Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Allergy Clin Immunol ; 149(2): 671-684.e9, 2022 02.
Article in English | MEDLINE | ID: mdl-34186142

ABSTRACT

BACKGROUND: Administering allergens in increasing doses can temporarily suppress IgE-mediated allergy and anaphylaxis by desensitizing mast cells and basophils; however, allergen administration during desensitization therapy can itself induce allergic responses. Several small molecule drugs and nutraceuticals have been used clinically and experimentally to suppress these allergic responses. OBJECTIVES: This study sought to optimize drug inhibition of IgE-mediated anaphylaxis. METHODS: Several agents were tested individually and in combination for ability to suppress IgE-mediated anaphylaxis in conventional mice, FcεRIα-humanized mice, and reconstituted immunodeficient mice that have human mast cells and basophils. Hypothermia was the readout for anaphylaxis; therapeutic efficacy was measured by degree of inhibition of hypothermia. Serum mouse mast cell protease 1 level was used to measure extent of mast cell degranulation. RESULTS: Histamine receptor 1 (HR1) antagonists, ß-adrenergic agonists, and a spleen tyrosine kinase (Syk) inhibitor were best at individually inhibiting IgE-mediated anaphylaxis. A Bruton's tyrosine kinase (BTK) inhibitor, administered alone, only inhibited hypothermia when FcεRI signaling was suboptimal. Combinations of these agents could completely or nearly completely inhibit IgE-mediated hypothermia in these models. Both Syk and BTK inhibition decreased mast cell degranulation, but only Syk inhibition also blocked desensitization. Many other agents that are used clinically and experimentally had little or no beneficial effect. CONCLUSIONS: Combinations of an HR1 antagonist, a ß-adrenergic agonist, and a Syk or a BTK inhibitor protect best against IgE-mediated anaphylaxis, while an HR1 antagonist plus a ß-adrenergic agonist ± a BTK antagonist is optimal for inhibiting IgE-mediated anaphylaxis without suppressing desensitization.


Subject(s)
Anaphylaxis/prevention & control , Immunoglobulin E/immunology , Adrenergic beta-Agonists/therapeutic use , Animals , Drug Therapy, Combination , Histamine Antagonists/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors
2.
Front Med (Lausanne) ; 8: 758311, 2021.
Article in English | MEDLINE | ID: mdl-34805224

ABSTRACT

The endoplasmic reticulum (ER) is not only responsible for protein synthesis and folding but also plays a critical role in sensing cellular stress and maintaining cellular homeostasis. Upon sensing the accumulation of unfolded proteins due to perturbation in protein synthesis or folding, specific intracellular signaling pathways are activated, which are collectively termed as unfolded protein response (UPR). UPR expands the capacity of the protein folding machinery, decreases protein synthesis and enhances ER-associated protein degradation (ERAD) which degrades misfolded proteins through the proteasomes. More recent evidences suggest that UPR also amplifies cytokines-mediated inflammatory responses leading to pathogenesis of inflammatory diseases. UPR signaling also activates autophagy; a lysosome-dependent degradative pathwaythat has an extended capacity to degrade misfolded proteins and damaged ER. Thus, activation of autophagy limits inflammatory response and provides cyto-protection by attenuating ER-stress. Here we review the mechanisms that couple UPR, autophagy and cytokine-induced inflammation that can facilitate the development of novel therapeutic strategies to mitigate cellular stress and inflammation associated with various pathologies.

3.
Front Immunol ; 9: 249, 2018.
Article in English | MEDLINE | ID: mdl-29497423

ABSTRACT

Because of the morbidity and limited therapeutic options of autoimmune diseases, there is a high, and thus far, unmet medical need for development of novel treatments. Pemphigoid diseases, such as epidermolysis bullosa acquisita (EBA), are prototypical autoimmune diseases that are caused by autoantibodies targeting structural proteins of the skin, leading to inflammation, mediated by myeloid cells. To identify novel treatment targets, we performed cutaneous genome-wide mRNA expression profiling in 190 outbred mice after EBA induction. Comparison of genome-wide mRNA expression profiles in diseased and healthy mice, and construction of a co-expression network identified Sykb (spleen tyrosine kinase, SYK) as a major hub gene. Aligned, pharmacological SYK inhibition protected mice from experimental EBA. Using lineage-specific SYK-deficient mice, we identified SYK expression on myeloid cells to be required to induce EBA. Within the predicted co-expression network, interactions of Sykb with several partners (e.g., Tlr13, Jdp2, and Nfkbid) were validated by curated databases. Additionally, novel gene interaction partners of SYK were experimentally validated. Collectively, our results identify SYK expression in myeloid cells as a requirement to promote inflammation in autoantibody-driven pathologies. This should encourage exploitation of SYK and SYK-regulated genes as potential therapeutic targets for EBA and potentially other autoantibody-mediated diseases.


