• Non ci sono risultati.

44 Autoantibodies in Atopic Eczema

N/A
N/A
Protected

Academic year: 2022

Condividi "44 Autoantibodies in Atopic Eczema"

Copied!
9
0
0

Testo completo

(1)

44 Autoantibodies in Atopic Eczema

N. Mothes, I. Mittermann, K. Aichberger, P. Valent, R. Valenta

44.1

Introduction

Atopic eczema (AE) is a chronic inflammatory skin dis- ease with a biphasic course consisting of an acute inflammatory Th2-dominated phase and a chronic phase, reminiscent of a delayed-type immune reaction, with the appearance of Th1-like immune responses.

AE is thus a rather unusual manifestation of IgE- mediated allergies because the immediate reaction caused by the cross-linking of effector cell (i.e., mast cells, basophils) -bound IgE that leads to the release of inflammatory mediators within a few minutes (e.g., histamine and leukotrienes) does not dominate the disease. Skin lesions in AE are characterized by the influx of T cells and thus resemble eczematous features [1 – 3]. It was demonstrated very early that AE patients exhibit much stronger lymphoproliferative responses in response to allergens than patients suffering from other manifestations of atopy (e.g., allergic rhinocon- junctivitis, asthma) [4]. The important contribution of T cells to the pathogenesis of AE has now been well established [1, 5, 6].

AE affects 10 % – 20 % of children and 1 % – 3 % of adults. The majority of AE cases belong to the extrinsic form, which is due to IgE recognition of a broad variety of allergens. Patients suffering from extrinsic forms of AE are typically sensitized against a broad variety of allergens and frequently exhibit elevated levels of total serum IgE antibodies directed against allergens [7 – 10]. However, also intrinsic forms of AE have been described that resemble the typical clinical criteria of AE but are not characterized by the production of IgE antibodies with specificity for defined allergens [7].

For most forms of respiratory allergy (e.g., rhino- conjunctivitis, allergic asthma) and gastrointestinal allergy, IgE-sensitization to certain allergens and dis-

ease symptoms are highly related. In AE, it is possible that patients mount IgE antibodies against a variety of environmental allergens but disease exacerbations often lack an obvious association between contact to allergens and clinical symptoms [7].

Several explanations for this phenomenon can be considered. First, it is possible that the disease-elicit- ing allergens are not known and hence the association cannot be demonstrated. In this context, it should be mentioned that a broad variety of unusual allergens have been discovered to be related to AE (e.g., bacte- ria, yeast) [1, 11, 12]. Second, it may be considered that different pathogenetic mechanisms are operative in AE vs other forms of allergy that are more difficult to reveal by diagnostic tests. For example, skin prick testing with allergens is useful for most forms of aller- gy, whereas atopy patch testing, a test principle simi- lar to epicutaneous testing used for delayed-type hypersensitivity, has been found suitable for AE diag- nosis [13].

Third, allergens and allergen-derived peptides may

reach the skin also via endogenous routes. In this con-

text, it has been shown that AE patients contain aller-

gen-specific T cells that home to the skin, and anti-

gen-presenting cells in AE may undergo extensive

trafficking [14, 15]. Thus even allergens and allergen-

derived peptides taken up via the gastrointestinal

tract may reach the skin via various mechanisms [7,

16 – 18]. This book chapter is dedicated to summariz-

ing recent findings pointing to the possibility that also

IgE recognition of autoantigens and other autoim-

mune phenomena may be involved in the pathogene-

sis of AE [19].

(2)

44.2

Similarities and Cross-Reactivities Between Environmental Allergens and Human Proteins:

The Concept of IgE Autoimmunity is Reborn

Already in the 1920s, several investigators reported that human skin dander could trigger immediate hypersen- sitivity reactions and it has been demonstrated by cuta- neous testing and RAST technology that atopic patients form IgE antibodies not only against environmental allergens, but also against human proteins [20 – 25].

However, with the discovery and characterization of several extremely potent environmental allergens, the concept that IgE autoreactivity could play a pathogenet- ic role in atopy fell into oblivion. It was reborn when the cDNA coding for a birch pollen allergen was isolated

Table 44.1. List of autoantigens in atopic eczema patients. The antigenic structures, their molecular weight (kDa), their function and references are displayed and grouped in target structures of IgE antibodies and IgG antibodies in atopic eczema (AE)

Environmental allergens with similarity to human proteins

MW (kDa)

Origin Function Refer-

ences

Profilin 12 – 17 All eukaryotic cells Actin-binding protein partici-

pating in the phosphoinositide pathway and a signal transduction

[26]

Albumin (dog, cat) 60 – 70 Mammalian serum proteins [29]

Calcium-binding allergens (plants, fish)

8 – 23 Binding and transport of calcium [30]

Cytochrome allergens [31]

MnSOD Manganese superoxide dismutase [34]

P2-protein

Most eukaryotic cells

Ribosomal phosphoprotein type 2 [33]

