scholar 

  68. Akkerman A, Huang W, Wang X, Ramanujam M, Schiffer L, Madaio M, Factor SM, Davidson A (2004) CTLA4Ig prevents initiation but not evolution of anti-phospholipid syndrome in NZW/BXSB mice. Autoimmunity 37(6–7):445–451. doi:10.1080/08916930400008524

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Renaudineau Y, Bariller E, JO P (2014) B1- and CD5-positive B cells. eLS. Wiley, Chichester. doi:10.1002/9780470015902.a0024242

  70. Mageed RA, Garaud S, Taher TE, Parikh K, Pers JO, Jamin C, Renaudineau Y, Youinou P (2012) CD5 expression promotes multiple intracellular signaling pathways in B lymphocyte. Autoimmun Rev 11:795–798. doi:10.1016/j.autrev.2012.02.007

    Article  CAS  PubMed  Google Scholar 

  71. Mahmoud F, Diejomaoh M, Omu AE, Abul H, Haines D (2001) Lymphocyte subpopulation frequency and presence of anti-cardiolipin and anti-nuclear antibodies in peripheral blood of Kuwaiti women experiencing recurrent pregnancy loss. J Obstet Gynaecol 21(6):587–590. doi:10.1080/01443610120087805

    Article  CAS  PubMed  Google Scholar 

  72. Sims GP, Ettinger R, Shirota Y, Yarboro CH, Illei GG, Lipsky PE (2005) Identification and characterization of circulating human transitional B cells. Blood 105(11):4390–4398. doi:10.1182/blood-2004-11-4284

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Cuss AK, Avery DT, Cannons JL, Yu LJ, Nichols KE, Shaw PJ, Tangye SG (2006) Expansion of functionally immature transitional B cells is associated with human-immunodeficient states characterized by impaired humoral immunity. J Immunol 176(3):1506–1516

    Article  CAS  PubMed  Google Scholar 

  74. Palanichamy A, Barnard J, Zheng B, Owen T, Quach T, Wei C, Looney RJ, Sanz I, Anolik JH (2009) Novel human transitional B cell populations revealed by B cell depletion therapy. J Immunol 182(10):5982–5993. doi:10.4049/jimmunol.0801859

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Alonso R, Buors C, Le Dantec C, Hillion S, Pers JO, Saraux A, Montero E, Marianowski R, Loisel S, Devauchelle V, Youinou P, Renaudineau Y (2010) Aberrant expression of CD6 on B-cell subsets from patients with Sjogren’s syndrome. J Autoimmun 35(4):336–341. doi:10.1016/j.jaut.2010.07.005

    Article  CAS  PubMed  Google Scholar 

  76. Lemoine S, Morva A, Youinou P, Jamin C (2011) Human T cells induce their own regulation through activation of B cells. J Autoimmun 36(3–4):228–238. doi:10.1016/j.jaut.2011.01.005

    Article  CAS  PubMed  Google Scholar 

  77. Nouel A, Pochard P, Simon Q, Segalen I, Le Meur Y, Pers JO, Hillion S (2015) B-cells induce regulatory T cells through TGF-beta/IDO production in A CTLA-4 dependent manner. J Autoimmun 59:53–60. doi:10.1016/j.jaut.2015.02.004

    Article  CAS  PubMed  Google Scholar 

  78. Chang NH, Li TT, Kim JJ, Landolt-Marticorena C, Fortin PR, Gladman DD, Urowitz MB, Wither JE (2015) Interferon-alpha induces altered transitional B cell signaling and function in systemic lupus erythematosus. J Autoimmun 58:100–110. doi:10.1016/j.jaut.2015.01.009

    Article  CAS  PubMed  Google Scholar 

  79. Simon Q, Pers JO, Cornec D, Le Pottier L, Mageed RA, Hillion S (2016) In-depth characterization of CD24highCD38high transitional human B cells reveals different regulatory profiles. J Allergy Clin Immunol 137:1577–1584.e10. doi:10.1016/j.jaci.2015.09.014

    Article  CAS  PubMed  Google Scholar 

  80. Al Kindi M, Mundy J, Sullivan T, Smith W, Kette F, Smith A, Heddle R, Hissaria P (2012) Utility of peripheral blood B cell subsets analysis in common variable immunodeficiency. Clin Exp Immunol 167(2):275–281. doi:10.1111/j.1365-2249.2011.04507.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Wasserstrom H, Bussel J, Lim LC, Cunningham-Rundles C (2008) Memory B cells and pneumococcal antibody after splenectomy. J Immunol 181(5):3684–3689