Subject(s)
Autoimmune Diseases/immunology , Epidermolysis Bullosa Acquisita/immunology , Myeloid Cells/immunology , Skin/metabolism , Syk Kinase/immunology , Aged, 80 and over , Animals , Biopsy , Case-Control Studies , Disease Models, Animal , Epidermolysis Bullosa Acquisita/pathology , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Protein Interaction Maps/immunology , Skin/cytology , Skin/immunology , Skin/pathology , Syk Kinase/genetics , Whole Genome Sequencing
4.
Mol Med ; 22: 918-926, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27999842

ABSTRACT

Epidermolysis bullosa acquisita (EBA) is a difficult-to-treat subepidermal autoimmune blistering skin disease (AIBD) with circulating and tissue-bound anti-type VII collagen antibodies. Different reports have indicated an increased concentration of tumor necrosis factor alpha (TNF) in the serum and blister fluid of patients with subepidermal AIBDs. Furthermore, successful anti-TNF treatment has been reported for individual patients with AIBDs. Here, we show that in mice, induction of experimental EBA by repeated injections of rabbit-anti mouse type VII collagen antibodies led to increased expression of TNF in skin, as determined by real-time PCR and immunohistochemistry. To investigate if the increased TNF expression is of functional relevance in experimental EBA, we inhibited TNF function using the soluble TNF receptor fusion protein etanercept (Enbrel®) or a monoclonal antibody to murine TNF. Interestingly, mice receiving either of these two treatments showed significantly milder disease progression than controls. In addition, immunohistochemical staining demonstrated reduced numbers of macrophages in lesional skin in mice treated with TNF inhibitors compared to controls. Furthermore, etanercept treatment significantly reduced the disease progression in immunization-induced EBA. In conclusion, the increased expression of TNF in experimental EBA is of functional relevance, as both the prophylactic blockade of TNF and the therapeutic use of etanercept impaired the induction and progression of experimental EBA. Thus, TNF is likely to serve as a new therapeutic target for EBA and AIBDs with a similar pathogenesis.

5.
J Immunol ; 195(5): 1945-54, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26202985

ABSTRACT

Animal models have enhanced our understanding of the pathogenesis of autoimmune diseases. For these models, genetically identical, inbred mice have commonly been used. Different inbred mouse strains, however, show a high variability in disease manifestation. Identifying the factors that influence this disease variability could provide unrecognized insights into pathogenesis. We established a novel Ab transfer-induced model of epidermolysis bullosa acquisita (EBA), an autoimmune disease characterized by (muco)-cutaneous blistering caused by anti-type VII collagen (COL7) autoantibodies. Blistering after anti-COL7 IgG (directed against the von Willebrand factor A-like domain 2) transfer showed clear variability among inbred mouse strains, that is, severe cutaneous blistering and inflammation in C57BL/6J and absence of skin lesions in MRL/MpJ mice. The transfer of anti-COL7 IgG into irradiated, EBA-resistant MRL/MpJ mice, rescued by transplantation with bone marrow from EBA-susceptible B6.AK-H2k mice, induced blistering. To the contrary, irradiated EBA-susceptible B6.AK-H2k mice that were rescued using MRL/MpJ bone marrow were devoid of blistering. In vitro, immune complex activation of neutrophils from C57BL/6J or MRL/MpJ mice showed an impaired reactive oxygen species release from the latter, whereas no differences were observed after PMA activation. This finding was paralleled by divergent expression profiles of immune complex-activated neutrophils from either C57BL/6J or MRL/MpJ mice. Collectively, we demonstrate that radiosensitive cells determine the varying extent of skin inflammation and blistering in the end-stage effector phase of EBA.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow Transplantation/methods , Dermatitis/immunology , Epidermolysis Bullosa Acquisita/immunology , Animals , Autoantibodies/immunology , Blister/immunology , Bone Marrow Cells/radiation effects , Collagen Type VII/immunology , Disease Models, Animal , Gene Regulatory Networks/immunology , Humans , Immunoglobulin G/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred MRL lpr , Mice, Knockout , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Neutrophils/immunology , Neutrophils/metabolism , Oligonucleotide Array Sequence Analysis , Rabbits , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Species Specificity , Transcriptome/immunology
6.
J Pathol ; 237(1): 111-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25953430