Target structures of IgE autoantibodies in AE

Hom s 1 (SART 1) 73.4 Mammalian cells: skin, lung, colon, liver

Recognized by cytotoxic T cells of cancer patients

[39, 40]

Hom s 2 ( [ -NAC) 23.2 Most eukaryotic cells: skeletal muscle, liver

Sequence-specific sorting and trans- location of intracellular proteins

[41 – 43, 71]

Hom s 3 (BCL7B) 20.1 Mammalian cells: gall bladder, skeletal muscle, placenta, liver, ocular ciliary body

Putative oncogene [41, 44]

Hom s 4 (CALC) 54 Eukaryotic cells: skin, brain, lung, breast, liver, heart

Calcium-binding protein [41, 45]

Hom s 5 (KER)

Atopy related autoantigens (ara)

42.6 Mammalian cells: epithelial tissues

Formation of intermediate filaments

[41; 46]

Target structures of IgG autoantibodies in AE

IgE antibodies ⎫

⎬ ⎭

Mammalian cells

Cell nucleus

Most euka- ryotic cells

[56 – 64]

Nuclear proteins [66, 67]

DFS-70 70 Transcription coactivator p75, lens

epithelium-derived growth factor (LEDGF)

[68]

p80-coilin

⎭ Nuclear proteins

80 RNA processing and cellular

trafficking [69, 70]

and the corresponding allergen was identified as profi- lin, an ubiquitous actin-binding protein found in dif- ferent pollen, fungi and even in humans [26 – 28].

Recombinant profilin from birch reacted with IgE anti- bodies from sensitized allergic patients, induced spe- cific basophil activation and profilin-sensitized indi- viduals were found who, due to cross-reactivity with birch profilin, mounted IgE autoantibodies to human profilin [26]. The molecular nature of several other environmental allergens with similarity and/or cross- reactivity to human proteins was revealed (Table 44.1).

These allergens included dog and cat albumin [29], cal-

cium-binding allergens from plants and fish (reviewed

in [30]) and the cytochrome family of plant allergens

[31], manganese superoxide dismutase (MnSOD) and a

ribosomal P2-protein (P2-protein) from Aspergillus

418 44 Autoantibodies in Atopic Eczema

(3)

S KIN G A S T R OINT E S T INA L

T R A C T

E YE S R E S P IR A T OR Y

T R A C T

R eleas e of A utoallergens

IgE

FcHRI mas t c ell

IFN-J A P C

E c zema, P ruritus T is s ue damage

T cell epitope MHC

TCR

FcHRII FcHRI

T c ells T

E xogenous A llergens

Inflammation

A utos ens itization, Inflammation fumigatus. The latter two showed cross-reactivity with

human proteins and induced immediate-type and T cell-mediated autoimmune reactions [32, 33]. Humoral and cell-mediated autoreactivity to the corresponding human proteins in vitro and in vivo were demonstrat- ed. For MnSOD, inhibition studies showed that patients’ IgE antibodies recognized common epitopes between the enzymes from humans and other species, including Drosophila enzyme. Rare exposure to MnSOD of the latter species suggested molecular mim- icry as the mechanism for cross-reactivity [34].

Studies on the three-dimensional structure of aller- gens revealed that despite low or lacking sequence identity between environmental allergens and human proteins (e.g., profilin, timothy grass pollen allergen, Phl p 2), environmental allergens can mimic the struc- ture of human proteins [35, 36].

The identification of highly cross-reactive allergens that even cross-reacted with endogenous antigens gave rise to the rediscovery of IgE autoreactivity, but the pathogenetic relevance of this finding remained unclear. Furthermore, IgE recognition of cross-reac- tive autoantigens was not associated with certain forms/manifestations of atopy, extent of disease severi- ty, or other clinically important findings.

44.3

The Discovery that the Occurrence of IgE Autoantibodies Is Frequently Associated with Atopic Eczema

In order to investigate whether IgE autoreactivity is associated with certain manifestations of atopy or oth- er immunologically mediated diseases, the approach of testing sera from various patient groups and healthy non-allergic individuals for IgE reactivity to nitrocellu- lose-blotted human proteins from various cell types was taken [37]. The latter study revealed several impor- tant aspects. First, it was found that IgE autoreactivity can be found in a high percentage of AE patients but not in healthy persons, in patients suffering from mild forms of allergy and not in patients suffering from oth- er immunologically-mediated disorders (Fig. 44.2). IgE autoreactivity was directed against a large variety of human proteins expressed in various cell types and no major autoantigens could be identified. The specificity of IgE autoreactivity was demonstrated by inhibition studies by testing for IgG autoreactivity, and IgE allore-

Fig. 44.1. Pathomechanisms of autoallergy in AE patients. In sensitized atopic individuals, contact with exogenous allergens (orange asterisks) activates effector cells (e.g., mast cells) for immediate-type reactions and Th2 cell activation, which then may lead to tissue damage and the release of autoantigens (blue asterisks). These autoallergens can autosensitize and then cross-link IgE antibodies on mast cells or be presented to T cells

activity was excluded [37]. Although no major IgE-

reactive autoantigens, which are recognized by the

majority of AE patients, could be identified, a logical

next step was to attempt to characterize some of the

IgE-reactive autoantigens in detail.

