Full-text resources of PSJD and other databases are now available in the new Library of Science.
Visit https://bibliotekanauki.pl

PL EN


Preferences help
enabled [disable] Abstract
Number of results
2011 | 38 | suplement 2 | 5-260

Article title

Ocena zjawisk immunologicznych i enzymatycznych w patogenezie wybranych podnaskórkowych chorób pęcherzowych

Content

Title variants

Languages of publication

EN

Abstracts

EN
The pathogenesis of subepidermal autoimmune blistering diseases, such as pemphigoid (BP, ang. bullous pemphigoid) and dermatitis herpetiformis (DH, ang. dermatitis herpetiformis, morbus Duhring) is connected with the destruction of basement membrane components and blister formation. Ultrastructural studies confirmed the basement membrane zone as a structure responsible for the integrity of dermo-epidermal junction. The binding of autoantibodies in BMZ zone leads to the activation of immunological and enzymatic mechanisms leading to the destruction of hemidesmosomes and anchoring fibres. Despite a similar initial stage: the connection of autoantibodies with autoantigens ,which are the proteins of the basement membrane structures, the further development of the disease mechanisms is different. The Differences of antigens and the activation of other cytokines, chemokines and enzymes imply the characteristic histological immunological picture, and require a different therapy. In BP and DH the impact of the disorders of certain cytokines expression , especially proinflammatory and complement components, on the development of inflammatory infiltrates and the formation of subepidermal blisters was shown. These phenomena are the cause of the activation of adhesion molecules and subsequent inflammatory mediators such as chemokines, neuropeptides and proteolitic enzymes. Recent experimental studies have also shown that these processes can activate procoagulant factors causing the development of blood thrombus, which is the most common cause of complications in patients with BP. The goal of the study was: 1. The assessment of the role of cytokine network components, such as adhesion molecules, chemokines, neuropeptides, and markers of activation of eosinophils and mast cells in the formation of skin lesions in pemphigoid and dermatitis herpetiformis. 2. The assessment of the role of selected proteolytic enzymes: metalloproteinases, adamalysins and their tissue inhibitors in the development of subepidermal blisters. 3. The assessment of procoagulation factors activity and basement membrane destruction caused by them in pemphigoid and dermatitis herpetiformis. 4. The indication of coagulation parameters useful in the prevention of complications in therapy of BP patients. The study included 61 persons: 27 untreated patients with BP (range: 58 to 84 years, average - 68,5) and 14 with DH (range: 18 to 70 years, average – 49,8) in an active stage of the disease. A Control group consisted of 20 healthy individuals (range:50 to 80 years, average – 71,6 for BP patients and ranged:19 to 49, average – 42 for DH patients). All the study subjects gave their consent before entering the study and the study protocol was approved by The Local Ethical Committee of Medical University of Lodz. Paraffin-embedded sections were used for routine staining and for immunohistochemistry to assess the expression of adhesion molecules, chemokines and their receptors, neuropeptides, metalloproteinases and their inhibitors and adamalysines. The following primary monoclonal antibodies were used: 1. adhesive molecules: anty- CD29 (β1 family), CD61 (GPIII) (β3 family), CD104 (β4 family) CD 62E (E-selectin), CD62L (L-selectin) from Novocastra 2. chemokines and their receptors: anty- eotaksyna, TARC, MCP-1, CCR- 1, CXCR-1, CXCR-2 from R&D. 3. neuropeptides: anty- Neurokinin B antibody, Endothelin B Receptor, CGRP, Corticotropin Releasing Factor and Substance P, from Abcam. 4. metalloproteinases: anty- MMP1, MMP2, MMP9, MMP1, TIMP1, TIMP2, TIMP 3 from Novocastra. 5. adamalisyns: anty- ADAM8, ADAM10, ADAM15, ADAM17 from R&D. 6. anty- TNF α from R&D. 7. tissue factors: anty- TF from American Diagnostica Inc. The Significantly increased expression of adamalysines: ADAM8, ADAM10, ADAM15, ADAM17 in the tissues (immunohistochemistry technique assessed) was confirmed by in situ hybridization and immunoblotting. In order to determine the concentrations of the examined protein in the serum the enzyme-linked immunoassay method was used using ELISA kits: TARC, MCP-1, eotaxin from R&D ECP – ELISA Kit for Human Ribonuclease A3 from Uscn Life Science; PAF ELISA Kit for Human Platelet Activating Factor (PAF from Uscn Life Science, Tryptaza, ELISA Kit for Human Tryptase) from Uscn Life Science, Chymaza ELISA Kit for Human Chymase 1 Mast Cell from Uscn Life Science, TNFα Human TNFα -ELISA Kit from Gen-Probe Diaclone, IL-4 Human IL-4 ELISA Kit from Gen-Probe Diaclone, Neurokinin B, Endotelin B, CGRP, Substance P ELISA kit from R&D, MMP1, MMP2, MMP9 i TIMP2 ELISA kit from Quantikine, R&D Systems, IMUBIND Tissue Factor ELISA Kit from American Diagnostica Inc. The Indications of d-dimers were performed using the method of optical test apparatus Turbidity STA Compact (Roche Diagnostics). The Determination of the concentration of fibrinogen was performed using chronometric Clauss and Turbidity method (during the determination coagulation parameters). The results were statistically analyzed. The Results were considered statistically significant with p < 0.05. The localization and expression of adhesive molecules: E- and L-selectins and β1, β3, β4 integrins was studied by immunohistochemistry.The Expression of selectins was detected in the epithelium and skin leukocytes in all DH and BP samples. The expression of β1, β3 integrins was detected mainly in basal keratinocytes as well as focally in the other layers of the epidermis. The expression of β4 integrin was irregular and detected mainly in the blister. In the control group only weak expression of the examined molecules was detected. The expression of one of the chemokines: eotaxin was found in the epidermis and in the cells of inflammatory infiltrate in the epidermis. The expression of eotaxin was significantly higher in the patients with BP compared to the healthy subjects, as well as in the patients with DH compared to the healthy subjects (infiltration - p < 0.02, keratinocytes - p < 0.03). In the patients with BP TARC (thymus and activation regulated chemokine) expression was present in keratinocytes of the lower layers of the epidermis around lesions in individual cells and the inflammatory infiltrate located in the epidermis. TARC expression was significantly higher in the patients with BP compared to the healthy subjects (infiltration - p < 0.001, keratinocytes – p < 0.006) and skin lesions in relation to the perilesional skin in BP (infiltration p < 0.009 and keratinocytes, p < 0.001). In the biopsies from the skin lesions in DH the average intensity of expression TARC was observed in most cases. In keratinocytes within the lesions it differed significantly between the patients with expression of BP and DH (p < 0.05), as well as in keratinocytes of the perilesional skin (p < 0.05). In the control group there was very weak, focal expression of TARC of single keratinocytes located in the lower layer of the epidermis. The expression of chemokin MCP-1 (monocyte chemoattractant protein-1) was present in skin lesions and the perilesional skin in the patients with BP. There was no expression of MCP-1 in DH patients and the control group. The expression of chemokine receptor: CCR-1 in skin lesions and the perilesional skin of the patients with BP was similarly weak. There was no statistically significant difference between the expression of CCR-1 in skin lesions and the perilesional skin in BP and DH. The expression of receptor for chemokines CXCR-1 was present in the biopsies of skin lesions in the patients with BP. CXCR-1 expression was significantly higher in the skin lesions in relation to the perilesional skin in BP (p < 0.03). In all the cases of patients with DH cells expressing CXCR-1were found in lesions. CXCR-1 expression was significantly higher in the patients with DH compared to the healthy subjects (p < 0.001). The Patients with DH showed higher expression of CXCR-1 in the biopsies of lesions compared to the patients with BP (0.67±0.18 vs. 0.15±0.24, p < 0.001). In the skin lesions in BP patients, receptor for chemokines CXCR-2 expression was demonstrated in all specimens and was significantly higher compared to the perilesional skin in BP (p < 0.02) as well as the expression in the healthy subjects (p < 0.04). In the biopsies taken from the blisters in DH patients in all cases the expression of CXCR-2 was present. The Patients with DH showed significantly higher expression of CXCR-2 in skin lesions compared to the patients with BP (1.1±0.87 vs. 0.31±0.24, p < 0.001). There was no expression of CCR-1, CXCR-1, CXCR-2 in the control group. The concentrations of MCP-1 levels were statistically higher