Eosinophil Cationic Protein (ECP), a predictive marker of bullous pemphigoid severity and outcome

Delphine Giusti, Gregory Gatouillat, Sébastien Le Jan, Julie Plée, Philippe Bernard, Frank Antonicelli, Bach Nga Pham, Delphine Giusti, Gregory Gatouillat, Sébastien Le Jan, Julie Plée, Philippe Bernard, Frank Antonicelli, Bach Nga Pham

Abstract

Bullous Pemphigoid (BP) is an inflammatory rare autoimmune bullous dermatosis, which outcome cannot be predicted through clinical investigations. Eosinophils are the main immune infiltrated cells in BP. However, the release of Major Basic Protein (MBP), Eosinophil Derived Neurotoxin (EDN), and Eosinophil Cationic Protein (ECP) upon eosinophil activation has still not been evaluated with respect to BP development. MBP, EDN and ECP were measured by ELISA in serum (n = 61) and blister fluid (n = 20) of patients with BP at baseline, and in serum after 2 months of treatment (n = 41). Eosinophil activation in BP patients was illustrated at baseline by significantly higher MBP, EDN and ECP serum concentrations as compared with control subjects (n = 20), but without distinction according to disease severity or outcome. EDN and ECP values were even higher in the blister fluids (P < 0.01 and P < 0.05, respectively), whereas MBP values were lower (P < 0.001). ECP serum concentration decreased after 60 days of treatment in BP patients with ongoing remission but not in patients who later relapsed (P < 0.05). A reduction of at least 12.8 ng/mL in ECP concentrations provided a positive predictive value for remission of 81%, showing that ECP serum variation could be a useful biomarker stratifying BP patients at risk of relapse.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Serum concentrations of Eosinophil granule proteins at baseline. Major Basic Protein (MBP) (A), Eosinophil Derived Neurotoxin (EDN) (B) and Eosinophil Cationic Protein (ECP) (C) serum levels from 61 patients with active bullous pemphigoid and 24 healthy control subjects were measured by specific ELISA at the time of diagnosis. Results are expressed as ng/mL. (**P < 0.01; ***P < 0.001). Lines correspond to median and interquartile ranges.
Figure 2
Figure 2
MBP, EDN and ECP serum variations between baseline and day 60. Variations in the serum concentrations of MBP (A), EDN (B) and ECP (C) were measured by mean of specific ELISAs between days 0 and 60. (MD: Mild disease; R0: Remission). (*P < 0.05).
Figure 3
Figure 3
Prognostic value of the decrease in ECP serum concentrations between days 0 and 60. The prognostic properties of the decrease of ECP serum concentrations measured by means of ELISA between baseline and day 60 with respect of the occurrence of a clinical remission in patients with BP are depicted as a receiver operating characteristic (ROC) curve.
Figure 4
Figure 4
Comparison of serum and blister fluid concentrations for MBP, ECP and EDN in 20 patients with active bullous pemphigoid. The serum blister fluids concentrations of MPB (A), EDN (B) and ECP (C) were measured by means of specific ELISAs. Variations between serum and blister fluids were analyzed by means of Wilcoxon test for MBP and EDN and paired t test for ECP. Correlation between EDN concentrations in the serum and in the blister fluids was performed by means of Spearman’s correlation coefficient analysis. Statistical significance was as follow: (*P < 0.05, **P < 0.01; ***P < 0.001). Lines define median and interquartile ranges.