(4)

44.4

Identification of IgE-Reactive Autoantigens by Molecular Cloning

For the identification and characterization of IgE-reac- tive autoantigens, an approach was taken that had been applied for the characterization of environmental aller- gens before (reviewed in [38]). Expression cDNA libraries prepared from the mRNA of human tissues were screened with serum IgE from AE patients to search for cDNAs coding for IgE-reactive autoantigens.

Five IgE-reactive autoantigens have been described by screening of human cDNA libraries and were named according to the allergen nomenclature as Hom s (Homo sapiens) antigens (Table 44.1).

Hom s 1 was the most frequently detected autoanti- gen recognized by serum IgE from patients suffering from AE. The DNA sequence of Hom s 1 codes for a 72.3-kDa protein highly expressed in the skin and to a lesser extent in other target organs of atopy (e.g., lung, gastrointestinal tract) [39]. Hom s 1 shares almost complete sequence identity with SART1, an antigen recognized by cytotoxic T cells of patients suffering from squamous esophageal cancer [40]. Using a rabbit anti-serum against purified Hom s 1, this autoantigen was detected within the epidermis, especially in the cytoplasm of suprabasal keratinocytes and in fibro- blasts and endothelial cells within the dermis [39].

Hom s 2 showed sequence identity with the human [ -chain of the nascent polypeptide-associated complex (NAC), a protein required for signal sequence-specific sorting and translocation of intracellular proteins [41 – 43]. The cDNA sequence codes for a protein with 23.2-kDa and by sequence analysis was found to be expressed in histogenetically unrelated tissues (Table 44.1). Hom s 2-homologous proteins were found in mice, insects, plants (Arabidopsis thaliana), yeast, and protozoa [42]. Using the purified and recombinant E.

coli-expressed Hom s 2 in circular dichroism experi- ments, it was demonstrated that [ -NAC represented a folded protein with mixed [ - and q -sheet conforma- tion and exhibited remarkable stability as well as refol- ding capacity [42].

Hom s 3, a protein that is expressed in skeletal mus- cle, gall bladder, placenta, liver, and ocular ciliary body is a 20.1-kDa protein called BCL7B. It possibly is an oncogene which occurs in many different tissues of the human body [41, 44].

Another recently identified autoantigen recognized

by IgE antibodies of AE patients was termed Hom s 4. It belongs to a new subfamily of calcium-binding pro- teins, which like Hom s 2, was found to induce T cell autoreactivity [42]. The complete Hom s 4 cDNA codes for a 54-kDa basic protein containing two calcium- binding domains. Using Hom s 4-specific antibodies Aichberger et al. demonstrated that the protein is strongly expressed within epidermal keratinocytes and dermal endothelial cells [45]. Moreover, it was shown that the recombinant Hom s 4 exhibited IgE cross-reac- tivity with exogenous calcium-binding allergens from plants and fish.

Hom s 5 is human cytokeratin type II (Table 44.1), a component of the mammalian cytoskeleton taking part in the formation of intermediate filaments in epithelial tissues [41, 46].

For some cloned IgE-reactive autoantigens, e.g., Hom s 2 sequence similarities with exogenous antigens present in yeast, plants, animals, and bacteria were found [42], whereas for other autoallergens no proteins with similarity to exogenous allergens have been found yet.

The unusual finding that most of the characterized IgE-reactive autoantigens represented intracellular proteins was confirmed by biochemical studies dem- onstrating that these autoantigens can be detected in the nuclear > microsomal > mitochondrial > cyto- plasmic fraction of A431 cells as well as in a variety of human tissue specimens (brain, bone, intestine, liver, lung, muscle, skin, uterus) and effector cells of atopy (basophils, mast cells, T cells) [47]. The biochemical studies thus confirmed the broad expression of IgE- reactive autoantigens in a variety of cell types and tis- sues. Skin-specific expression thus does not seem to be the reason why these autoantigens are recognized by IgE antibodies from AE patients. The biochemical studies on IgE-reactive autoantigens point to another feature of IgE-reactive autoantigens: it appears that these autoantigens are “better” recognized by IgE when they are altered by denaturation or other modifi- cations, indicating that “slightly altered self ” could be the target for IgE autoantibodies.

As stated above, molecular and histochemical char- acterization of autoallergens as well as fractionation experiments showed that most of them represented intracellular proteins [41, 47]. The fact that autoaller- gens represent mainly intracellular proteins raises the question of how these autoallergens are released to induce inflammation in AE patients.