in the patients with BP (374.8±30.8 pg/mL) compared to the patients with DH (282.2±21 pg/mL, p < 0.05), although there was no statistically significant difference in comparison to the control group. The concentration of ECP (283.9±66.92 pg/mL) and PAF (136.9±29.87 pg/mL) levels was significantly higher in BP than in the healthy subjects and the patients with DH (p < 0.05). The Concentration of chymase (0.21±0.98 pg/mL) was significantly higher in the patients with BP compared to the healthy subjects and the patients with DH. There was no difference in the concentrations of tryptase, IL-4 and TNF in the treatment groups. The expression of TNFα was demonstrated in inflammatory infiltrates and keratinocytes and in blister liquid of BP and DH patients. The Expression of TNFα was also confirmed in the perilesional skin. Morphometric analysis showed a higher expression of TNFα in the skin lesions as amended in relation to the perilesional skin (1.59±0.58 BP vs. 1.01±0.43, DH 1.39±0.84 vs. 0.69±0.59) as well as in the control groups vs. the control group (p < 0.01). The expression of neither the examined chemokines nor their receptors was observed in the biopsies from the healthy people. The expression of the neuropeptide: corticoliberin CRF (corticotrophin releasing factor) was present in keratinocytes, cells of the sebaceous glands and sweat glands, and the infiltration of inflammatory cells. The results had significantly higher expression of CRF in the biopsies derived from the skin lesions than in the skin biopsies taken from the perilesional skin, and this applied both to the individuals with BP (1.58±0.64 vs. 1.04±0.48, p < 0.001) and with the DH (1.36±0.89 vs. 0.71±0.61, p < 0.05). The lowest expression of CRF was demonstrated in the healthy subjects (0.36±0.15). The expression of endothelin receptor B was found in the endothelial cells, vascular muscle and keratinocytes. This phenomenon was observed for the increased expression of endothelin receptor B in the individuals with DH. This value was significantly higher in the skin lesions than in the perilesional skin (1.24±0.81 vs. 0.66±0.43, p < 0.04). There was no difference in the expression of endothelin B receptor in the perilesional skin in DH patients and the control group (0.66±0.43 vs. 0.58±0.32). The Expression of Substance P, CGRP and neurokinin B was demonstrated in the nerve endings in the epidermis and the skin. The expression of these neuropeptides in the groups was not statistically significant. The expression of metalloproteinases - MMP1, 2, 9 and 10 (MMP, matrix metalloproteinase) in patients with BP was found both in the basal layer and the whole epidermis and in the fluid blister and inflammatory infiltrates. In biopsies from the skin lesions with the blister the linear expression of MMPs has been shown in the bottom of the blister. In patients with DH MMPs expression was found in the basal layer of keratinocytes in all the biopsies from the lesion, in most biopsies it was very intense. The Expression of MMPs is not limited to the basal layer of keratinocytes, but was also present in medium intensity in the other layers of epidermal keratinocytes. The expression of these MMPs was also found in neutrophils, blister fluid and microabscesses. In contrast to the MMPs, the expression of their inhibitors - TIMPs (tissue inhibitor of martix metelloproteinases) was moderately or poorly pronounced. The expression of TIMP1, 2, 3 in the patients with BP was found throughout the epidermis. In the presence of the blister it was ascertained at its bottom. Moderate activity of these enzymes was demonstrated in inflammatory infiltrates and blister fluid. The expression of TIMP1, 2, 3 of medium intensity in all specimens was demonstrated throughout the epidermis of the patients with DH, as well as in neutrophilic microabscesses. In the studied serum of the patients with DH and BP the concentrations of MMP1, MMP2 and TIMP2 are within normal limits. Individual values of MMP9 concentrations were elevated in the patients with BP, in the remaining members of the studied groups they were normal. In the comparison group they were not elevated in the serum concentrations of these enzymes. The expression of adamalysins: ADAM8, 10, 15 and 17 (ADAM, a disintegrin and metalloproteinases domain) was found in the cells of the basal layer and other layers of the epidermis and dermis in BP and DH. Eosinophilic and neutrophilic infiltration in the skin lesions of patients with BP showed a mean intensity expression. The Expression of ADAMs was also shown in perivascular influx and microabscesses in DH. The Expression of ADAM8, 10, 15 and 17 in the control group in all the biopsies was detected only in single keratinocytes. Morphometric analysis showed a higher expression of ADAM8, 10, 15 and 17 in the skin lesions compared to the perilesional skin in keratinocytes, inflammatory infiltration in both the group of patients with BP and DH. Morphometric analysis showed a significantly higher expression of ADAM15 in the skin lesions in BP and in the perilesional skin compared to the patients with DH (p < 0.001). In a study by immunoblotting a significantly higher expression of ADAM17 was detected in the patients with BP compared to the patients with DH (p = 0.007 and 0.007, respectively) and the control group (p = 0.007 and p = 0.006 respectively). The Immunohistochemical study was confirmed by in situ hybridization. The expression of tissue factor (TF, tissue factor) was demonstrated in the neutrophils in inflammatory infiltrates and keratinocytes and blisters fluid of the patients with BP. TF expression was also shown in the perilesional skin. Morphometric analysis showed a higher expression of TF in the skin lesions in comparsion to the perilesional skin in the patients with BP (p < 0.02). The studies of the patients with DH showed no significant expression of the tissue factor in pathologically changed, and normal appearing skin. TF was also detected in the specimens from the healthy individuals. The Expression, however, in this group was significantly lower than in the lesions in BP and comparable to the lesions in DH. In the group of the patients with BP serum TF concentration was 847.2±402.7 values, in the patients with DH 285.7±187.7 pg/mL, and the comparison group for BP - 252.6±169.3 pg/mL, and for DH 221.9±153.3 pg/mL. These results indicate a statistically significant difference in the TF values between the group of patients with BP and DH and BP and the control group (p <0.001). The values of d-dimers in the group of patients with BP before the treatment, most patients were in the upper limits of normal and averaged: 0.49±0.40 mg/mL (0.33-2.9 mg/mL). In 7 patients the values were elevated and had a range between 1.2 and 2.9 μg/mL. These values were higher than in the group of patients with DH and the comparison group. In all patients with DH values of d-dimers correspond to the standard dimers and their average value was 0.31±0.19 mg/mL (0.26-0.36 g/mL). In the control group for patients with BP in an individual d-dimer values exceeded the normal in the other corresponded to the standard 0.29±0.3 mg/mL (0.24-0.67 g/mL), the comparison group for all patients with DH values were normal - mean: 0.34±0.6 mg/mL. There is a statistically significant difference between the group of patients with BP and DH and BP and a group comparison of the value of d-dimers (p < 0.05). In the patients with BP the value of fibrinogen is an average 4.50±0.18 g/L at normal 2.0-5.0 g/L. The Patients with DH had a mean fibrinogen value 2.80±0.14 g/L in the control group for BP - 2.60±0.3 g/L and for DH - 2.45±0.26 g/L. There is a statistically significant difference between the group of patients with BP and DH and BP and the comparison group in the value of fibrinogen (p < 0.05). The obtained results allowed to formulate the following conclusions: 1. Differences in the expression of adhesion molecules and chemokines and their receptors are the cause of a different composition of the inflammatory infiltrate in dermatitis herpetiformis and pemphigoid, in which a special role is played by tissue factor activating eosinophiles. 2. Increase in expression of neuropeptides is one of the factors of development neuroimmunological inflammation and itching in the DH and BP. 3. The increased activity of metalloproteinases and adamalysines and their tissue inhibitors and their balance disorder is the cause of the activation of cytokines and tissue destruction in the course of BP and DH. These data offer new prospects for therapeutic BP and DH. 4. The increase in tissue factor expression in skin lesions and the serum of patients with BP indicates the activation of the coagulation cascade and the coexistence of elevated levels of d-dimer and fibrinogen, points to the particular predisposition to thromboembolic complications of glicocorticosteroids in therapy. 5. The determination of coagulation parameters should be the standard before the planned treatment with glicocorticosteroids in patients with pemphigoid.