References

    1. Gammon WR, et al. An in vitro model of immune complex-mediated basement membrane zone separation caused by pemphigoid antibodies, leukocytes, and complement. J. Invest. Dermatol. 1982;78:285–290. doi: 10.1111/1523-1747.ep12507222.
    1. Schmidt E, Obe K, Bröcker EB, Zillikens D. Serum levels of autoantibodies to BP180 correlate with disease activity in patients with bullous pemphigoid. Arch. Dermatol. 2000;136:174–178. doi: 10.1001/archderm.136.2.174.
    1. Bernard P, Bedane C, Bonnetblanc JM. Anti-BP180 autoantibodies as a marker of poor prognosis in bullous pemphigoid: a cohort analysis of 94 elderly patients. Br. J. Dermatol. 1997;136:694–698. doi: 10.1111/j.1365-2133.1997.tb03654.x.
    1. Joly P, et al. Incidence and mortality of bullous pemphigoid in France. J. Invest. Dermatol. 2012;132:1998–2004. doi: 10.1038/jid.2012.35.
    1. Bernard P, et al. Incidence and distribution of subepidermal autoimmune bullous skin diseases in three French regions. Bullous Diseases French Study Group. Arch. Dermatol. 1995;131:48–52. doi: 10.1001/archderm.1995.01690130050009.
    1. Joly P, et al. A Comparison of Oral and Topical Corticosteroids in Patients with Bullous Pemphigoid. N. Engl. J. Med. 2002;346:321–327. doi: 10.1056/NEJMoa011592.
    1. Joly P, et al. A comparison of two regimens of topical corticosteroids in the treatment of patients with bullous pemphigoid: a multicenter randomized study. J. Invest. Dermatol. 2009;129:1681–1687. doi: 10.1038/jid.2008.412.
    1. Fichel F, et al. Clinical and immunologic factors associated with bullous pemphigoid relapse during the first year of treatment: a multicenter, prospective study. JAMA Dermatol. 2014;150:25–33. doi: 10.1001/jamadermatol.2013.5757.
    1. Le Jan S, et al. Innate immune cell-produced IL-17 sustains inflammation in bullous pemphigoid. J. Invest. Dermatol. 2014;134:2908–2917. doi: 10.1038/jid.2014.263.
    1. Plée J, et al. Integrating longitudinal serum IL-17 and IL-23 follow-up, along with autoantibodies variation, contributes to predict bullous pemphigoid outcome. Sci. Rep. 2015;5:18001. doi: 10.1038/srep18001.
    1. Leighty L, Li N, Diaz LA, Liu Z. Experimental models for the autoimmune and inflammatory blistering disease, Bullous pemphigoid. Arch. Dermatol. Res. 2007;299:417–422. doi: 10.1007/s00403-007-0790-5.
    1. Liu Z, et al. Subepidermal blistering induced by human autoantibodies to BP180 requires innate immune players in a humanized bullous pemphigoid mouse model. J. Autoimmun. 2008;31:331–338. doi: 10.1016/j.jaut.2008.08.009.
    1. Messingham KN, et al. Eosinophil localization to the basement membrane zone is autoantibody- and complement-dependent in a human cryosection model of bullous pemphigoid. Exp. Dermatol. 2016;25:50–55. doi: 10.1111/exd.12883.
    1. Dubertret L, Bertaux B, Fosse M, Touraine R. Cellular events leading to blister formation in bullous pemphigoid. Br. J. Dermatol. 1980;103:615–624. doi: 10.1111/j.1365-2133.1980.tb01683.x.
    1. Hogan SP, et al. Eosinophils: biological properties and role in health and disease. Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 2008;38:709–750. doi: 10.1111/j.1365-2222.2008.02958.x.
    1. Walsh GM. Eosinophil granule proteins and their role in disease. Curr. Opin. Hematol. 2001;8:28–33. doi: 10.1097/00062752-200101000-00006.
    1. Acharya KR, Ackerman SJ. Eosinophil Granule Proteins: Form and Function. J. Biol. Chem. 2014;289:17406–17415. doi: 10.1074/jbc.R113.546218.
    1. Muniz VS, Weller PF, Neves JS. Eosinophil crystalloid granules: structure, function, and beyond. J. Leukoc. Biol. 2012;92:281–288. doi: 10.1189/jlb.0212067.
    1. Borrego L, et al. Deposition of eosinophil granule proteins precedes blister formation in bullous pemphigoid. Comparison with neutrophil and mast cell granule proteins. Am. J. Pathol. 1996;148:897–909.
    1. Inaoki M, Takehara K. Increased serum levels of interleukin (IL)-5, IL-6 and IL-8 in bullous pemphigoid. J. Dermatol. Sci. 1998;16:152–157. doi: 10.1016/S0923-1811(97)00044-3.
    1. D’Auria L, et al. IL-5 levels in the serum and blister fluid of patients with bullous pemphigoid: correlations with eosinophil cationic protein, RANTES, IgE and disease severity. Arch. Dermatol. Res. 1998;290:25–27. doi: 10.1007/s004030050272.
    1. Karlen S, et al. Biological and molecular characteristics of interleukin-5 and its receptor. Int. Rev. Immunol. 1998;16:227–247. doi: 10.3109/08830189809042996.
    1. Büdinger L, et al. Identification and characterization of autoreactive T cell responses to bullous pemphigoid antigen 2 in patients and healthy controls. J. Clin. Invest. 1998;102:2082–2089. doi: 10.1172/JCI3335.
    1. Mishra A, Hogan SP, Lee JJ, Foster PS, Rothenberg ME. Fundamental signals that regulate eosinophil homing to the gastrointestinal tract. J. Clin. Invest. 1999;103:1719–1727. doi: 10.1172/JCI6560.
    1. Long H, Liao W, Wang L, Lu Q. A Player and Coordinator: The Versatile Roles of Eosinophils in the Immune System. Transfus. Med. Hemotherapy. 2016;43:96–108. doi: 10.1159/000445215.
    1. Venge R, et al. Eosinophil cationic protein (ECP): molecular and biological properties and the use of ECP as a marker of eosinophil activation in disease. Clin. Exp. Allergy. 1999;29:1172–1186. doi: 10.1046/j.1365-2222.1999.00542.x.
    1. Plager DA, et al. Eosinophil Ribonucleases and Their Cutaneous Lesion-Forming Activity. J. Immunol. Baltim. Md 1950. 2009;183:4013–4020.
    1. Tomassini M, et al. Release of granule proteins by eosinophils from allergic and nonallergic patients with eosinophilia on immunoglobulin-dependent activation. J. Allergy Clin. Immunol. 1991;88:365–375. doi: 10.1016/0091-6749(91)90099-A.
    1. Czech W, Schaller J, Schöpf E, Kapp A. Granulocyte activation in bullous diseases: release of granular proteins in bullous pemphigoid and pemphigus vulgaris. J. Am. Acad. Dermatol. 1993;29:210–215. doi: 10.1016/0190-9622(93)70170-X.
    1. Riani, M. et al. Bullous pemphigoid outcome is associated with CXCL10-induced matrix metalloproteinase 9 secretion from monocytes and neutrophils but not lymphocytes. J. Allergy Clin. Immunol. doi:10.1016/j.jaci.2016.08.012 (2016).
    1. Vaillant L, et al. Evaluation of clinical criteria for diagnosis of bullous pemphigoid. French Bullous Study Group. Arch. Dermatol. 1998;134:1075–1080. doi: 10.1001/archderm.134.9.1075.
    1. Bernard P, et al. [Bullous pemphigoid. Guidelines for the diagnosis and treatment. Centres de référence des maladies bulleuses auto-immunes. Société Française de Dermatologie] Ann. Dermatol. Vénéréologie. 2011;138:247–251. doi: 10.1016/j.annder.2011.01.009.

Source: PubMed

3
Subscribe