420 44 Autoantibodies in Atopic Eczema

(5)

44.5

How Intracellular Antigens Can Contribute to the Pathogenesis of Atopic Eczema

Obviously, intracellular antigens are released whenever cells die, either due to necrosis or apoptosis. In fact, it has been demonstrated that keratinocytes in AE lesions undergo apoptosis and it is therefore possible that intracellular proteins, including autoantigens, are released by this mechanism [48]. Keratinocyte damage may occur via immunological mechanisms in AE patients but may be simply caused by allergic inflam- mation induced by exogenous allergens, by scratching and superinfections (Fig. 44.1). That IgE-reactive auto- antigens are indeed released into the circulation and can be detected complexed to IgE antibodies in AE patients has been demonstrated [39, 41]. Using anti- BCL7B antibodies, IgE-specific BCL7B immune com- plexes isolated with Sepharose-coupled anti-human IgE antibodies were detected by immunoblotting [41].

Likewise, Hom s 1 IgE immune complexes were detect- ed [39]. Circulating IgE autoallergen immune com- plexes may reach target organs of atopy and, when bound via Fc 5 -receptors on effector and inducer cells of atopy and may induce degranulation, mediator release or activation of T cells. For exogenous allergens, it has in fact been demonstrated that presentation via the low-affinity receptor (Fc 5 RII/CD23) as well as via the high-affinity receptor (Fc 5 RI) leads to enhanced T cell activation [49, 50]. The clinical relevance of the lat- ter findings has, however, not yet been established and it is unclear whether this mechanism is valid also for autoantigens. The description of IgE-dependent hista- mine-releasing factors in sera of atopic individuals [51, 52] has prompted the investigation of the possibility that these factors may be IgE–autoallergen immune complexes that can activate mast cells or basophils.

Sera from patients containing such histamine-releas- ing factors were described as IgE

+

sera by their ability to induce basophil degranulation without the addition of allergens [51, 52]. However, a recent study conduct- ed to establish a possible link between IgE autoreactivi- ty and IgE-dependent histamine release factors has indicated that the two phenomena are rather distinct from each other [53].

44.6

IgE Autoreactivity as a Possible Marker for Chronic Inflammation and Tissue Damage in Atopic Eczema

While the pathogenetic relevance of IgE autoreactivity has not been proven, it has been demonstrated that the intensity of IgE autoreactivity is associated with dis- ease severity and exacerbation [41]. In another study, it was found that IgE autoreactivity to nitrocellulose blotted human proteins increased when skin symp- toms occurred in response to contact with environ- mental allergens [54]. Furthermore, it was shown that IgE antibody reactivities to autoallergens decreased treatment of skin manifestations with cyclosporin A [55].

The latter studies indicate that IgE autoreactivity is a useful parameter to monitor chronic inflammation and tissue damage in AE patients.

44.7

IgG Autoantibodies in Atopic Eczema Patients

While IgE autoreactivity is strongly associated with AE and disease severity of AE, the importance of IgG auto- reactivity in AE is less clear. Some investigations showed the presence of anti-IgE antibodies in the sera of AE patients [56 – 58] but also in normal individuals [59, 60]. Because of the difficulty of purifying anti-IgE antibodies, which seem to exist in the form of immune complexes with IgE in serum only in very small quanti- ties, controversial results were obtained and the ques- tion of biological relevance still remains unproven [61 – 64]. Other IgG autoantibodies comprising antinu- clear antibodies (ANAs), well known in systemic rheu- matic diseases (e.g., SLE, Sjögren syndrome, scleroder- ma) [65] were recently reported to be present in some patients with AE [66, 67]. However, no association between positive ANA levels and a high amount of total IgE, disease duration, or the presence of respiratory atopy could be found.

Other authors described a typical, dense, fine-

speckled ANA pattern on Hep-2 cells of AE patients

who also recognized a 70-kDa protein on immuno-

blots. The antigen thus was termed dense fine-speckled

70 (DSF 70) [68]. The authors reported that 9 % – 30 %

of AE patients from different populations had anti-

(6)

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 N 220

97 66 45 30

20 14

kDa

A 431 DSF70 antibodies compared to patients with other autoimmune diseases or normal controls. Interesting- ly, almost all patients (16/18) with DFS70 antibodies had facial dermatitis, and in some AE patients IgE anti- DFS70 antibodies could also be detected [68].

Recently another autoantigen named p80-coilin was described in the nuclear coiled body of most eukaryot- ic cells [69, 70]. Sera of AE patients contained anti- bodies against this 80-kDa nuclear protein, which is thought to play a role in RNA processing and cellular trafficking. Almost 5 % of AE patients had p80-coilin- specific autoantibodies; however, further research is needed to clarify the relationship between anticoiled antibodies and AE [70].