Discipline

Year

Volume

38

Issue

Pages

5-260

Physical description

Contributors

  • Klinika Dermatologii i Wenerologii, Uniwersytet Medyczny w Łodzi

References

  • Goebler M, Zillikens D. Bullous pemphigoid: diagnosis and management. Exp Rev Dermatol. 2006; 1: 401-411.
  • Olasz EB, Yancey KB. Bullous pemphigoid and related subepidermal autoimmune blistering diseases. Curr Dir Autoimmun. 2008; 10: 141-166.
  • Jordon RE, Beutner EH, Witebsky E, Blumental G, Hale WL, Lever WF. Basement zone antibodies in bullous pemphigoid. JAMA. 1967; 200: 751-756.
  • Stanley JR. Cell adhesion molecules as targets of autoantibodies in pemphigus and pemphigoid, bullous diseases due to defective epidermal cell adhesion. Adv Immunol. 1992; 53: 291-325.
  • 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.
  • Alexander JO. Dermatitis herpetiformis. W: Major problems in dermatology. WB Saunders Company Ltd, London, 1975; 47-56.
  • Beutner EH, Chorzelski TP, Bean SF, Jordon R. Dermatitis herpetiformis: Basic Findings w: Immunopathology of the skin. Wiley Publication New York, 1979; 283-301.
  • Gawkrodger DJ, Blakwell JN, Gilmour HM. Dermatitis herpetiformis: diagnosis, diet and dermography. Gut 1984; 25: 151-157.
  • Reunala T. Dermatitis herpetiformis. Clin Dermatol. 2001; 19: 728-736.
  • Wankiewicz A, Gwieździński Z. Zapalenie opryszczkowate skóry w świetle wieloletnich badań. Przegl Dermatol. 1999; 5: 491-497.
  • Garioch JJ, Baker BS, Leonard JN, Fry L. T lymphocytes in lesional skin of patients with dermatitis herpetiformis. Br J Dermatol. 1994; 131: 822-826.
  • Dieterich W, Ehnis T, Bauer M, Donner P, Volta U, Riecken EO i wsp. Identification of tissue transglutaminase as the autoantigen of celiac disease. Nat Med. 1997; 3: 797-801.
  • Dieterich W, Laag E, Bruckner-Tuderman L, Reunala T, Kárpáti S, Zágoni T i wsp. Antibodies to tissue transglutaminase as serologic markers in patients with dermatitis herpetiformis. J Invest Dermatol. 1999; 113: 133-136.
  • Malnick S, Beergabel M. Celiakia - Przesłanki diagnostyczne w przypadkach nietypowych. Medycyna po dyplomie. 1998; 9: 73-77.
  • Rosińska-Borkowska D. Choroby pęcherzowe o podłożu autoimmunologicznym. W: Dermatologia pediatryczna. red. Miklaszewska M, Wąsik F. Volumed Wrocław, 1999; 15-37.
  • Gammon WR. Dermatologic immunology and allergy. Julius Stone. St. Louis 1985; 457-493.
  • Marks J, Shuster S, Watson AJ. Small bowell changes in dermatitis herpetiformis. Lancet. 1966; 2: 1280-1284.
  • Witas H, Młynarski J. Immune response in coeliac disease. Cent Eur Jour of Immunol. 2000; 25, 4: 180-184.
  • Rose C, Dieterich W, Bröcker EB, Schuppan D, Zillikens D. Circulating autoantibodies to tissue transglutaminase differentiate patients with dermatitis herpetiformis from these with linear IgA disease. J Am Acad Dermatol. 1999; 41: 957-961.
  • Chorzelski TP, Beutner EH, Sulej J, Tchorzewska H, Jablonska S, Kumar V i wsp. IgA anti-endomysium antibody. A new immunological marker of dermatitis herpetiformis and coeliac disease. Br J Dermatol. 1984; 111: 395-402.
  • Kolho KL, Farkkila MA, Savilahti E. Undiagnosed coeliac disease is common in Finnish adults. Scand J Gastroentrol. 1998; 33: 1280-1283.
  • Mäki M, Holm K, Collin P, Savilahti E. Increase in γ/δ T cell receptor bearning lymphocytes in normal small bowel mucosa in latent celiac disease. Gut. 1991; 32: 1412-1414.
  • Wojnarowska F, Briggaman R. Managment of Blistering Diseases. New York 1990; 139-160.
  • Sárdy M, Odenthal U, Kárpáti S, Paulsson M, Smyth N. Recombinant human tissue transglutaminase ELISA for the diagnosis of gluten-sensitive enteropathy. Clin Chem. 1999; 45: 2142-2149.
  • Dieterich W, Laag E, Schöpper H, Volta U, Ferguson A, Gillett H i wsp. Autoantibodies to tissue transglutaminase as predictors of celiac disease. Gastroenterology. 1998; 115: 1317-1321.
  • Sulkanen S, Halttunen T, Laurila K, Kolho KL, Korponay-Szabó IR, Sarnesto A i wsp. Tissue transglutaminase autoantibody enzyme-linked immunosorbent assay in detecting celiac disease. Gastroenterology. 1998; 115: 1322-1328.
  • Sárdy M, Kárpáti S, Merkl B, Paulsson M, Smyth N. Epidermal transglutaminase (Tgase 3) is the autoantigen of dermatitis herpetiformis. J Exp Med. 2002; 195: 747-757.
  • Aeschliman D, Paulsson M. Cross-lining of laminin-nidogen complexes by tissue transglutaminase. A novel mechanism for basement membrane stabilization. J Biol Chem. 1991; 266: 15308-15317.
  • Kleman JP, Aeschlimann D, Paulsson M, van der Rest M. Transglutaminase-catalyzed cross-linking of fibrils of collagen V/XI in A204 rhabdomyosarcoma cells. Biochemistry. 1995; 34: 13768-13775.
  • Aeschliman D, Paulsson M, Mann K. Identification of Gln726 in nidogen as the amine acceptor in transglutaminase-catalyzed cross-links of laminin-nidogen complexes. J Biol Chem. 1992; 267: 11316-11321.
  • Aeschliman D, Kaupp O, Paulsson M. Osteonectin is a major glutaminyl substrate for transglutaminase-catalyzed cross-linking in cartilage matrix. J Cell Biol. 1995; 129: 881-892.
  • Martinez J, Chalupowicz R, Roush A. Transglutaminase-mediated processing of fibronectin by endothelial cell monolayers. Biochemistry 1994; 33: 2538-2545.
  • Franzke CW, Tasanen K, Schäcke H, Zhou Z, Tryggvason K, Mauch C i wsp. Transmembrane collagen XVII, an epithelial adhesion protein, is shed from the cell surface by ADAMs. EMBO J. 2002; 21: 5026-5035.
  • Zone J, Meyer L, Petersen M. Deposition of granular IgA relative to clinical lesions in dermatitis herpetiformis. Arch Dermatol. 1996; 132: 912-918.
  • Gur P, Kaushal Sudhir V. The new kids on the block: ADAMTSs potentialy multifunctional metalloproteinases of the ADAM family. J Clin Invest. 2000; 105: 1335-1337.
  • Reitamo S, Reunala T, Konttinen YT, Saksela O, Salo OP. Inflammatory cells, IgA, C3, fibrin and fibronectin in skin lesions in dermatitis herpetiformis. Br J Dermatol. 1981; 105: 167-177.
  • Fry L, Seah PP, Riches DJ, Hoffbrand AV. Clearance of skin lesions in dermatitis herpetiformis after gluten
  • Reunala T, Blomqvist K, Tarpila S, Halme H, Kangas K. Gluten-free diet in dermatitis herpetiformis. I. Clinical response of skin lesions in 81 patients. Br J Dermatol. 1977; 97: 473-480.
  • Garioch JJ, Lewis HM, Sargent SA, Leonard JN, Fry L. 25 years’ experience
  • of a gluten-free diet in the treatment of dermatitis herpetiformis. Br J Dermatol. 1994; 9: 403-408.
  • Leonard JN, Fry L. Treatment and management of dermatitis herpetiformis. Clin Dermatol. 1991; 9: 404-408
  • Burgeson RE, Christiano AM. The dermal-epidermal junction. Curr Opin Cell Biol. 1997; 9: 651-658.
  • Chan RY, Bhol K, Tesavibul N, Letko E, Simmons RK, Foster CS i wsp. The role of antibody to human beta4 integrin in conjunctival basement membrane separation: possible in vitro model for ocular cicatricial pemphigoid. Invest Ophthalmol Vis Sci. 1999; 40: 2283-2290.
  • Borradori L, Sonnenberg A. Structure and function of hemidesmosomes: More than simple adhesion complexes. J Invest Dermatol. 1999, 112: 411-418.
  • Okumura Y, Sato H, Seiki M, Kido H. Proteolytic activation of membrane type 1 matrix metalloproteinase by human plasmin. A possible cell surface activator. FEBS Lett. 1997; 402: 181-184.
  • Hirako Y, Owaribe K. Hemidesmosomes and their unique transmembrane protein BP180. Microsc Res Tech. 1998; 43: 207-217.
  • Jones JC, Hopkinson SB, Goldfinger LE. Structure and assembly of hemidesmosomes. Bioessays. 1998; 20: 488-494.
  • Moll R, Moll I. Epidermal adhesion molecules and basement membrane components as target structures of autoimmunity. Virchows Arch. 1998; 432: 487-450
  • Guo L, Degenstein L, Dowling J, Yu QC, Wollmann R, Perman B i wsp. Gene targeting of BPAG1: abnormalities in mechanical strength and cell migration in stratified epithelia and neurologicdegeneration. Cell. 1995; 81: 233-243.
  • McMillan JR, McGrath JA, Tidman MJ, Eady RA. Hemidesmosomes show abnormal association with the keratin filament network in junctional forms of epidermolysis bullosa. J Invest Dermatol. 1998; 100: 132-137.
  • Uitto J, Pulkkinen L. Molecular complexity of the cutaneous basement membrane zone. Mol Biol Rep. 1996; 23: 35-46.
  • Tasanen K, Eble JA, Aumailley M, Schumann H, Baetge J, Tu H i wsp. Collagen XVII is destabilized by a glycine substitution mutation in the cell adhesion domain Col15. J Biol Chem. 2000; 275: 3093-3099.
  • Schumann H, Baetge J, Tasanen K, Wojnarowska F, Schäcke H, Zillikens D. The shed ectodomain of collagen XVII/BP180 is targed by autoantibodies in different blistering skin diseases. Am J Pathol. 2000; 156: 685-695.
  • Goldberg DJ, Sabolinski M, Bystryn JC. Regional variation in the expression of bullous pemphigoid antigen and location of lesions in bullous pemphigoid. J Invest Dermatol. 1984; 82: 326-328.
