New Insights in the Pathogenesis of Autoimmune Hemolytic Anemia

Wilma Barcellini, Wilma Barcellini

Abstract

Autoimmune hemolytic anemia (AIHA) is caused by the increased destruction of red blood cells (RBCs) by anti-RBC autoantibodies with or without complement activation. RBC destruction may occur both by a direct lysis through the sequential activation of the final components of the complement cascade (membrane attack complex), or by antibody-dependent cell-mediated cytotoxicity (ADCC). The pathogenic role of autoantibodies depends on their class (the most frequent are IgG and IgM), subclass, thermal amplitude (warm and cold forms),as well as affinity and efficiency in activating complement. Several cytokines and cytotoxic mechanisms (CD8+ T and natural killer cells) are further involved in RBC destruction. Moreover, activated macrophages carrying Fc receptors may recognize and phagocyte erythrocytes opsonized by autoantibodies and complement. Direct complement-mediated lysis takes place mainly in the circulations and liver, whereas ADCC, cytotoxicity, and phagocytosis occur preferentially in the spleen and lymphoid organs. The degree of intravascular hemolysis is 10-fold greater than extravascular one. Finally, the efficacy of the erythroblastic compensatory response can greatly influence the clinical picture of AIHA. The interplay and relative burden of all these pathogenic mechanisms give reason for the great clinical heterogeneity of AIHAs, from fully compensated to rapidly evolving fatal cases.

Keywords: Autoantibodies; Autoimmune hemolytic anemia; Cytokines.

Figures

Fig. 1
Fig. 1
T-lymphocyte subsets and cytokine interactions in AIHA. Cytokines that were found elevated are highlighted in yellow: IL-2 and IL-12, that induce the differentiation of CD4+ naïve T cells into Th1 subset, and IL-4, that promotes Th2 switch. Elevated levels of TGF-β favor the differentiation of Th17 subset, which amplifies the pro-inflammatory and autoimmune response. On the contrary, decreased levels of both IFN-γ and Tregs were found (highlighted in green). The former resulted in decreased inhibition of Th2 response, i.e. an amplification of the autoantibody-mediated autoimmune disease, and the latter may cause a lack of down-regulation of inflammatory and autoimmune pathways.