44.8

Pathomechanisms of IgE Autoreactivity

The work done so far has shown that more than 60 % of AE patients mount IgE antibodies against endogenous human proteins and the molecular nature of several of these autoantigens has been identified [41] (Fig. 44.2, Table 44.1). These autoantigens are apparently released after tissue damage, which can be caused by numerous events, among them inflammation induced by exoge- nous allergens, mechanical skin irritation through scratching or bacterial skin infections (Fig. 44.1). The importance of inflammation induced by exogenous allergens for autosensitization can be studied in exper- imental animal models. In a very recent work it was shown that mice can be sensitized to human as well as

Fig. 44.2. Serum IgE reactivity of AE patients with nitrocellulose-blotted A431 proteins. Nitrocellulose-blotted human epithelial cell line (A431) proteins were probed with sera from 29 AE patients (lanes 1 – 29) and a serum from a non-atopic control individu- al (N). The molecular masses are shown in kilo daltons (kDa) on the left

murine [ -NAC resulting in a condition of autoallergy characterized by allergic asthma [71]. Mice sensitized to Hom s 2 developed IgE and IgG autoantibodies, T cell autoreactivity, immediate type skin sensitivity, asthma and lung inflammation. Interestingly, it turned out that autosensitization resulted in an allergic auto- immune response of a mixed Th2 and Th1 profile, as observed in AE. The important question that remains to be clarified is the exact mode of how IgE autoreactiv- ity can contribute to skin inflammation in AE patients.

For this purpose, it will be important to use the recom- binant autoallergens as paradigmatic tools to study whether they can induce immediate and chronic skin inflammation and to dissect the underlying pathome- chanisms.

Acknowledgements. This chapter was supported by grants F01808 and F01815 of the Austrian Science Fund.

References

1. Herz U, Bunikowski R, Renz H (1998) Role of T cells in atop- ic dermatitis. New aspects on the dynamics of cytokine pro- duction and the contribution of bacterial superantigens. Int Arch Allergy Immunol 115:179 – 190

2. Allam JP, Bieber T, Novak N (2005) Recent highlights in the pathophysiology of atopic eczema. Int Arch Allergy Immu- nol 136:191 – 197

3. Leung DY, Boguniewicz M, Howell MD, Nomura I, Hamid QA (2004) New insights into atopic dermatitis. J Clin Invest 113:651 – 657

4. Rawle FC, Mitchell EB, Platts-Mills TA (1984) T cell

responses to the major allergen from the house dust mite

422 44 Autoantibodies in Atopic Eczema

(7)

Dermatophagoides pteronyssinus, antigen P1: comparison of patients with asthma, atopic dermatitis, and perennial rhinitis. J Immunol 133:195 – 201

5. Trautmann A, Akdis M, Klunker S, Blaser K, Akdis CA (2001) Role of apoptosis in atopic dermatitis. Int Arch Allergy Immunol 124:230 – 232

6. Wohlfahrt JG, Kunzmann S, Menz G, Kneist W, Akdis CA, Blaser K, Schmidt-Weber CB (2003) T cell phenotype in allergic asthma and atopic dermatitis. Int Arch Allergy Immunol 131:272 – 282

7. Novak N, Bieber T (2003) Allergic and nonallergic forms of atopic diseases. J Allergy Clin Immunol 112:252 – 262 8. Wistokat-Wulfing A, Schmidt P, Darsow U, Ring J, Kapp A,

Werfel T (1999) Atopy patch test reactions are associated with T-lymphocyte-mediated allergen-specific immune responses in atopic dermatitis. Clin Exp Allergy 29:513 – 521 9. Schafer T, Heinrich J, Wjst M, Adam H, Ring J, Wichmann HE (1999) Association between severity of atopic eczema and degree of sensitization to aeroallergens in schoolchil- dren. J Allergy Clin Immunol 104:1280 – 1284

10. Ring J, Darsow U, Behrendt H (2001) Role of aeroallergens in atopic eczema: proof of concept with the atopy patch test. J Am Acad Dermatol 45:49 – 52

11. Novak N, Allam JP, Bieber T (2003) Allergic hyperreactivi- ty to microbial components: a trigger factor of “intrinsic”

atopic dermatitis? J Allergy Clin Immunol 112:215 – 216 12. Scheynius A, Johansson C, Buentke E, Zargari A, Linder

MT (2002) Atopic eczema/dermatitis syndrome and Malassezia. Int Arch Allergy Immunol 127:161 – 169 13. Darsow U, Ring J (2000) Airborne and dietary allergens in

atopic eczema: a comprehensive review of diagnostic tests.