  • Shimizu H. New insights into the immunoultrastructural organization of cutaneous basement membrane zone molecules. Exp Dermatol. 1998; 7: 303 313.
  • Borradori L, Chavanas S, Schaapveld RQJ, Gagnoux-Palacios L, Calafat J, Meneguzzi G i wsp. Role of the bullous pemphigoid antigen 180 (BP180)
  • in the assembly of hemidesmosomes and cell adhesion - reexpression of BP180 in generalized atrophic benign epidermolysis bullosa keratinocytes. Exp Cell Res. 1998; 239: 463-476.
  • Woodley D, Sauder D, Talley MJ, Silver M, Grotendorst G, Qwarnstrom E. Localization of basement membrane components after dermal-epidermal junction separation. J Invest Dermatol. 1983; 81: 149-153.
  • Giancotti FG. Signal transduction by the α6β4 integrin: charting the path between laminin binding and nuclear events. J Cell Sci. 1996; 109: 1165-1172.
  • Spinardi L, Einheber S, Cullen T, Milner TA, Giancotti FG. A recombinant tail-less integrin β4 subunit disrupts hemidesmosomes, but does not suppress α6β4-mediated cell adhesion to laminins. J Cell Biol. 1995; 129: 473-487.
  • Champliaud MF, Lunstrum GP, Rousselle P, Nishiyama T, Keene DR, Burgeson RE. Human amnion contains a novel laminin variant, laminin 7, which like laminin 6, covalently associates with laminin 5 to promote stable epithelial-stromal attachment. J Cell Biol. 1996; 132: 1189-1198.
  • Zhang K, Kramer RH. Laminin 5 deposition promotes keratinocyte motility. Exp Cell Res. 1996; 227: 309-322.
  • O´Toole EA, Marinkovich MP, Hoeffler WK, Furthmayr H, Woodley DT. Laminin-5 inhibits human keratinocyte migration. Exp Cell Res. 1997; 233: 330-333.
  • Koshikawa N, Giannelli G, Cirulli V, Miyazaki K, Quaranta V. Role of cell surface metalloprotease MT1-MMP in epithelial cell migration over laminin 5. J Cell Biol. 2000; 148: 615-624.
  • Miner JH, Patton BL, Lentz SI, Gilbert DJ, Snider WD, Jenkins NA i wsp. The laminin αchains: Expression, developmental transitions, and chromosomal locations of α1- 5, identification of heterotrimeric laminins 8-11, and cloning of a novel α3 isoform. J Cell Biol. 1997; 137: 685-701.
  • Virtanen I, Lohi J, Tani T, Korhonen M, Burgeson RE, Lehto VP i wsp. Distinct changes in the laminin composition of basement membranes in human seminiferous tubules during development and degeneration. Am J Pathol. 1997; 150: 1421-1431.
  • Eble JA, Wucherpfennig KW, Gauthier L, Dersch P, Krukonis E, Isberg RR i wsp. Recombinant soluble human α3β1 integrin: purification, processing, regulation, and specific binding to laminin-5 and invasin in a mutually exclusive manner. Biochemistry. 1998; 37: 10945-10955.
  • Koch M, Olson PF, Albus A, Jin W, Hunter DD, Brunken WJ i wsp. Characterization and expression of the laminin α3 chain: a novel, non-basement membrane-associated, laminin chain. J Cell Biol. 1999; 145: 605-617.
  • Aumailley M, Smyth N. The role of laminins in basement membrane function. J Anat. 1998; 193: 1-21
  • Ninomiya Y, Kagawa M, Iyama K, Naito I, Kishiro Y, Seyer JM i wsp. Differential expression of two basement membrane collagen genes, COL4A6 and COL4A5, demonstrated by immunofluorescence staining using peptide-specific monoclonal antibodies. J Cell Biol. 1995; 130: 1219-1229.
  • Tanaka K, Iyama KI, Kitaoka M, Ninomiya Y, Oohashi T, Sado Y i wsp. Differential expression of α1(IV), α 2(IV), α 5(IV) and α 6(IV) collagen chains in the basement membrane of basal cell carcinoma. Histochem J. 1997; 29: 563 570.
  • Timpl R, Brown JC. Supramolecular assembly of basement membranes. Bioessays. 1996; 18: 123-132.
  • Mayer U, Kohfeldt E, Timpl R. Structural and genetic analysis of laminin-nidogen interaction. Ann N Y Acad Sci. 1998; 857: 130-142.
  • Meredith JE Jr, Winitz S, Lewis JM, Hess S, Ren XD, Renshaw MW i wsp. The regulation of growth and intracellular signaling by integrins. Endocr Rev. 1996; 17: 207-220.
  • Sheppard D. Epithelial integrins. Bioessays. 1996; 18: 655-660.
  • Kim JP, Zhang K, Chen JD, Kramer RH, Woodley DT. Vitronectin-driven human keratinocyte locomotion is mediated by the αvβ5 integrin receptor. J Biol Chem. 1994; 269: 26926-26932
  • Yokosaki Y, Matsuura N, Higashiyama S, Murakami I, Obara M, Yamakido M i wsp. Identification of the ligand binding site for the integrin α9β1 in the third fibronectin type III repeat of tenascin-C. J Biol Chem. 1998; 273: 11423-11428.
  • Palmer EL, Rüegg C, Ferrando R, Pytela R, Sheppard D. Sequence and tissue distribution of the integrin α9 subunit, a novel partner of β1 that is widely distributed in epithelia and muscle. J Cell Biol. 1993; 123: 1289-1297.
  • Stepp MA, Spurr-Michaud S, Tisdale A. a6b4 integrin heterodimer is a compo-nent of hemidesmosomes. Proc Natl Acad Sci 1990; 87: 8970-8974.
  • Prieto AL, Edelman GM, Crossin KL. Multiple integrins mediate cell attachment to cytotactin/tenascin. Proc Natl Acad Sci U S A. 1993; 90: 10154-10158.
  • Schmidt E, Reimer S, Kruse N, Jainta S, Bröcker EB, Marinkovich MP Autoantibodies to BP 180 associated with bullous pemphigoid relase interleukin-6 and interleukin-8 from cultured human keratynocytes. J Invest Dermatol. 2000; 115: 842-848.
  • Borradori L, Koch PJ, Niessen CM, Erkeland S, van Leusden MR, Sonnenberg A. The localization of bullous pemphigoid antigen 180 (BP180) in hemidesmosomes is mediated by its cytoplasmic domain and seems to be regulated by the beta4 integrin subunit. J Cell Biol. 1997; 136: 1333-1347.
  • Grando SA, Glukhenky BT, Drannik GN, Epshtein EV, Kostromin AP, Korostash TA. Mediators of inflammation in blister fluids from patients with pemphigus vulgaris and bullous pemphigoid. Arch Dermatol. 1989; 125: 925 930.
  • Oikarinen AI, Zone JJ, Ahmed AR, Kiistala U, Uitto J. Demonstration of collagenase and elastase activities in the blister fluids from bullous skin diseases. Comparison between dermatitis herpetiformis and bullous pemphigoid. J Invest Dermatol. 1983; 81: 261-266.
  • Baker BS, Garioch JJ, Bokth S, Leonard J, Fry L. Absence of gluten specific T lymphocytes in the skin of patients with dermatitis herpetiformis. J Autoimmun. 1995; 8: 75-82.
  • Caproni M, Feliciani C, Fuligni A, Salvatore E, Atani L, Bianchi B i wsp. Th2-like cytokine activity in dermatitis herpetiformis. Br J Dermatol. 1998; 138: 242-247.
  • Graeber M, Baker BS, Garioch JJ, Valdimarsson H, Leonard JN, Fry L. The role of cytokines in the generation of skin lesions in dermatitis herpetiformis.
  • Br J Dermatol. 1993; 129: 530-532.
  • Hendrix JD, Mangum KL, Zone JJ, Gammon WR. Cutaneous IgA deposits in bullous diseases function as ligands to mediate adherence of activated neutrophils. J Invest Dermatol. 1990; 94: 667-672.
  • Dahl MV, Falk RJ, Carpenter R, Michael AF. Membrane attack complex of complement in dermatitis herpetiformis. Arch Dermatol. 1985; 121: 70-72.
  • Lorand L, Conrad SM. Transglutaminases. Mol Cell Biochem. 1984; 58: 9-35.
  • Tyrell D, Sale W. Fibronectin is a component of the sodium dodecyl sulfate- in soluble transglutaminase substrate. J Biol Chem. 1988; 263: 8468-8469.
  • Freemont AJ. Demystified … adhesion molecules. Mol Pathol. 1998, 51: 175 184.
  • Parish WE. Inflammation. W: Rook, Wilkinson, Ebling Textbook of Dermatology. Champion RH, Burton JL, Burner DA, Breathnach SM. Sixth edition, Blackwell Science Ltd. Oxford, London, Edinburgh, Massachusetts. 1998; 229-276.
  • Hynes RO. Integrins: Versatility, modulation and signaling in cell adhesion. Cell. 1992; 69: 11-25.
  • Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell. 2002; 110: 673-687.
  • Hynes RO. Integrins: a family of cell surface receptors. Cell. 1987; 48: 549-554.
  • Bouvard D, Brakebusch C, Gustafsson E, Aszódi A, Bengtsson T, Berna A i wsp. Functional consequences of integrin gene mutations in mice. Circ Res. 2001, 89: 211-223.
  • Ginsberg MH, Du X, Plow EF. Inside-out integrin signaling. Curr Opin Cell Biol.1992; 4: 766-771.
  • O'Toole TE, Katagiri Y, Faull RJ, Peter K, Tamura R, Quaranta V i wsp. Integrin cytoplasmic domains mediate inside-out signal transduction. J Cell Biol. 1994; 124: 1047-1059.
  • Stepp MA, Zhu L Cranfill R. Changes in beta 4 integrin expression and localization in vivo in response to corneal epithelial injury. Invest Ophthalmol Vis Sci. 1996; 37: 1593-1601.
  • Andrian UH, Engrlhardt B: a4 integrins as therapeutic targets in autoimmune disease. N Eng J Med. 2003; 348: 65-69.
  • Plow EF, Haas TA, Zhang L, Loftus J, Smith JW. Ligand binding to Integrins. J Bilol Chem. 2000; 275: 21785-21788.
  • Berman AE, Kozlovz NI, Morozevich GE. Integrins: structure and signaling. Biochemistry. 2003;1284-1299.
  • Eody RAJ. The hemidesmosome: a target in autoimmunmo bullous disease. Dermatology 1994; 189(suppl 1): 38-41.
  • Xiong JP, Stehle T, Goodman SL, Arnaout MA. New insights into the structural basis of integrin activation. Blood. 2003; 102: 1155-1159.