References

    1. Petz LD, Garratty G. Immune Hemolytic Anemias, 2nd ed. Philadelphia, Churchill Livingstone, 2004.
    1. Berentsen S. How I manage cold agglutinin disease. Br J Haematol. 2011;153:309–317.
    1. Gehrs BC, Friedberg RC. Autoimmune hemolytic anemia. Am J Hematol. 2002;69:258–271.
    1. Abbas AK, Lichtman AH, Pillai S. Cellular and Molecular Immunology, Philadelphia, Saunders, 2012.
    1. McQueen F. A B cell explanation for autoimmune disease: the forbidden clone returns. Postgrad Med J. 2012;88:226–233.
    1. Garratty G, Arndt PA. Drugs that have been shown to cause drug-induced immune hemolytic anemia or positive direct antiglobulin tests: some interesting findings since 2007. Immunohematology. 2014;30:66–79.
    1. Salama A. Drug-induced immune hemolytic anemia. Expert Opin Drug Saf. 2009;8:73–79.
    1. Ahrens N, Pruss A, Kähne A, Kiesewetter H, Salama A. Coexistence of autoantibodies and alloantibodies to red blood cells due to blood transfusion. Transfusion. 2007;47:813–816.
    1. Alijotas-Reig J. Human adjuvant-related syndrome or autoimmune/inflammatory syndrome induced by adjuvants. Where have we come from? Where are we going? A proposal for new diagnostic criteria. Lupus 2015;2015, doi: 10.1177/0961203315579092.
    1. Barcellini W, Fattizzo B, Zaninoni A, Radice T, Nichele I, Di Bona E, Lunghi M, Tassinari C, Alfinito F, Ferrari A, Leporace AP, Niscola P, Carpenedo M, Boschetti C, Revelli N, Villa MA, Consonni D, Scaramucci L, De Fabritiis P, Tagariello G, Gaidano G, Rodeghiero F, Cortelezzi A, Zanella A. Clinical heterogeneity and predictors of outcome in primary autoimmune hemolytic anemia: a GIMEMA study of 308 patients. Blood. 2014;124:2930–2936.
    1. Fattizzo B, Zaninoni A, Nesa F, Sciumbata VM, Zanella A, Cortelezzi A, Barcellini W. Lessons from very severe, refractory and fatal primary autoimmune hemolytic anemias. Am J Hematol. 2015;90:E149–151.
    1. Arndt PA, Leger RM, Garratty G. Serologic findings in autoimmune hemolytic anemia associated with immunoglobulin M warm autoantibodies. Transfusion. 2009;49:235–242.
    1. Bartolmas T, Salama A. A dual antiglobulin test for the detection of weak or nonagglutinating immunoglobulin M warm autoantibodies. Transfusion. 2010;50:1131–1134.
    1. Bass GF, Tuscano ET, Tuscano JM. Diagnosis and classification of autoimmune hemolytic anemia. Autoimmun Rev. 2014;13:560–564.
    1. Garratty G. Immune hemolytic anemia caused by drugs. Expert Opin Drug Saf. 2012;11:635–642.
    1. Packman CH. Hemolytic anemia due to warm autoantibodies. Blood Rev. 2008;22:17–31.
    1. Dacie J. Auto-immune haemolytic anaemia [AIHA]: pathogenesis; in Dacie J (ed): The Haemolytic Anaemias. Vol. 3. London, Churchill Livingstone, 1992, pp. 392-451.
    1. Kurlander RJ, Rosse WF, Logue GL. Quantitative influence of antibody and complement coating of red cells on monocyte-mediated cell lysis. J Clin Invest. 1978;61:1309–1319.
    1. Berentsen S, Beiske K, Tjønnfjord GE. Primary chronic cold agglutinin disease: an update on pathogenesis, clinical features and therapy. Hematology. 2007;12:361–370.
    1. Berentsen S, Ulvestad E, Langholm R, Beiske K, Hjorth-Hansen H, Ghanima W, Sørbø JH, Tjønnfjord GE. Primary chronic cold agglutinin disease: a population based clinical study of 86 patients. Haematologica. 2006;91:460–466.
    1. Swiecicki PL, Hegerova LT, Gertz MA. Cold agglutinin disease. Blood. 2013;122:1114–1121.
    1. Ulvestad E, Berentsen S, Mollnes TE. Acute phase haemolysis in chronic cold agglutinin disease. Scand J Immunol. 2001;54:239–242.
    1. Roth A, Huttmann A, Rother RP, Duhrsen U, Philipp T. Long-term efficacy of the complement inhibitor eculizumab in cold agglutinin disease. Blood. 2009;113:3885–3886.