ABSTRACT

Genetic studies have added to the understanding of complex diseases. Here, we used a combined genetic approach for risk-loci identification in a prototypic, organ-specific, autoimmune disease, namely experimental epidermolysis bullosa acquisita (EBA), in which autoantibodies to type VII collagen (COL7) and neutrophil activation cause mucocutaneous blisters. Anti-COL7 IgG induced moderate blistering in most inbred mouse strains, while some showed severe disease or were completely protected. Using publicly available genotyping data, we identified haplotype blocks that control blistering and confirmed two haplotype blocks in outbred mice. To identify the blistering-associated genes, haplotype blocks encoding genes that are differentially expressed in EBA-affected skin were considered. This procedure identified nine genes, including retinoid-related orphan receptor alpha (RORα), known to be involved in neurological development and function. After anti-COL7 IgG injection, RORα+/- mice showed reduced blistering and homozygous mice were completely resistant to EBA induction. Furthermore, pharmacological RORα inhibition dose-dependently blocked reactive oxygen species (ROS) release from activated neutrophils but did not affect migration or phagocytosis. Thus, forward genomics combined with multiple validation steps identifies RORα to be essential to drive inflammation in experimental EBA.


Subject(s)
Epidermolysis Bullosa Acquisita/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Skin/metabolism , Animals , Autoantibodies/immunology , Collagen Type VII/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Partial Agonism , Epidermolysis Bullosa Acquisita/genetics , Epidermolysis Bullosa Acquisita/immunology , Epidermolysis Bullosa Acquisita/pathology , Genetic Predisposition to Disease , Genomics/methods , Haplotypes , Heterozygote , Homozygote , Immunoglobulin G/immunology , Mice, Knockout , Neutrophil Activation , Neutrophils/immunology , Neutrophils/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/immunology , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction , Skin/drug effects , Skin/immunology , Skin/pathology , Species Specificity , Sulfonamides/pharmacology , Thiophenes/pharmacology , Time Factors
7.
J Immunol ; 194(8): 3656-63, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25795756

ABSTRACT

Although reports documented aberrant cytokine expression in autoimmune bullous dermatoses (AIBDs), cytokine-targeting therapies have not been established in these disorders. We showed previously that IL-6 treatment protected against tissue destruction in experimental epidermolysis bullosa acquisita (EBA), an AIBD caused by autoantibodies to type VII collagen (COL7). The anti-inflammatory effects of IL-6 were mediated by induction of IL-1ra, and prophylactic IL-1ra administration prevented blistering. In this article, we demonstrate elevated serum concentrations of IL-1ß in both mice with experimental EBA induced by injection of anti-COL7 IgG and in EBA patients. Increased IL-1α and IL-1ß expression also was observed in the skin of anti-COL7 IgG-injected wild-type mice compared with the significantly less diseased IL-1R-deficient or wild-type mice treated with the IL-1R antagonist anakinra or anti-IL-1ß. These findings suggested that IL-1 contributed to recruitment of inflammatory cells into the skin. Accordingly, the expression of ICAM-1 was decreased in IL-1R-deficient and anakinra-treated mice injected with anti-COL7. This effect appeared to be specifically attributable to IL-1 because anakinra blocked the upregulation of different endothelial adhesion molecules on IL-1-stimulated, but not on TNF-α-stimulated, cultured endothelial cells. Interestingly, injection of caspase-1/11-deficient mice with anti-COL7 IgG led to the same extent of skin lesions as in wild-type mice. Collectively, our data suggest that IL-1, independently of caspase-1, contributes to the pathogenesis of EBA. Because anti-IL-1ß in a prophylactic setting and anakinra in a quasi-therapeutic setting (i.e., when skin lesions had already developed) improved experimental EBA, IL-1 appears to be a potential therapeutic target for EBA and related AIBDs.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Blister/immunology , Caspase 1/immunology , Epidermolysis Bullosa Acquisita/immunology , Immunoglobulin G/immunology , Intercellular Adhesion Molecule-1/immunology , Interleukin-1beta/immunology , Animals , Autoantibodies/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Blister/genetics , Caspase 1/genetics , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Collagen Type VII/genetics , Collagen Type VII/immunology , Epidermolysis Bullosa Acquisita/genetics , Immunoglobulin G/genetics , Intercellular Adhesion Molecule-1/genetics , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/genetics , Mice , Mice, Knockout , Skin/immunology , Skin/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/drug effects , Up-Regulation/genetics , Up-Regulation/immunology
8.
Exp Dermatol ; 24(6): 471-3, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25739426