  • Stupack DG, Cheresh DA. Get a ligand, get a life: integrins, signaling and cell survival. J Cell Sci. 2002; 115: 3729-3738.
  • Sterk LM, Geuijen CA, Oomen LC, Calafat J, Janssen H, Sonnenberg A. The tetraspan molecule CD151, a novel constituent of hemidesmosomes, associates with the integrin alpha6beta4 and may regulate the spatial organization of hemidesmosomes. J Cell Biol. 2000; 149: 969-982.
  • Marchisio PC, Bondanza S, Cremona O, Cancedda R, De Luca M. Polarized expression of integrin receptors and their relationship with the cytoskeleton and basement membrane matrix in cultured human keratinocytes. J Cell Biol. 1991; 112: 761-773.
  • Schaapveld RQ, Borradori L, Geerts D, van Leusden MR, Kuikman I, Nievers MG i wsp. Hemidesmosome formation is initiated by the beta4 integrin subunit, requires complex formation of beta4 and HD1/plectin, and involves a direct interaction between beta4 and the bullous pemphigoid antigen 180. J Cell Biol. 1998; 142: 271-284.
  • Hopkinson SB, Findlay K, deHart GW, Jones JCR. Interaction of BP180 (type XVII collagen) and a6 integrin is necessary for stabilization of hemidesmosome structure. J Invest Dermatol. 1988; 111: 1015-1022.
  • Koster J, Geerts D, Favre B, Borradori L, Sonnenberg A. Analysis of interactions between BP180, BP230, plectin and the integrin a6b4 important for hemidesmosome assembly. J Cell Sci. 2003; 116: 387-389.
  • Leverkus M, Bhol K, Hirako Y, Pas H, Sitaru C, Baier G i wsp. Cicatrical pemphigoid with circulating autoantibodies to b4 integrin, bullous pemphigoid 180 and bullous pemphigoid 230. Br J Dermatol. 2001; 145: 998-1004.
  • Kumari S, Bhol KC, Simmons RK, Razzaque MS, Letko E, Foster CS i wsp. Identification of ocular cicatrical pemphigoid antibody binding site(s) in human beta4 integrin. Invest Ophthalmol Vis Sci. 2001; 42: 379-385.
  • Liu Z, Zhao M, Li N, Diaz LA, Mayadas TN. Differential roles for b2 integrins in experimental autoimmune bullous pemphigoid. Blood. 2006; 107: 1063-1069.
  • Kuhns DB, DeCarlo E, Hawk DM, Gallin JI. Dynamics of the cellular and humoral components of the inflammatory responce elicited in skin blisters in humans. J Clin Invest. 1992; 89: 1734-1740.
  • Ameglio F, D'Auria L, Bonifati C, Ferraro C, Mastroianni A, Giacalone B. Cytokine pattern in blister fluid and serum of patients with bullous pemphigoid: relationship with disease intensity. Br J Dermatol. 1998; 138: 611-614.
  • Kristensen M, Larsen CG, Jørgensen P, Paludan K. RNA purification from epidermal suction blisters. Acta Derm Venereol. 1991; 71: 423-426.
  • D’Auria L, Pietravalle M, Cordiali-Fei P, Ameglio F. Increased tryptase and myeloporoxudase levels in blister fluids of patients with bullous pemphogoid: correlation with cytokines, adhesion molecules and anti-basement membrane zone antibodies. Exp Dermatol. 2000; 9: 131-137.
  • Wakugawa M, Nakamura K, Hino H, Toyama K, Hattori N, Okochi H i wsp. Elevated levels of eotaxin and interleukin-5 in blister fluid of bullous pemphigoid: correlation with tissue eosynophilia. Br J Dermatol. 2000; 143: 112 116.
  • Shrikhande M, Hunziker T, Braathen LR, Pichler WJ, Dahinden CA, Yawalkar N. Increased Coexpression of Eotaxin and Interleukin 5 in Bullous Pemphigoid. Acta Derm Venerol. 2000; 80: 277-280.
  • Kakinuma T, Wakugawa M, Nakamura K, Hino H, Matsushima K, Tamaki K. High level of thymus and activation-regulated chemokine in blister fluid and sera of patients with bullous pemphigoid. Br J Dermatol 2003; 148: 203-210.
  • Sallusto F, Lenig D, Mackay CR, Lanzavecchia A. Flexible programs of chemokine receptors expression on human polarized T helper 1 and 2 lymphocytes. J Exp Med. 1998; 187: 857-883.
  • Amerio P, Verdolini R, Giangiacomi M, Proietto G, Feliciani C, Offidani A i wsp. Expression of eotaxin, interleukin 13 and tumor necrosis factor-α in dermatitis herpetiformis. Br J Dermatol. 2000; 143: 974-978.
  • Caproni M, Cardinalli C, D’Agata A, Selvaggi W, Fabbri P. Serum eosinophil cationic protein, myeloperoxidase, tryptase, eotaxin and Th2-L-like cytokines in dermatitis herpetiformis. Int Arch Allerg Immunol. 2002; 128: 67-72.
  • Oikarinen AI, Reunala T, Zone JJ, Kiistala U, Uitto J. Proteolytic enzymes in blister fluids from patients with dermatitis herpetiformis. Br J Dermatol. 1986; 114: 295-230.
  • Airola K, Vaalamo M, Reunala T, Saarialho-Kere UK. Enhanced expresion of intertitial collagenase, stromelysin-1 and urokinase plasminogen activator in lesions of dermatitis herpetiformis. J Invest Dermatol. 1995; 15: 184-189.
  • Baggioloni M. Chemokines in pathology and medicine. J Inter Med. 2001; 250: 91-104.
  • Hall RP 3rd, Takeuchi F, Benbenisty KM, Streilein RD. Cutaneous endothelial cell activation in normal skin of patients with dermatitis herpetiformis associated with increased serum levels of IL-8, sE-selectin, and TNF-α. J Invest Dermatol. 2006; 126: 1331-1337.
  • Bornscheuer E, Zillikens D, Schrőder JM, Sticherling M. Lack of expression of Interleukin 8 and RANTES in autoimmune bullous skin diseases. Dermatology. 1999; 198: 118-121.
  • Kaneko F, Minagawa T, Takiguchi Y, Suzuki M, Itoh N. Role of Cell-Mediated Immune Reaction in Blister Formation of Bullous Pemphigoid. Dermatology. 1992; 184: 34-39.
  • Saeki H, Tamaki K. Thymus and activation regulated chemokine (TARC)/CCL17 and skin diseases. J Dermatol Sci. 2006; 43: 75-84.
  • Zlotnik A, Yoshie O. Chemokines. A new classification system and their role in immunity. Immunity. 2000; 12: 121-127.
  • Waśniowska K. Ludzkie receptory chemokin: budowa i funkcja. Post Hig Med Dosw. 1999; 53: 583-600.
  • Palová-Jelínková L, Rozková D, Pecharová B, Bártová J, Sedivá A, Tlaskalová-Hogenová H i wsp. Gliadin fragments induce phenotypic and functional maturation of human dendritic cells. J Immunol. 2005; 175: 7038-7045.
  • Chowers Y, Marsh MN, De Grandpre L, Nyberg A, Theofilopoulos AN, Kagnoff MF. Increased proinflammatory cytokine gene expression in the colonic mucosa of coeliac disease patients in the early period after gluten challenge. Clin Exp Immunol. 1997; 107: 141-147.
  • Hub E, Rot A. Binding of RANTES, MCP-1, MCP-3, and MIP-1a to cells in human skin. Am J Pathol. 1998; 152: 749-757.
  • Cinova J, Palová-Jelínková L, Smythies LE, Cerná M, Pecharová B, Dvorák M i wsp. Gliadin peptides activate blood monocytes from patients with celiac disease. J Clin Immunol. 2007; 27: 201-209.
  • Greaves MW. Recent advances in pathophysiology and current management of itch. Ann Acad Med Singapore. 2007; 36: 788-792.
  • Rothman S. Physiology of itching. Physiol Rev. 1941, 21: 357-381.
  • Arck P, Paus R. From the brain-skin connection: the neuroendocrine-immune misalliance of stress and itch. Neuroimmunomodulation. 2006; 13: 347-356.
  • Steinhoff M, Ständer S, Seeliger S, Ansel JC, Schmelz M, Luger T. Modern aspects of cutaneous neurogenic inflammation. Arch Dermatol. 2003; 139: 1479 1488.
  • Pincelli C, Fantini F, Giannetti A. Neuropeptides and skin inflammation. Dermatology. 1993; 187: 153-158.
  • Luger TA. Neuromediators - a crucial component of the skin immune system. J Drmatol Sci. 2002; 30: 87-93.
  • Słomski A, Wortsman J. Neuroendocrinology of the Skin. Endocr Rev. 2000; 21: 457-487.
  • Lotti T, Hautmann G, Panconesi E. Neuropeptides in skin. J Am Acad Dermatol. 1995; 33: 482-496.
  • Weldon D. What lies beneath the surface of the itch in adults? Allergy Asthma Proc. 2007; 28: 153-162.
  • Nagase H. Activation mechanisms of matrix metalloproteinases. Biol Chem. 1997; 378: 151-160.
  • Reynolds JJ. Collagenases and tissues inhibitors of metalloproteinases: functional balance in tissue degradation. Oral Dis. 1996; 2: 70-76.
  • Pauschinger M, Chandrasekharan K, Li J, Schwimmbeck PL, Noutsias M, Schultheiss HP. Mechanisms of extracellular matrix remodeling in dilated cardiomyopathy. Herz. 2002; 27: 677-682.
  • Reynolds JJ. Connective tissue degradation in health and periodontal disease and the roles of metalloproteinases and their natural inhibitors. Adv Dent Res. 1994; 8: 312-319.
  • Reynolds JJ, Meikle MC. The funcional blance of metalloproteinases and inhibitors in tissue degradation: relevance to oral pathologies. J R Coll Surg Edinb. 1997; 42: 154-160.
  • Airola K, Ahonen M, Johansson N, Heikkilä P, Kere J, Kähäri VM i wsp. Human TIMP-3 is expressed during fatal development, hair growth cycle and cancer progression. J Histochem Cytochem. 1998; 46: 437-447.
  • Birkedal Hansen H. Role of cytokines and inflammatory mediators in tissue destruction. J Periodontal Res. 1993; 28: 500-510.
  • Ruhul Amin AR, Senga T, Oo ML, Thant AA, Hamaguchi M. Secretion of matrix metalloproteinase-9 by the proinflammatory cytokine, IL-1beta: a role for the dual signalling pathways, Akt and Erk. Genes Cells. 2003; 8: 515-523.