    1. Hill A, Rother RP, Arnold L, Kelly R, Cullen MJ, Richards SJ, Hillmen P. Eculizumab prevents intravascular hemolysis in patients with paroxysmal nocturnal hemoglobinuria and unmasks low-level extravascular hemolysis occurring through C3 opsonization. Haematologica. 2010;95:567–573.
    1. Shi J, Rose EL, Singh A, Hussain S, Stagliano NE, Parry GC, Panicker S. TNT003, an inhibitor of the serine protease C1s, prevents complement activation induced by cold agglutinins. Blood. 2014;123:4015–4022.
    1. Wouters D, Stephan F, Strengers P, de Haas M, Brouwer C, Hagenbeek A, van Oers MH, Zeerleder S. C1-esterase inhibitor concentrate rescues erythrocytes from complement-mediated destruction in autoimmune hemolytic anemia. Blood. 2013;121:1242–1244.
    1. Risitano AM, Notaro R, Pascariello C, Sica M, del Vecchio L, Horvath CJ, Fridkis-Hareli M, Selleri C, Lindorfer MA, Taylor RP, Luzzatto L, Holers VM. The complement receptor 2/factor H fusion protein TT30 protects paroxysmal nocturnal hemoglobinuria erythrocytes from complement-mediated hemolysis and C3 fragment. Blood. 2012;119:6307–6316.
    1. Risitano AM, Ricklin D, Huang Y, Reis ES, Chen H, Ricci P, Lin Z, Pascariello C, Raia M, Sica M, Del Vecchio L, Pane F, Lupu F, Notaro R, Resuello RR, DeAngelis RA, Lambris JD. Peptide inhibitors of C3 activation as a novel strategy of complement inhibition for the treatment of paroxysmal nocturnal hemoglobinuria. Blood. 2014;123:2094–2101.
    1. Harboe M, van Furth R, Schubothe H, Lind K, Evans RS. Exclusive occurrence of K chains in isolated cold haemagglutinins. Scand J Haematol 1965:2:259-266.
    1. Berentsen S, Tjønnfjord GE. Diagnosis and treatment of cold agglutinin mediated autoimmune hemolytic anemia. Blood Rev. 2012;26:107–115.
    1. Berentsen S, Randen U, Vågan AM, Hjorth-Hansen H, Vik A, Dalgaard J, Jacobsen EM, Thoresen AS, Beiske K, Tjønnfjord GE. High response rate and durable remissions following fludarabine and rituximab combination therapy for chronic cold agglutinin disease. Blood. 2010;116:3180–3184.
    1. Quinquenel A, Willekens C, Dupuis J, Royer B, Ysebaert L, De Guibert S, Michallet AS, Feugier P, Guieze R, Levy V, Delmer A. Bendamustine and rituximab combination in the management of chronic lymphocytic leukemia-associated autoimmune hemolytic anemia: a multicentric retrospective study of the French CLL intergroup (GCFLLC/MW and GOELAMS) Am J Hematol. 2015;90:204–207.
    1. Carson KR, Beckwith LG, Mehta J. Successful treatment of IgM-mediated autoimmune hemolytic anemia with bortezomib. Blood. 2010;115:915.
    1. Fagiolo E. Immunological tolerance loss vs. erythrocyte self antigens and cytokine network disregulation in autoimmune hemolytic anaemia. Autoimmun Rev. 2004;3:53–59.
    1. Gilsanz F, de La Serna J, Moltó L, Alvarez-Mon M. Hemolytic anemia in chronic large granular lymphocytic leukemia of natural killer cells: cytotoxicity of natural killer cells against autologous red cells is associated with hemolysis. Transfusion. 1996;36:463–466.
    1. Xu L, Zhang T, Liu Z, Li Q, Xu Z, Ren T. Critical role of Th17 cells in development of autoimmune hemolytic anemia. Exp Hematol. 2012;40:994–1004.
    1. Ward FJ, Hall AM, Cairns LS, Leggat AS, Urbaniak SJ, Vickers MA, Barker RN. Clonal regulatory T cells specific for a red blood cell autoantigen in human autoimmune hemolytic anemia. Blood. 2008;111:680–687.
    1. Sakaguchi S, Ono M, Setoguchi R, Yagi H, Hori S, Fehervari Z, Shimizu J, Takahashi T, Nomura T. Foxp3+ CD25+ CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol Rev. 2006;212:8–27.