Clin Exp Dermatol 25:544 – 551

14. Santamaria-Babi LF, Picker LJ, Perez-Soler MT, Drzimalla K, Flohr P, Blaser K, Hauser C (1995) Circulating allergen- reactive T cells from patients with atopic dermatitis and allergic contact dermatitis express the skin-selective hom- ing receptor, the cutaneous lymphocyte-associated anti- gen. J Exp Med 181:1935 – 1940

15. Abernathy-Carver KJ, Sampson HA, Picker LJ, Leung DY (1995) Milk-induced eczema is associated with the expan- sion of T cells expressing cutaneous lymphocyte antigen. J Clin Invest 95:913 – 918

16. Werfel T, Ahlers G, Schmidt P, Boeker M, Kapp A (1996) Detection of a kappa-casein-specific lymphocyte response in milk-responsive atopic dermatitis. Clin Exp Allergy 26:1380 – 1386

17. Werfel T, Ahlers G, Schmidt P, Boeker M, Kapp A, Neu- mann C (1997) Milk-responsiveness atopic dermatitis is associated with a casein-specific lymphocyte response in adolescent and adult patients. J Allergy Clin Immunol 99:124 – 133

18. Werfel T, Reekers R, Busche M, Schmidt P, Constien A, Wittmann M, Kapp A (1999) Evidence for a birch pollen- specific cutaneous T-cell response in food-responsive atopic dermatitis. Int Arch Allergy Immunol 118:230 – 231 19. Valenta R, Seiberler S, Natter S, Mahler V, Mossabeb R,

Ring J, Stingl G (2000) Autoallergy: a pathogenetic factor in atopic dermatitis? J Allergy Clin Immunol 105:432 – 437 20. Storm van Leeuwen W, Bien Z, Varekamp H (1926) Über

die Hautreaktion mit Extrakten menschlicher Kopfhaut-

schuppen bei allergischen Krankheiten. Klin Wochenschr 5:1023 – 1025

21. Hampton SF, Cooke RA (1941) The sensitivity of man to human dander, with particular reference to eczema (aller- gic dermatitis). J Allergy 13:63 – 76

22. Simon FA (1944) On the allergen in human dander. J Aller- gy 15:338 – 345

23. Simon FA (1947) The allergen of human dander present in the skin of the general body surface. J Invest Dermatol 9:329 – 332

24. Parish WE (1960) Autosensitization to skin. In: Rock A (ed) Progress in the Biological sciences in relation to der- matology. Cambridge University Press, New York, p 250 25. Berrens L (1970) The allergens in house dust. Prog Allergy

14:259 – 339

26. Valenta R, Duchene M, Pettenburger K, Sillaber C, Valent P, Bettelheim P, Breitenbach M, Rumpold H, Kraft D, Schei- ner O (1991) Identification of profilin as a novel allergen:

IgE autoreactivity in sensitized individuals. Science 253:

557 – 560

27. Valenta R, Duchene M, Ebner C, Valent P, Sillaber C, Devil- ler P, Ferreira F, Teijkl M, Edelmann H, Kraft D, Scheiner O (1992) Profilins constitute a novel family of functional plant-panallergens. J Exp Med 175:377 – 385

28. Valenta R, Ferreira F, Grote M, Swoboda I, Vrtala S, Duche- ne M, Deviller P, Meagher RB, McKinney E, Heberle-Bors E, Kraft D, Scheiner O (1993) Identification of profilin as an actin-binding protein in higher plants. J Biol Chem 268:22777 – 22781

29. Spitzauer S, Schweiger C, Sperr WR, Pandjaitan B, Valent P, Mühl S, Ebner C, Scheiner O, Kraft D, Rumpold H, Valenta R (1994) Molecular characterization of dog albumin as a cross-reactive allergen. J Allergy Clin Immunol 93:614 – 627

30. Valenta R, Hayek B, Seiberler S, Bugajska-Schretter A, Nie- derberger V, Twardosz A, Natter S, Vangelista L, Pastore A, Spitzauer S, Kraft D (1998) Calcium-binding allergens: from plants to man. Int Arch Allergy Immunol 117:160 – 166 31. Matthews PA, Baldo BA, Howden ME (1988) Cytochrome c

allergens isolated from the pollens of the dicotyledons English plantain (Plantago lanceolata) and Paterson’s curse (Echium plantagineum). Mol Immunol 25:63 – 68 32. Crameri R, Faith A, Hemmann S, Jaussi R, Ismail C, Menz

G, Blaser K (1996) Humoral and cell-mediated autoimmu- nity in allergy to Aspergillus fumigatus. J Exp Med 184:

265 – 270

33. Mayer C, Appenzeller U, Seelbach H, Achatz G, Oberkofler H, Breitenbach M, Blaser K, Crameri R (1999) Humoral and cell-mediated autoimmune reactions to human ribo- somal P-2 protein in individuals sensitized to Aspergillus fumigatus P-2 protein. J Exp Med 189:1507 – 1512 34. Mayer C, Hemmann S, Faith A, Blaser K, Crameri R (1997)

Cloning, production, characterisation and IgE cross-reac- tivity of different manganese superoxide dismutases in individuals sensitised to Aspergillus fumigatus. Int Arch Allergy Immunol 113:213 – 215

35. Fedorov AA, Ball T, Mahoney NM, Valenta R, Almo SC (1997) The molecular basis for allergen cross-reactivity:

Crystal structure and IgE epitope mapping of birch pollen

profilin. Structure 5:33 – 45

(8)

36. De Marino S, Morelli MAC, Fraternali F, Tamvorini E, Mus- co G, Vrtala S, Dolecek C, Arosio P, Valenta R, Pastore A (1999) An immunoglobulin fold in a major plant allergen:

the solution structure of Phl p 2 from timothy grass pollen.