  • Larry M, Corcoran W. Regulation of Monocyte/Macrophage Metalloproteinase Production by Cytokines. J Periodontol. 1993; 64: 467-473.
  • Tummalapalli CM, Heath BJ, Tyagi SC. Tissue inhibitor of metalloproteinase-4 instigates apoptosis in transformed cardiac fibroblasts. J Cell Biochem. 2001; 80: 512-521.
  • Bhide M. Use of Fluorogenic Septapeptide Matrix Metalloproteinase Assay to Assess Responses to Periodontal Treatment. J Periodontol. 2000; 71: 690-700.
  • Reinhardt D, Sigusch HH, Hensse J, Tyagi SC, Körfer R, Figulla HR. Cardiac remodelling in end stage heart failure: upregulation of matrix metalloproteinase (MMP) irrespective of the underlying disease, and evidence for a direct inhibitory effect of ACE inhibitors on MMP. Heart. 2002; 88: 525-530.
  • Korostoff JM. Analysis of in situ protease activity in chronic adult periodontitis patients: expression of activated MMP-2 and a 40-kD serine protease. J Periodontol. 2000; 71: 353-360.
  • Cleutjens JP, Creemers EE. Integration of concepts: cardiac extracellular matrix remodeling after myocardial infarction. J Card Fail. 2002; 8(suppl 6): 344-348.
  • Caterson B. Mechanisms involved in cartilage proteoglycan catabolism. Matrix Biol. 2000; 19: 333-344.
  • Leco KJ, Khokha R, Pavloff N, Hawkes SP, Edwards DR. Tissue inhibitor of metalloproteinases-3 (TIMP-3) is an extracellular matrix-associated protein with a distinctive pattern of expresion in mouse cells and tissues. J Biol Chem. 1994; 269: 9352-9360.
  • Lohmander LS. Metalloproteinases, tissue inhibitor and proteoglycan fragments in knee synovial fluid in human osteoarthritis. Arthritis Rheum. 1993; 36: 181-189.
  • Stearns M, Rhim J, Wang M. Interleukin-10 (IL-10) inhibition of primary human prostate cell- induced angiogenesis: IL-10 stimulation of tissue inhibitor of metalloproteinase-1 and inhibition of matrix metalloproteinase (MMP)-2/MMP-9 secretion. Clin Cancer Res. 1999; 5: 189-196.
  • Martel-Pelletier J. Excess of metalloproteases over tissue inhibitor of metalloprotease may contribute to cartilage degradation in osteoarthritis and rheumatoid arthritis. Lab Invest. 1994; 70: 807-815.
  • Campbell IK. Recombinant human interleukin-1 stimulates human articular cartilage to undergo resorption and human chondrocytes to produce both tissue- and urokinase-type plasminogen activator. Biochem Biophys Acta. 1988; 967: 183-194.
  • Goldring MB. The role of cytokines as inflammatory mediators in osteoarthritis: lessons from animal models. Connect Tissue Res. 1999; 40: 1-11.
  • Su S, Grover J, Roughley PJ, DiBattista JA, Martel-Pelletier J, Pelletier JP. i wsp. Expression of the tissue inhibitors of metalloproteinases (TIMPs) gene family in normal and osteoarthritic joints. Rheumatol Int. 1999; 18: 183-191.
  • Dean DD. Evidence for metalloproteinase and metalloproteinase inhibitor imbalance in human osteoarthritic cartilage. J Clin Invest. 1989; 84: 678-685.
  • Baumann G, Frank SJ. Metalloproteinases and the modulation of GH signaling. Journal of Endocrinol. 2002; 174: 361-368.
  • Gomis-Rüth FX. Structural aspects of metzincin clan of metalloendopeptidases. Mol Biotechnol. 2003; 24: 157-202.
  • Schlöndorff J, Blobel CP. Metalloprotease-disintegrins: modular proteins capable of promoting cell-cell interaction and triggering signals by protein-ectodomain shedding. J Cell Sci. 1999; 112: 3603-3617.
  • Stone AI, Kroeger M, Xiang QX. Structure-function analysis of the ADAM family of disintegrin-like and metalloproteinase-containing proteins. J Protein Chem. 2000; 18: 447-465.
  • Primakoff P, Myles DG. The ADAM gene family: surface proteins with adhesion and protease activity. Trends Genet. 2000; 16: 83-87.
  • Karan D, Lin FC, Bryan M, Ringel J, Moniaux N, Lin MF i wsp. Expression of ADAMs (a disintegrin and metalloproteases) and TIMP-3 (tissue inhibitor of metalloproteinase-3) in human prostatic adenocarcinomas.International Int J of Oncol. 2003; 23: 1365-1371.
  • Schäcke H, Schumann H, Hammami-Hauasli N, Raghunath M, Bruckner-Tuderman L. Two Forms of Collagen XVII in Keratinocytes a full-length transmembrane protein and soluble ectodomain. J Biol Chem. 1998; 273: 25937-25943.
  • Hirako Y, Usukura J, Uematsu J, Hashimoto T, Kitajima Y, Owaribe K. Cleavage of BP180, a 180-kD Bullous Pemphigoid Antigen, Yields a 120-kD Collagenous Extracellular Polypeptide. J Biol Chem. 1998; 273: 9711-9717.
  • Franzke CW, Tasanen K, Borradori L, Huotari V, Bruckner-Tuderman L. Shedding of collagen XVII/BP180: structural motifs influence cleavage from cell surface. J Biol Chem. 2004; 279: 24521-24529.
  • Kuno K. Molecular cloning of gene encoding a new type of metalloproteinases – disintegrin family protein with trombospondin motifs as an inflammation associated gene. J Biol Chem. 1998; 272: 556-562.
  • Cerretti DP, Poindexter K, Castner BJ, Means G, Copeland NG, Gilbert DJ i wsp. Characterization of the cDNA and gene for mouse tumour necrosis factor alpha converting enzyme (TACE/ADAM17) and its location to mouse chromosome 12 and human chromosome 2p25. Cytokine. 1999; 11: 541-551.
  • Marzano AV, Tedeschi A, Spinelli D, Fanoni D, Crosti C, Cugno M. Coagulation activation in autoimmune bullous diseases. Clin Exp Immunol. 2009; 158: 31-36.
  • Marzano AV, Tedeschi A, Fanoni D, Bonanni E, Venegoni L, Berti E i wsp. Activation of blood coagulation in bullous pemphigoid: role of eosinophils, and local systemic implications. Br J Dermatol. 2009; 160: 266-272.
  • De Quatrebarbes J, Joly P. Bullous pemphigoid. Rev Prat. 2005; 55: 1165-1168.
  • Rico MJ, Benning C, Weingart ES, Streilein RD, Hall RP 3rd. Characterization of skin cytokines in bullous pemphigoid and pemphigus vulgaris. Br J Dermatol. 1999; 140: 1079-1086.
  • Engineer L, Bhol K, Kumari S, Razzaque Ahmed A. Bullous pemphigoid: interaction of interleukin 5 antibasement membrane zone antibodies and eosinophils. A preliminary observation. Cytokine. 2001; 13: 32-38.
  • Gerber BO, Zanni MP, Uguccioni M, Loetscher M, Mackay CR, Pichler WJ. Functional expression of the eotaxin receptor CCR3 in T lymphocytes colocalizing with eosinophils. Curr Biol. 1997; 7: 836-843.
  • Venge P, Byström J, Carlson M, Hâkansson L, Karawacjzyk M, Peterson C i wsp. 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.
  • Kita H, Abu-Ghazaleh R, Sanderson CJ, Gleich GJ. Effect of steroids on immunoglobulin–induced eosinophil degranulation. J Allergy Clin Immunol. 1991; 87: 70-77.
  • Muller YA, Ultsch MH, Kelley RF, deVos AM. Structure of the extracellular domain of human tissue factor: location of the factor VIIa binding sile. Biochemistry. 1994; 33: 10864-10867.
  • Moosbauer C, Morgenstern E, Cuvelier SL, Manukyan D, Bidzhekov K, Albrecht S i wsp. Eosinophils are a major intravascular location for tissue factor storage and exposure. Blood. 2007; 109: 995-1002.
  • Levi M, van der Poll T, Ten Cate H, van Deventer SJ. The cytokine – mediated imbalance between coagulant ant
  • Tremoli E, Camera M, Toschi V, Colli S. Tissue factor in atherosclerosis. Atherosclerosis. 1999; 144: 273-283.
  • Gouin–Thibault I, Achkar A, Samama MM. The trombophilic state in cancer patients. Acta Haematol 2001; 106: 33-42.
  • Debaugnies F, Azerad MA, Noubouossié D, Rozen L, Hemker HC, Corazza F i wsp. Evaluation of the procoagulant activity in the plasma of cancer patients using a thrombin generation assay. Thromb Res. 2010; 126: 531-535.
  • Brotman DJ, Girod JP, Posch A, Jani JT, Patel JV, Gupta M i wsp. Effect of short – term glucocorticoids on hemostatic factor in healthy volunteers. Thromb Res. 2006; 118: 247-252.
  • Van Zaane B, Nur E, Squizzato A. Systemic review on the effect of glucocorticoid use on procoagulant, anticoagulant and fibrynolityc factors. J Thromb Haemost. 2010; 11: 2483-2493.
  • Łubińska M, Kazimierska E, Sworczak K. Hiperchomocysteinemia jako nowy czynnik ryzyka wielu chorób. Adv Clin Exp Med. 2006; 15: 897-903.
  • Kohler HP. Insulin resistance syndrome: interaction with coagulation and fibrinolysis. Swiss Med Wkly. 2002; 132: 241-252.
  • Langan SM, Hubbard R, Fleming K, West J. A population – based study of acute medical conditions associated with bullous pemphigoid. Br J Dermatol. 2009; 161: 1149-1152.
  • Langan SM, Smeeth L, Hubbard R, Fleming KM, Smith CJ, West J. Bullous pemphigoid and pemphigus vulgaris – incidence and mortality in tke UK: population based cohort study. BMJ. 2008; 337: 160-163.
  • Weibel ER. Point Counting Methods. w: Weibel ER. Stereological Methods. 1979; Academic Press, London, New York, Toronto, Sydney, San Francisco. vol. 1: 101-159.
  • D'Auria L, Cordiali Fei P, Pietravalle M, Ferraro C, Mastroianni A, Bonifati C i wsp. The serum levels of sE-selectin are increased in patients with bullous pemphigoid or pemphigus vulgaris. Correlation with the number of skin lesions and recovery after corticosteroid therapy. Br J Dermatol. 1997; 137: 59-64.