    1. Ahmad E, Elgohary T, Ibrahim H. Naturally occurring regulatory T cells and interleukins 10 and 12 in the pathogenesis of idiopathic warm autoimmune hemolytic anemia. J Investig Allergol Clin Immunol. 2011;21:297–304.
    1. Barcellini W, Clerici G, Montesano R, Taioli E, Morelati F, Rebulla P, Zanella A. In vitro quantification of anti-red blood cell antibody production in idiopathic autoimmune haemolytic anaemia: effect of mitogen and cytokine stimulation. Br J Haematol. 2000;111:452–460.
    1. Toriani-Terenzi C, Fagiolo E. IL-10 and the cytokine network in the pathogenesis of human autoimmune hemolytic anemia. Ann N Y Acad Sci. 2005;1051:29–44.
    1. Barcellini W, Zaja F, Zaninoni A, Imperiali FG, Battista ML, Di Bona E, Fattizzo B, Consonni D, Cortelezzi A, Fanin R, Zanella A. Low-dose rituximab in adult patients with idiopathic autoimmune hemolytic anemia: clinical efficacy and biologic studies. Blood. 2012;119:3691–3697.
    1. Lechner K, Jager U. How I treat autoimmune hemolytic anemias in adults. Blood. 2010;16:1831–1838.
    1. Crowther M, Chan YL, Garbett IK, Lim W, Vickers MA, Crowther MA. Evidence-based focused review of the treatment of idiopathic warm immune hemolytic anemia in adults. Blood. 2011;118:4036–4040.
    1. Mahévas M, Michel M, Vingert B, Moroch J, Boutboul D, Audia S, Cagnard N, Ripa J, Menard C, Tarte K, Mégret J, Le Gallou S, Patin P, Thai L, Galicier L, Bonnotte B, Godeau B, Noizat-Pirenne F, Weill JC, Reynaud CA. Emergence of long-lived autoreactive plasma cells in the spleen of primary warm auto-immune hemolytic anemia patients treated with rituximab. J Autoimmun 2015; doi: 10.1016/j.jaut.2015.05.006.
    1. Conley CL, Lippman SM, Ness P. Autoimmune hemolytic anemia with reticulocytopenia. A medical emergency. JAMA. 1980;244:1688–1690.
    1. Liesveld JL, Rowe JM, Lichtman MA. Variability of the erythrocyte response in autoimmune hemolytic anemias: analysis of 109 cases. Blood. 1987;69:820–826.
    1. Aladjidi N, Leverger G, Leblanc T, Picat MQ, Michel G, Bertrand Y, Bader-Meunier B, Robert A, Nelken B, Gandemer V, Savel H, Stephan JL, Fouyssac F, Jeanpetit J, Thomas C, Rohrlich P, Baruchel A, Fischer A, Chêne G, Perel Y, Centre de Référence National des Cytopénies Auto-immunes de l'Enfant (CEREVANCE) New insights into childhood autoimmune hemolytic anemia: a French national observational study of 265 children. Haematologica. 2011;96:655–663.
    1. Arbach O, Funck R, Seibt F, Salama A. Erythropoietin may improve anemia in patients with autoimmune hemolytic anemia associated with reticulocytopenia. Transfus Med Hemother. 2012;39:221–223.
    1. Kuter DJ. The biology of thrombopoietin and thrombopoietin receptor agonists. Int J Hematol. 2013;98:10–23.
    1. Akl EA, Ramly EP, Kahale LA, Yosuico VE, Barba M, Sperati F, Cook D, Schünemann H. Anticoagulation for people with cancer and central venous catheters. Cochrane Database Syst Rev. 2014;10:CD006468.
    1. Wang M, Wang W, Abeywardane A, Adikarama M, McLornan D, Raj K, de Lavallade H, Devereux S, Mufti GJ, Pagliuca A, Potter VT, Mijovic A. Autoimmune hemolytic anemia after allogeneic hematopoietic stem cell transplantation: analysis of 533 adult patients who underwent transplantation at King's College Hospital. Biol Blood Marrow Transplant. 2015;21:60–66.
    1. Hoffman PC. Immune hemolytic anemia - selected topics. Hematology Am Soc Hematol Educ Program 2009:80-86.
    1. ElAnsary M, Hanna MO, Saadi G, ElShazly M, Fadel FI, Ahmed HA, Aziz AM, ElSharnouby A, Kandeel MM. Passenger lymphocyte syndrome in ABO and Rhesus D minor mismatched liver and kidney transplantation: A prospective analysis. Hum Immunol. 2015;76:447–452.

Source: PubMed

3
Sottoscrivi