Structure 7:943 – 952

37. Valenta R, Maurer D, Steiner R, Seiberler S, Sperr WR, Valent P, Spitzauer S, Kapiotis S, Smolen J, Stingl G (1996) Immunoglobulin E responses to human proteins in atopic patients. J Invest Dermatol 107:203 – 207

38. Valenta R, Kraft D (2002) From allergen structure to new forms of allergen-specific immunotherapy. Curr Opin Immunol 14:718 – 727

39. Valenta R, Natter S, Seiberler S, Wichlas S, Maurer D, Hess M, Pavelka M, Grote M, Ferreira F, Szephalusi Z, Valent P, Stingl G (1998) Molecular characterization of an autoaller- gen, Hom s 1, identified by serum IgE from atopic dermati- tis patients. J Invest Dermatol 111:1178 – 1183

40. Sehichijo S, Nakao M, Imai Y, Takasu H, Kawamoto M, Nijya F, Yang D, Toh Y, Yamana H, Itoh K (1998) A gene encoding antigenic peptide of human squamous cell carci- noma recognized by cytotoxic T lymphocytes. J Exp Med 187:277 – 288

41. Natter S, Seiberler S, Hufnagl P, Binder BR, Hirschl AM, Ring J, Abeck D, Schmidt T, Valent P, Valenta R (1998) Iso- lation of cDNA clones coding for IgE autoantigens with serum IgE from atopic dermatitis patients. FASEB J 12:1559 – 1569

42. Mossabeb R, Seiberler S, Mittermann I, Reininger R, Spit- zauer S, Natter S, Verdino P, Keller W, Kraft D, Valenta R (2002) Characterization of a novel isoform of [ -nascent polypeptide-associated complex as IgE-defined autoanti- gen. J Invest Dermatol 119:820 – 829

43. Wiedmann B, Sakai H, Davis TA, Wiedmann M (1994) A protein complex required for signal-sequence-specific sorting and translocation. Nature (London) 370:434 – 440 44. Zani VJ, Asou N, Jadayel D, Heward JM, Shipley J, Nacheva

E, Takasuki K, Catovsky D, Dyer MJ (1996) Molecular clon- ing of complex chromosomal translocation t (8;14;12) (q24.1; q32.3, q24.1) in a Burkitt lymphoma cell line defines a new gene (BCL7A) with homology to caldesmon.

Blood 87:3124 – 3134

45. Aichberger KJ, Mittermann I, Reininger R, Seiberler S, Swoboda I, Spitzauer S, Kopp T, Stingl G, Sperr WR, Valent P, Repa A, Bohle B, Kraft D, Valenta R (2005) Hom s 4, an IgE-reactive autoantigen belonging to a new subfamily of calcium-binding proteins can induce T helper cell type 1- mediated autoreactivity. J Immunol 175:1286 – 1294 46. Glass C, Kim KH, Fuchs E (1985) Sequence and expression

of a human type II mesothelial keratin. J Cell Biol 101:

2366 – 2373

47. Seiberler S, Bugajska-Schretter A, Hufnagl P, Binder BR, Stöckl J, Spitzauer S, Valent P, Valenta R (1999) Character- ization of IgE-reactive autoantigens in atopic dermatitis, 1:

subcellular distribution and tissue-specific expression. Int Arch Allergy Immunol 120:108 – 116

48. Trautmann A, Akdis M, Kleemann D, Altznauer F, Simon HU, Graeve T, Noll M, Bröcker EB, Blaser K, Akdis CA (2000) T cell-mediated Fas-induced keratinocyte apopto- sis plays a key pathogenetic role in eczematous dermatitis.

J Clin Invest 106:25 – 35

49. Stingl G, Maurer D (1997) IgE-mediated allergen presenta- tion via Fc epsilon RI on antigen-presenting cells. Int Arch Allergy Immunol 113:24 – 29

50. Bieber T (1996) Fc epsilon RI on antigen-presenting cells.

Curr Opin Immunol 8:773 – 777

51. MacDonald SM, Rafnar T, Langdon J, Lichtenstein LM (1995) Molecular identification of an IgE-dependent hista- mine-releasing factor. Science 269:688 – 690

52. MacDonald SM (1996) Histamine-releasing factors. Curr Opin Immunol 8:778 – 783

53. Budde IK, de Heer PG, Natter S, Mahler V, van der Zee JS, Valenta R, Aalberse RC (2002) Studies on the association between immunoglobulin E autoreactivity and immuno- globulin E-dependent histamine-releasing factors. Immu- nology 107:243 – 251

54. Seiberler S, Natter S, Hufnagl P, Binder BR, Valenta R (1999) Characterization of IgE-reactive autoantigens in atopic dermatitis; 2. A pilot study on IgE versus IgG sub- class response and seasonal variation of IgE autoreactivity.