ABSTRACT

A broad range of immunosuppressive and immunomodulatory effects of heat shock protein 90 (Hsp90) blockade has been described in models of autoimmune bullous diseases, but the direct contribution of this chaperone to neutrophil effector pathways in the context of autoantibody-driven blistering is generally unknown. Therefore, this has been addressed in the current study on the basis of the subepidermal blistering disease epidermolysis bullosa acquisita (EBA) characterized by autoantibodies against type VII collagen, in which a crucial role of neutrophils and both their reactive oxygen species and matrix metalloproteinases in mediating tissue injury has been established. First, the Hsp90 antagonist 17-DMAG dose-dependently inhibited dermal-epidermal separation ex vivo in cryosections of human skin induced by co-incubation of EBA patient autoantibodies with neutrophils from healthy blood donors. Next, 17-DMAG dose-dependently suppressed production and release of reactive oxygen species by human neutrophils induced by both fMLP ± LPS and EBA-specific immune complexes. In addition, co-immunoprecipitation studies revealed that extracellular Hsp90 interacted with secreted matrix metalloproteinases 2 and 12 in sera of EBA patients, suggesting that these basement membrane-degrading proteolytic enzymes are client proteins of Hsp90 and dependent on its chaperone function. Our findings add to the knowledge of the multimodal anti-inflammatory effects of Hsp90 blockade and implicate that Hsp90 is closely involved in the effector mechanisms of neutrophil-driven autoantibody-induced tissue damage, thus being a relevant therapeutic target in patients with neutrophil-mediated autoimmune diseases such as inflammatory types of EBA.


Subject(s)
Autoantibodies/physiology , Epidermolysis Bullosa Acquisita/pathology , Epidermolysis Bullosa Acquisita/physiopathology , HSP90 Heat-Shock Proteins/physiology , Neutrophils/pathology , Skin/pathology , Autoantibodies/immunology , Benzoquinones/pharmacology , Case-Control Studies , Cells, Cultured , Collagen Type VII/immunology , Dose-Response Relationship, Drug , Epidermolysis Bullosa Acquisita/immunology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/drug effects , Humans , Immunoprecipitation , Lactams, Macrocyclic/pharmacology , Matrix Metalloproteinase 12/physiology , Matrix Metalloproteinase 2/physiology , Neutrophils/drug effects , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Skin/drug effects , Skin/physiopathology
9.
J Invest Dermatol ; 135(6): 1565-1573, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25689103

ABSTRACT

Epidermolysis bullosa acquisita (EBA) is an autoimmune blistering disease of the skin and mucous membranes, characterized by autoantibodies against type VII collagen (COL7), a major component of anchoring fibrils. Different clinical EBA phenotypes are described, including mechanobullous and inflammatory variants. Most EBA patients' sera react with epitopes located within the non-collagenous 1 (NC1) domain of human COL7. However, it has remained unclear whether antibody binding to these different epitopes is pathogenically relevant. To address this issue, we generated recombinant proteins covering the entire NC1 domain. IgG reactivity with these proteins was analyzed in sera of 69 EBA patients. Most recognized clusters of epitopes throughout the NC1 domain. No correlation was detected between antibody specificity and clinical phenotype. To study the pathogenicity of antibodies specific to different NC1 subdomains, rabbit antibodies were generated. All these antibodies caused dermal-epidermal separation ex vivo. Antibodies against two of these subdomains were injected into mice carrying null mutations of mouse COL7 and the human COL7 transgene and induced subepidermal blisters. We here document that autoantibodies to COL7, independent of the targeted epitopes, induce blisters both ex vivo and in vivo. In addition, using COL7-humanized mice, we provide in vivo evidence of pathogenicity of autoantibodies binding to human COL7.


Subject(s)
Autoantibodies/chemistry , Blister/metabolism , Collagen Type VII/chemistry , Epidermolysis Bullosa Acquisita/metabolism , Epitopes/chemistry , Animals , Antibodies/chemistry , Autoantibodies/immunology , Dermis/metabolism , Epidermis/metabolism , Female , Humans , Immunoglobulin G/chemistry , Inflammation , Male , Mice , Mice, Transgenic , Mutation , Protein Structure, Tertiary , Rabbits , Recombinant Proteins/chemistry , Transgenes
10.
J Immunol ; 192(2): 559-71, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24337383