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Binard A, Le Pottier L, Devauchelle-Pensec V, Saraux A, Youinou P, Pers JO (2009) Is the blood B-cell subset profile diagnostic for Sjogren syndrome? Ann Rheum Dis 68(9):1447–1452. doi:10.1136/ard.2008.096172

    Article  CAS  PubMed  Google Scholar 

  83. Bohnhorst JO, Thoen JE, Natvig JB, Thompson KM (2001) Significantly depressed percentage of CD27+ (memory) B cells among peripheral blood B cells in patients with primary Sjogren’s syndrome. Scand J Immunol 54(4):421–427

    Article  CAS  PubMed  Google Scholar 

  84. Martinez-Gamboa L, Mei H, Loddenkemper C, Ballmer B, Hansen A, Lipsky PE, Emmerich F, Radbruch A, Salama A, Dorner T (2009) Role of the spleen in peripheral memory B-cell homeostasis in patients with autoimmune thrombocytopenia purpura. Clin Immunol 130(2):199–212. doi:10.1016/j.clim.2008.09.009

    Article  CAS  PubMed  Google Scholar 

  85. Weller S, Braun MC, Tan BK, Rosenwald A, Cordier C, Conley ME, Plebani A, Kumararatne DS, Bonnet D, Tournilhac O, Tchernia G, Steiniger B, Staudt LM, Casanova JL, Reynaud CA, Weill JC (2004) Human blood IgM “memory” B cells are circulating splenic marginal zone B cells harboring a prediversified immunoglobulin repertoire. Blood 104(12):3647–3654. doi:10.1182/blood-2004-01-0346

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Alijotas-Reig J, Ferrer-Oliveras R, Ruffatti A, Tincani A, Lefkou E, Bertero MT, Coloma-Bazan E, de Carolis S, Espinosa G, Rovere-Querini P, Kuzenko A, Valverde EE, Robles A, Cervera R, Canti V, Fredi M, Gil-Aguado A, Lundelin K, Llurba E, Melnychuk T, Nalli C, Picardo E, Silvestro E, del Ross T, Farran-Codina I (2015) The European Registry on Obstetric Antiphospholipid Syndrome (EUROAPS): a survey of 247 consecutive cases. Autoimmun Rev 14(5):387–395. doi:10.1016/j.autrev.2014.12.010

    Article  PubMed  Google Scholar 

  87. Ostensen M, Andreoli L, Brucato A, Cetin I, Chambers C, Clowse ME, Costedoat-Chalumeau N, Cutolo M, Dolhain R, Fenstad MH, Forger F, Wahren-Herlenius M, Ruiz-Irastorza G, Koksvik H, Nelson-Piercy C, Shoenfeld Y, Tincani A, Villiger PM, Wallenius M, von Wolff M (2015) State of the art: reproduction and pregnancy in rheumatic diseases. Autoimmun Rev 14(5):376–386. doi:10.1016/j.autrev.2014.12.011

    Article  PubMed  Google Scholar 

  88. Viall CA, Chamley LW (2015) Histopathology in the placentae of women with antiphospholipid antibodies: a systematic review of the literature. Autoimmun Rev 14(5):446–471. doi:10.1016/j.autrev.2015.01.008

    Article  CAS  PubMed  Google Scholar 

  89. van den Hoogen LL, van Roon JA, Radstake TR, Fritsch-Stork RD, Derksen RH (2016) Delineating the deranged immune system in the antiphospholipid syndrome. Autoimmun Rev 15(1):50–60. doi:10.1016/j.autrev.2015.08.011

    Article  PubMed  Google Scholar 

  90. Carbone J, Chean C, Lanio N, Gallego A, del Pozo N, Sarmiento E (2010) Peripheral blood lymphocyte subset abnormalities in antiphospholipid syndrome. J Clin Rheumatol Musculoskel Med (in press)

  91. Arnaud L, Mathian A, Devilliers H, Ruffatti A, Tektonidou M, Forastiero R, Pengo V, Lambert M, Lefevre G, Martinez-Zamora MA, Balasch J, Wahl D, Amoura Z (2015) Patient-level analysis of five international cohorts further confirms the efficacy of aspirin for the primary prevention of thrombosis in patients with antiphospholipid antibodies. Autoimmun Rev 14(3):192–200. doi:10.1016/j.autrev.2014.10.019