  • Dahlman-Ghozlan K, Heilborn JD, Stephansson E. Circulating levels of soluble E-selectin, ICAM-1 and VCAM-1 in bullous pemphigoid during low-dose methotrexate therapy. A prospective study. Exp Dermatol. 2000; 9: 336-340.
  • Smith AD, Streilein RD, Hall III RP. Neutrophil CD11b, L-selectin and Fc IgA receptors in patients with dermatitis herpetiformis. Br J Dermatol. 2002; 147: 1109-1117.
  • Springer TA. Adhesion receptors of the immune system. Nature 1990; 346: 425 434.
  • Zittermann SI, Issekutz AC. Basic fibroblast growth factor (bFGF, FGF-2) potentiates leukocyte recruitment to inflammation by enhancing endothelial adhesion molecule expression. Am J Pathol. 2006; 168: 835-846.
  • Watt FM. Role of integrins in regulating epidermal adhesion, growth and differentiation. EMBO J. 2002; 21: 3919-3926.
  • Kumar V, Jarząbek-Chorzelska M, Sulej J, Rajadhyaksha M, Jablońska S. Tissue transglutaminase and endomysial antibodies-diagnostic markers of gluten- sensitive enteropathy in dermatitis herpetiformis. Clin Immunol. 2001; 98: 378 382.
  • Petersen MJ, Woodley DT, Stricklin GP, O’Keefe EJ. Production of procollagenase by cultured human keratinocytes. J Biol Chem. 1987; 262: 835 840.
  • Airola K, Reunala T, Salo S, Saarialho-Kere UK. Urokinase plasminogen activator is expressed by basal keratinocytes before interstitial collagenase, stromelysin-1, and laminin-5 in experimentally induced dermatitis herpetiformis lesions. J Invest Dermatol. 1997; 108: 7-11.
  • Bhol KC, Goss L, Kumari S, Colon JE, Ahmed AR. Autoantibodies to human alpha6 integrin in patients with oral pemphigoid. J Dent Res. 2001; 80: 1711 1715.
  • Rashid KA, Stern JNH, Ahmed AR. Identification of an Epitope within Human Integrin a6 Subunit fo a Binding of Autiantibody and Its Role in Basement Membrane Separation in Oral Pemphigoid. J Immunol. 2006; 176: 1968-1977.
  • Kurpakus MA, Quaranta V, Jones JCR. Surface relocation of alpha6beta4 integrins and assembly of hemidesmosomes in an in vitro model of wound healing. J Cell Biol. 1991; 115: 1737-1750.
  • Merono A, Lucena C, López A, Garrido JJ, Perez de LL, Llanos D. Immunohistochemical analysis of beta 3 integrin (CD61): expression in pig tissues and human tumors. Histol Histopathol. 2002; 17: 347-352.
  • Liu KY, Timmons S, Lin YZ, Hawiger J. Identification of a functionally important sequence in the cytoplasmic tail of integrin beta3 by using cell permeable peptide analogs (signal transduction/cell adhesion/glycoprotein Ilb-IlIa/peptide delivery). Proc Natl Acad Sci U S A. 1996; 93: 11819-11824.
  • Sami N, Bhol KC, Ahmed AR: Treatment of oral pemphigoid with intravenous immunoglobulin as monotherapy. Long term follow-up: influence of treatment on antibody titres to human a6integrin. Clin Exp Immunol. 2002; 129: 533-540.
  • Ghosh S, Goldin E, Gordon FH, Malchow HA, Rask-Madsen J, Rutgeerts P i wsp. Natalizumab for active Crohn's disease. N Engl J Med. 2003; 348: 24-32.
  • Redhu NS, Saleh A, Shan L, Gerthoffer WT, Kung SK, Halayko AJ i wsp. Proinflammatory and Th2 cytokines regulate the high affinity IgE receptor (FcepsilonRI) and IgE-dependant activation of human airway smooth muscle cells. PLoS One. 2009;4: 6153.
  • Frezzolini A, Cianchini G, Ruffelli M, Cadoni S, Puddu P, De Pità O. Interleukin-16 expression and release in bullous pemphigoid. Clin Exp Immunol. 2004; 137: 595–600.
  • Nakashima H, Fujimoto M, Asashima N, Watanabe R, Kuwano Y, Yazawa N i wsp. Serum chemokine profile in patients with bullous pemphigoid.
  • Br J Dermatol. 2007;156: 454-459
  • Shrikhande M, Hunziker T, Braathen LR, Pichler WJ, Dahinden CA, Yawalkar N. Increased coexpression of eotaxin and interleukin 5 in bullous pemphigoid. Acta Derm Venereol. 2000; 80: 277-280.
  • Schmidt E, Ambach A, Bastian B, Bröcker EB, Zillikens D. Elevated levels of interleukin-8 in blister fluid of bullous pemphigoid compared with suction blisters of healthy control subjects. J Am Acad Dermatol. 1996; 34: 310-312.
  • Heimbach L, Li Z, Berkowitz P, Zhao M, Li N, Rubenstein DS i wsp. The C5a receptor on mast cells is critical for the autoimmune skin-blistering disease bullous pemphigoid. J Biol Chem. 2011; 286: 15003-15009.
  • Walker BA, Jacobson MA, Knight DA, Salvatore CA, Weir T, Zhou D i wsp. Adenosine A3 receptor expression and function in eosinophils. Am J Respir Cell Mol Biol. 1997;16: 531-537.
  • Chen R, Fairley JA, Zhao ML, Giudice GJ, Zillikens D, Diaz LA i wsp. Macrophages, but not T and B lymphocytes, are critical for subepidermal blister formation in experimental bullous pemphigoid: macrophage-mediated neutrophil infiltration depends on mast cell activation. J Immunol. 2002; 169: 3987-3992.
  • Chen R, Ning G, Zhao ML, Fleming MG, Diaz LA, Werb Z i wp. Mast cells play a key role in neutrophil recruitment in experimental bullous pemphigoid. J Clin Invest. 2001; 108: 1151-1158.
  • Rossi R, Johansson O. Cutaneous innervatio and the role of neuronal peptides in cutaneous inflammation: a minireview. Eur J Dermatol. 1998; 8: 299-306.
  • Amatya B, Nordlind K, Wahlgren CF. Responses to intradermal injections of substance P in psoriasis patients with pruritus. Skin Pharmacol Physiol. 2010; 23: 133-138.
  • Saraceno R, Kleyn CE, Terenghi G, Griffiths CE. The role of neuropeptides in psoriasis. Br J Dermatol. 2006; 154: 876-882.
  • Roosterman D, Goerge T, Schneider SW, Bunnett NW, Steinhoff M. Neuronal control of skin function: the skin as a neuroimmunoendocrine organ. Physiol Rev. 2006; 86: 1309-1379.
  • Misery L. Skin, immunity and the nervous system. Br J Dermatol. 1997; 37: 843 850.
  • O'Sullivan RL, Lipper G, Lerner EA. The neuro-immuno-cutaneous-endocrine network: relationship of mind and skin. Arch Dermatol. 1998; 134: 1431-1435.
  • Zhang B, Alysandratos KD, Angelidou A, Asadi S, Sismanopoulos N, Delivanis DA i wsp. Human mast cell degranulation and preformed TNF secretion require mitochondrial translocation to exocytosis sites: relevance to atopic dermatitis. J Allergy Clin Immunol. 2011;127: 1522-1531.
  • Salomon J, Baran E. The role of selected neuropeptides in pathogenesis of atopic dermatitis. JEADV 2008; 22: 223-228.
  • Bianchi B, Matucci R, Danesi A, Rossi R, Ipponi P, Giannotti B i wsp. Characterization of [3H] substance P binding sites in human skin. J Eur Acad Dermatol Venerol. 1999; 12: 6-10.
  • Amara SG, Jonas V, Rosenfeld MG, Ong ES, Evans RM. Alternative RNA processing in calcitonin gene expression generates mRNAs encoding different polypeptide products. Nature. 1982; 298: 240-244.
  • Slominski A, Ermak G, Mazurkiewicz JE, Baker J, Wortsman J. Characterization of corticotropin-releasing hormone (CRH) in human skin. J Clin Endocrinol Metab. 1998; 83: 1020-1024.
  • Slominski AT, Zmijewski MA, Zbytek B, Brozyna AA, Granese J, Pisarchik A i wsp. Regulated proenkephalin expression in human skin and cultured skin cells. J Invest Dermatol. 2011; 131: 613-622.
  • Linton EA, Woodman JR, Asboth G, Glynn BP, Plested CP, Bernal AL.Corticotrophin releasing hormone: its potential for a role in human myometrium. Exp Physiol. 2001; 86: 273-281.
  • Zbytek B, Slominski AT. Corticotropin-releasing hormone induces keratinocyte differentiation in the adult human epidermis. J Cell Physiol. 2005; 203: 118–126.
  • Quevedo ME, Slominski A, Pinto W, Wei E, Wortsman J. Pleiotropic effects of corticotropin releasing hormone on normal human skin keratinocytes. In Vitro Cell Dev Biol Anim. 2001; 37: 50–54.
  • McQueen DS, Noble MAH, Bond SM. Endothelin-1 activates ETA receptors to cause reflex scratching in BALB/c mice. Br J Pharmacol. 2007; 151: 278–284.
  • Lahav R, Suvà ML, Rimoldi D, Patterson PH, Stamenkovic I. Endothelin Receptor B Inhibition Triggers Apoptosis and Enhances Angiogenesis in Melanomas. Cancer Res. 2004; 64: 8945-8953.
  • Spinella F, Rosanò L, Di Castro V, Decandia S, Nicotra MR, Natali PG i wsp. Endothelin-1 and endothelin-3 promote invasive behavior via hypoxia-inducible factor-1a in human melanoma cells. Cancer Res 2007;67: 1725-1734.
  • Oikarinen A, Kylmäniemi M, Autio-Harmainen H, Autio P, Salo T. Demonstration of 72-kD and 92-kD forms of type Ivcollagenase in human skin: variable expression in various blistering diseases, inducting during re-epithelialization and decrease by topical glucocorticoids. J Invest Dermatol. 1993; 101: 205-210.
  • Saarialho-Kere UK, Vaalamo M, Airola K, Niemi KM, Oikarinen AI, Parks WC. Interstitial collagenase is expressed by keratinocytes that are actively involved in reepithelialization in blistering skin disease. J Invest Dermatol. 1995; 104: 982 988.