Int Arch Allergy Immunol 120:117 – 125

55. Kinaciyan T, Natter S, Kraft D, Stingl G, Valenta R (2002) IgE autoantibodies monitored in a patients with atopic dermatitis under cyclosporin A treatment reflect tissue damage. J Allergy Clin Immunol 109:717 – 719

56. Carini C, Brostoff J (1983) An antiglobulin: IgG anti-IgE.

Occurrence and specificity. Ann Allergy 51:251 – 253 57. Nawata Y, Koike T, Hosokawa H, Tomioka H, Yoshida S

(1985) Anti-IgE autoantibody in patients with atopic der- matitis. J Immunol 135:478 – 482

58. Quinti I, Brozek C, Wook N, Geha R, Leung DYM (1986) Circulating IgG autoantibodies to IgE in atopic syndromes.

J Allergy Clin Immunol 77:586 – 594

59. Inganas M, Johansson SGO, Bennich H (1981) Anti-IgE antibodies in human serum. Occurrence and specificity.

Int Arch Allergy Appl Immunol 65:51 – 61

60. Wilson PB, Fairfield JE, Beech N (1987) Detection of IgG subclass-specific anti-IgE antibodies in normal and atopic individuals. Int Arch Allergy Appl Immunol 84:198 – 204 61. Marone G, Casolaro V, Paganelli R, Quinti I (1989) IgG

anti-IgE from atopic dermatitis induces mediator release from basophils and mast cells. J Invest Dermatol 93:

246 – 252

62. Jensen-Jarolim E, Vogel M, de Weck AL, Stadler BM (1992) Anti-IgE autoantibodies mistaken for specific IgG. J Aller- gy Clin Immunol 89:31 – 43

63. Johansson SGO (1986) Anti-IgE antibodies in human sera.

J Allergy Clin Immunol 77:555 – 557

64. Ritter C, Battig M, Kraemer R, Stadler BM (1991) IgE hid- den in immune complexes with anti-IgE autoantibodies in children with asthma. J Allergy Clin Immunol 88:793 – 801 65. Von Muhlen CA, Tan EM (1995) Autoantibodies in the diagnosis of systemic rheumatic diseases. Semin Arthritis Rheum 24:323 – 358

66. Tada J, Toi Y, Yoshioka T, Fujiwara H, Arata J (1994) Anti- nuclear antibodies in patients with atopic dermatitis and severe facial lesions. Dermatology 189:38 – 40

67. Ohkouchi K, Mizutani H, Tanaka M, Takahashi M, Nakas- hima K, Shimizu M (1999) Anti-elongation factor-1 [ auto- antibody in adult atopic dermatitis patients. Int Immunol 11:1635 – 1640

424 44 Autoantibodies in Atopic Eczema

(9)

68. Ochs RL, Muro Y, Si Y, Ge H, Chan EKL, Tan EM (2000) Autoantibodies to DFS 70 kd/transcription coactivator p75 in atopic dermatitis and other conditions. J Allergy Clin Immunol 105:1211 – 1220

69. Onouchi H, Muro Y, Tomita Y (1999) Clinical features and IgG subclass distribution of anti-p80 coilin antibodies. J Autoimmun 12:225 – 232

70. Coilin Bellini M (2000) More than a molecular marker of the Cajal (coiled) body. BioEssays 22:861 – 867

71. Bünder R, Mittermann I, Herz U, Focke M, Wegmann M,

Valenta R, Renz H (2004) Induction of autoallergy with an

environmental allergen mimicking a self protein in a

murine model of experimental allergic asthma. J Allergy

Clin Immunol 114:422 – 428

Riferimenti

Documenti correlati

Wollenberg A, Kraft S, Hanau D, Bieber T (1996) Immuno- morphological and ultrastructural characterization of Langerhans cells and a novel, inflammatory dendritic epi- dermal

Lundberg L, Johannesson M, Silverdahl M, Hermansson C, Lindberg M (2000) Health-related quality of life in patients with psoriasis and atopic dermatitis measured with SF-36, DLQI and

Kobayashi Y (2001) Serum eosinophil-derived neurotoxin may reflect more strongly disease activity in childhood atopic dermatitis than eosinophil cationic protein.. Caproni M, Agata

Donnerer J, Schuligoi R, Stein C (1992) Increased content and transport of substance P and calcitonin gene-related peptide in sensory nerves innervating inflamed tissue: evi- dence

The nummular type of atopic eczema with oozing as a sign of secondary infection, also called impetiginous

) Personal or family history of atopic diseases ) IgE-mediated allergic sensitization 374 38 The Phenomenon of Irritable Skin in Atopic Eczema.. Stigmata of atopic constitution

But equally, the results suggest that certain sub- groups of patients are more likely to benefit from avoidance; however, the characteristics of these groups 394 40 The Role of

) Are probiotics effective for the prevention of other allergic diseases apart from atopic eczema (respiratory, gastrointestinal)? In the 4-year follow- up study, children in