    Article  CAS  PubMed  Google Scholar 

  92. Belizna C (2015) Hydroxychloroquine as an anti-thrombotic in antiphospholipid syndrome. Autoimmun Rev 14(4):358–362. doi:10.1016/j.autrev.2014.12.006

    Article  CAS  PubMed  Google Scholar 

  93. De Carolis S, Botta A, Salvi S, di Pasquo E, Del Sordo G, Garufi C, Lanzone A, De Carolis MP (2015) Is there any role for the hydroxychloroquine (HCQ) in refractory obstetrical antiphospholipid syndrome (APS) treatment? Autoimmun Rev 14(9):760–762. doi:10.1016/j.autrev.2015.04.010

    Article  PubMed  Google Scholar 

  94. Mekinian A, Costedoat-Chalumeau N, Masseau A, Tincani A, De Caroli S, Alijotas-Reig J, Ruffatti A, Ambrozic A, Botta A, Le Guern V, Fritsch-Stork R, Nicaise-Roland P, Carbonne B, Carbillon L, Fain O (2015) Obstetrical APS: is there a place for hydroxychloroquine to improve the pregnancy outcome? Autoimmun Rev 14(1):23–29. doi:10.1016/j.autrev.2014.08.040

    Article  CAS  PubMed  Google Scholar 

  95. Mekinian A, Lazzaroni MG, Kuzenko A, Alijotas-Reig J, Ruffatti A, Levy P, Canti V, Bremme K, Bezanahary H, Bertero T, Dhote R, Maurier F, Andreoli L, Benbara A, Tigazin A, Carbillon L, Nicaise-Roland P, Tincani A, Fain O, Snfmi, the European Forum on Antiphospholipid A (2015) The efficacy of hydroxychloroquine for obstetrical outcome in anti-phospholipid syndrome: data from a European multicenter retrospective study. Autoimmun Rev 14(6):498–502. doi:10.1016/j.autrev.2015.01.012

    Article  CAS  PubMed  Google Scholar 

  96. Noel N, Dutasta F, Costedoat-Chalumeau N, Bienvenu B, Mariette X, Geffray L, Sene D, Chaidi RB, Michot JM, Fain O, Darnige L, Ankri A, Cacoub P, Piette JC, Saadoun D (2015) Safety and efficacy of oral direct inhibitors of thrombin and factor Xa in antiphospholipid syndrome. Autoimmun Rev 14(8):680–685. doi:10.1016/j.autrev.2015.03.007

    Article  CAS  PubMed  Google Scholar 

  97. Raso S, Sciascia S, Kuzenko A, Castagno I, Marozio L, Bertero MT (2015) Bridging therapy in antiphospholipid syndrome and antiphospholipid antibodies carriers: case series and review of the literature. Autoimmun Rev 14(1):36–42. doi:10.1016/j.autrev.2014.09.002

    Article  CAS  PubMed  Google Scholar 

  98. Kronbichler A, Brezina B, Quintana LF, Jayne DR (2016) Efficacy of plasma exchange and immunoadsorption in systemic lupus erythematosus and antiphospholipid syndrome: a systematic review. Autoimmun Rev 15(1):38–49. doi:10.1016/j.autrev.2015.08.010

    Article  PubMed  Google Scholar 

  99. Tenti S, Cheleschi S, Guidelli GM, Galeazzi M, Fioravanti A (2016) Intravenous immunoglobulins and antiphospholipid syndrome: how, when and why? A review of the literature. Autoimmun Rev 15(3):226–235. doi:10.1016/j.autrev.2015.11.009

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We are grateful to Simone Forest and Geneviève Michel for their help in typing the paper and to Dr. Wesley H. Brooks (Tampa, USA) for editorial assistance.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yves Renaudineau.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Additional information

Francis Couturaud and Yves Renaudineau contributed equally to this work.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Simonin, L., Pasquier, E., Leroyer, C. et al. Lymphocyte Disturbances in Primary Antiphospholipid Syndrome and Application to Venous Thromboembolism Follow-Up. Clinic Rev Allerg Immunol 53, 14–27 (2017). https://doi.org/10.1007/s12016-016-8568-1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12016-016-8568-1

Keywords

Navigation