  • Welgus HG, Bauer EA, Stricklin GP. Elevated levels of human collagenase inhibitor in blister fluids of diverse etiology. J Invest Dermatol. 1986; 87: 592 596.
  • Verraes S, Hornebeck W, Polette M, Borradori L, Bernard P. Respective contribution of neutrophil elastase and matrix metalloproteinase 9 in the degradation of BP180 (type XVII collagen) in human bullous pemphigoid. J Invest Dermatol. 2001; 117: 1091-1096.
  • Ståhle-BäckDhl M, Inoue M, Guidice GJ, Parks WC. 92-kD gelatinase is produced by eosinophils at the site of blister formation in bullous pemphigoid and cleaves the extracellular domain of recombinant 180-kD bullous pemphigoid autoantigen. J Clin Invest. 1994; 93: 2022-2030.
  • Schmidt E, Reimer S, Kruse N, Bröcker EB, Zillikens D. The IL-8 release from cultured human keratinocytes,mediated by antibodies to bullous pemphigoid antigen 180, is inhibited by dapsone. Clin Exp Immunol. 2001; 124: 157-162.
  • Grando SA, Glukhenky BT, Drannik GN, Epshtein EV, Kostromin AP, Korostash TA. Mediators of inflammation in blister fluids from patients with pemphigus vulgaris and bullous pemphigoid. Arch Dermatol. 1989; 125: 925 930.
  • Beaudeux JL, Giral P, Bruckert E, Foglietti MJ, Chapman MJ. Matrix metalloproteinases and atherosclerosis. Therapeutic aspects. Ann Biol Clin. 2003; 61: 147-158.
  • Nip LH, Uitto VJ, Golub LM. Inhibition of epithelial cell matrix metalloproteinases by tetracyclines. J Periodontal Res. 1993; 28: 379-385.
  • Böhm BB, Aigner T, Roy B, Brodie TA, Blobel CP, Burkhardt H. Homeostatic effects of the metalloproteinase disintegrin ADAM15 in degenerative cartilage remodeling. Arthritis Rheum. 2005; 52: 1100-1109.
  • Böhm BB, Aigner T, Gehrsitz A, Blobel CP, Kalden JR, Burkhardt H. Up regulation of MDC15 (metargidin) messenger RNA in human osteoarthritic cartilage. Arthritis Rheum. 1999;42: 1946-1950.
  • Cugno M, Tedeschi A, Asero R, Meroni PL, Marzano AV. Skin autoimmunity and blood coagulation. Autoimmunity. 2010;43: 189-194.
  • Wojtukiewicz MZ, Rucińska M. Aktywacja krzepnięcia krwi u chorych na nowotwory: implikacje kliniczne. Nowotwory. 1999; 49: 381-391.
  • Wojtukiewicz MZ, Rybałtowski M. Powikłania zakrzepowo-zatorowe w chorobie nowotworowej. Leczenie Wspomagające. 2003; 4: 3-10.
  • Kucharewicz I, Kowal K, Buczko W, Bodzenia-Lukaszyk A. The plasmin system in airway remodeling. Thromb Res. 2003, 112: 1-7.
  • Kowal K, Kemona-Chetnik I, Kucharewicz I, Bodzenia-Luszczyk A. The involvment of coagulation in inflammatory lung diseases. Pol Arch Med Wew. 2003; 110: 775-781.
  • Cunningham MA, Romas P, Hutchinson P, Holdsworth SR, Tipping PG. Tissue factor and factor VIIa receptor/ligand interactions induce proinflammatory effects in macrophages. Blood. 1999; 15; 94: 3413-3420.
  • Del Prete G, De Carli M, Lammel RM, D'Elios MM, Daniel KC, Giusti B i wsp. Th1 and Th2 T-helper cells exert opposite regulatory effects on procoagulant activity and tissue factor production by human monocytes. Blood. 1995; 86: 250 257.
  • Asero R, Tedeschi A, Riboldi P, Cugno M. Plasma of patients with chronic urticaria shows signs of thrombin generation, and its intradermal injection causes wheal-and-flare reactions much more frequently than autologous serum. J Allergy Clin Immunol. 2006;117: 1113-1117.
  • So AK, Varisco PA, Kemkes-Matthes B, Herkenne-Morard C, Chobaz-Péclat V, Gerster JC i wsp. Arthritis is linked to local and systemic activation of coagulation and fibrinolysis pathways. J Thromb Haemost. 2003;1: 2510-2515.
  • Kume K, Yamasaki M, Tashiro M, Yoshikawa I, Otsuki M. Activations of coagulation and fibrinolysis secondary to bowel inflammation in patients with ulcerative colitis. Intern Med. 2007; 46: 1323-1329.
  • Liaw PC, Esmon CT, Kahnamoui K, Schmidt S, Kahnamoui S, Ferrell G i wsp. Patients with severe sepsis vary markedly in their ability to generate activated protein C. Blood. 2004;104: 3958-3964.
  • Levi M, van der Poll T, Büller HR. Bidirectional relation between inflammation and coagulation. Circulation. 2004; 109: 2698-2704.
  • Ludwicka-Bradley A, Tourkina E, Suzuki S, Tyson E, Bonner M, Fenton JW 2nd i wsp. Thrombin upregulates interleukin-8 in lung fibroblasts via cleavage of proteolytically activated receptor-I and protein kinase C-gamma activation. Am J Respir Cell Mol Biol. 2000; 22: 235-243.
  • Marietta E, Black K, Camilleri M. Krause P, Rogers RS, David C. i wsp.
  • A new model for dermatitis herpetiformis that uses HLA DQ8 transgenic NOD mice. J Clin Invest. 2004; 114: 1090-1097.
  • Rzany B, Partscht K, Jung M, Kippes W, Mecking D, Baima B i wsp. Risk factors for lethal outcome in patients with bullous pemphigoid: low serum albumin level, high dosage of glucocorticosteroids and old age. Arch Dermatol. 2002; 138: 903-908.
  • Beissert S, Werfel T, Frieling U, Böhm M, Sticherling M, Stadler R i wsp. A comparison of oral methylprednisolone plus azathioprine or mycophenolate mofetil for the treatment of bullous pemphigoid. Arch Dermatol. 2007; 143: 1536-1542.
  • Mignogna MD, Leuci S, Piscopo R, Bonovolontà G. Intravenous immune-globulins and mucous membrane pemphigoid. Ophthalmology. 2008; 115: 752 752.
  • Schmidt E, Bröcker EB, Goebeler M. Rituximab in treatment-resistant autoimmune blistering skin disorders. Clin Rev Allergy Immunol. 2008; 34: 56 64.
  • Joly P, Roujeau JC, Benichou J, Picard C, Dreno B, Delaporte E i wsp. A comparison of oral and topical corticosteroids in patients with bullous pemphigoid. N Engl J Med. 2002; 346: 321-327.
  • Ernst E. Plasma fibrynogen - an independent cardivascular risk factor. J. Intern. Med. 1990; 227: 365-372.
  • Senior RM, Skogen WF, Griffin GL, Wilner GD. Effects of fibrinogen derivatives upon the inflammatory response. Studies with human fibrinopeptide B. J. Clin. Invest. 1986; 77: 1014-1019.
  • Fairley JA, Baum CL, Brandt DS, Messingham KA. Pathogenicity of IgE in autoimmunity: successful treatment of bullous pemphigoid with omalizumab. J Allergy Clin Immunol. 2009; 123: 704-705.
  • Monnier-Murina K, Du Thanh A, Merlet-Albran S, Guillot B, Dereure O. Bullous pemphigoid occurring during efalizumab treatment for psoriasis: a paradoxical auto-immune reaction? Dermatology. 2009; 219: 89-90.
  • Joly P, Courville P, Lok C, Bernard P, Saiag P, Dreno B i wsp. Clinical criteria for the diagnosis of bullous pemphigoid: a reevaluation according to immunoblot analysis of patient sera. Dermatology. 2004; 208: 16-20.
  • Braun-Falco O, Plewig G, Wolff HH, Burgdorf WH. Choroby pęcherzowe.
  • W: Dermatologia. Braun-Falco O, Plewig G, Wolff HH, Burgdorf WH (red.). Czelej, Lublin 2002; 639-643.
  • Hall TC. Paraneoplastic syndromes: mechanisms. Semin Oncol. 1997; 24: 269 276.
  • Demitsu T, Kakurai M, Yoneda K, Iida E, Takada M, Hiratsuka Y i wsp. Localized pemphigoid (Brunsting-Perry type) with IgG antibody to BP180 NC16a domain resembling lupus erythematosus successfully treated with topical tacrolimus therapy. J Eur Acad Dermatol Venereol. 2009; 923: 79-80.
  • Kjellman P, Eriksson H, Berg P. A retrospective analysis of patients with bullous pemphigoid treated with methotrexate. Arch Dermatol. 2008; 144: 612-616.
  • Carrozzo M, Arduino PG, Baldovino S, Di Zenzo G, Salzano S, Calabresi V i wsp. Minocycline in combination with mycophenolate mofetil in oral mucous membrane pemphigoid. Eur J Dermatol. 2008; 18: 198-200.
  • Błaszczyk M. Leczenie pemfigoidu pęcherzowego. Czy miejscowa terapia mocnymi kortykosteroidami stanowi rozwiązanie problemu? Przegl Dermatol. 2005; 4: 271-277.
  • Schultz HY, Diaz LA, Sirois DA, Werth VP, Grando SA. Generating consensus research goals and treatment strategies for pemphigus and pemphigoid: The 2010 JC Bystryn Pemphigus and Pemphigoid Meeting. J Invest Dermatol. 2011; 131: 1395-1399.
  • Roujeau JC, Lok C, Bastuji-Garin S, Mhalla S, Enginger V, Bernard P. High risk of death in elderly patients with extensive bullous pemphigoid. Arch Dermatol. 1998; 134: 465-469.
  • Joly P, Benichou J, Lok C, Hellot MF, Saiag P, Tancrede-Bohin E i wsp. Prediction of survival for patients with bullous pemphigoid: a prospective study. Arch Dermatol. 2005; 141: 691-698.
  • Stockman A, Beele H, Vanderhaeghen Y, Naeyaert JM. Topical class I corticosteroids in 10 patients with bullous pemphigoid: correlation of the autcome with the severity degree of the disease and review of the literature. J Eur Acad Dermatol Venerol. 2004; 18: 164-168.
  • Jung M, Kippes W, Messer G, Zillikens D, Rzany B. Increased risk of bullous pemphigoid in male and very old patients: a population-based study on incidence. J Am Acad Dermatol. 1999; 41: 266-268.

Document Type

paper

Publication order reference

Identifiers

YADDA identifier

bwmeta1.element.psjd-c7ea8636-d515-433e-a3ff-b078af460c1c
JavaScript is turned off in your web browser. Turn it on to take full advantage of this site, then refresh the page.