ABSTRACT

GM-CSF activates hematopoietic cells and recruits neutrophils and macrophages to sites of inflammation. Inhibition of GM-CSF attenuates disease activity in models of chronic inflammatory disease. Effects of GM-CSF blockade were linked to modulation of the effector phase, whereas effects on early pathogenic events, for example, Ab production, have not been identified. To evaluate yet uncharacterized effects of GM-CSF on early pathogenic events in chronic inflammation, we employed immunization-induced epidermolysis bullosa acquisita (EBA), an autoimmune bullous disease caused by autoantibodies to type VII collagen. Compared to wild-type mice, upon immunization, GM-CSF(-/-) mice produced lower serum autoantibody titers, which were associated with reduced neutrophil numbers in draining lymph nodes. The same effect was observed in neutrophil-depleted wild-type mice. Neutrophil depletion in GM-CSF(-/-) mice led to a stronger inhibition, indicating that GM-CSF and neutrophils have additive functions. To characterize the contribution of GM-CSF specifically in the effector phase of EBA, disease was induced by transfer of anti-type VII collagen IgG into mice. We observed an increased GM-CSF expression, and GM-CSF blockade reduced skin blistering. Additionally, GM-CSF enhanced reactive oxygen species release and neutrophil migration in vitro. In immunization-induced murine EBA, treatment with anti-GM-CSF had a beneficial effect on established disease. We demonstrate that GM-CSF modulates both autoantibody production and skin blistering in a prototypical organ-specific autoimmune disease.


Subject(s)
Autoantibodies/immunology , Epidermolysis Bullosa Acquisita/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Animals , Autoimmune Diseases/immunology , Collagen Type VII/immunology , Disease Models, Animal , Humans , Immunoglobulin G/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/immunology , Reactive Oxygen Species/immunology , Skin/immunology
11.
J Invest Dermatol ; 133(10): 2390-2399, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23448878

ABSTRACT

Corticosteroids are regularly used to treat autoimmune diseases, such as bullous pemphigoid (BP). In BP, autoantibodies bind to type XVII collagen (COL17), located at the dermal-epidermal junction. A crucial role of neutrophils in experimental BP has been established. Specifically, reactive oxygen species and proteolytic granule enzymes mediate tissue injury. Therefore, we investigated the effects of methylprednisolone (MP) on neutrophils, which are likely to be affected by topical treatment. First, MP inhibited dermal-epidermal separation ex vivo in cryosections of the human skin induced by co-incubation of BP autoantibodies with neutrophils from healthy volunteers. Next, MP inhibited neutrophil activation in vitro induced by immune complexes (ICs) of COL17 and autoantibodies. This neutrophil activation was associated with phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), p38 mitogen-activated protein kinase (MAPK), and Akt. In turn, inhibition of ERK1/2, p38 MAPK, or Akt phosphorylation inhibited neutrophil activation by IC in vitro and dermal-epidermal separation ex vivo. In addition, we observed an increase of p38 MAPK phosphorylation in dermal infiltrates of BP patients. Treatment of mice with either MP or inhibitors of p38-MAPK or ERK1/2 phosphorylation impaired induction of autoantibody- or irritant-induced neutrophil-dependent inflammation. We here identify the inhibition of Akt, ERK1/2, and p38 MAPK phosphorylation as molecular mechanisms to promote MP's therapeutic effects.


Subject(s)
Autoantibodies/immunology , Epidermolysis Bullosa Acquisita/drug therapy , Methylprednisolone/pharmacology , Neutrophils/drug effects , Pemphigoid, Bullous/drug therapy , Adult , Animals , Antigen-Antibody Complex/immunology , Butadienes/pharmacology , Dermis/cytology , Dermis/immunology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Epidermal Cells , Epidermis/immunology , Epidermolysis Bullosa Acquisita/immunology , Epidermolysis Bullosa Acquisita/pathology , Glucocorticoids/pharmacology , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred C57BL , Neutrophils/cytology , Neutrophils/immunology , Nitriles/pharmacology , Pemphigoid, Bullous/immunology , Pemphigoid, Bullous/pathology , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , Respiratory Burst/drug effects , Respiratory Burst/immunology
12.
J Autoimmun ; 40: 74-85, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22980031

ABSTRACT

Cytokines are key regulators of physiological inflammatory responses, while aberrant cytokine expression contributes to pathogenesis of autoimmune diseases. We noted increased IL-6 levels in human and murine epidermolysis bullosa acquisita (EBA), a prototypic organ-specific autoimmune bullous dermatoses (AIBD) induced by autoantibodies to type VII collagen (COL7). In contrast to rheumatoid arthritis, blockade of IL-6 led to strikingly enhanced experimental EBA, while treatment with recombinant IL-6 was protective. This was due to classical IL-6 signalling and independent of IL-6 trans-signalling, as treatment of mice with sgp130Fc had no impact on EBA manifestation. Induction of EBA in mice led to increased IL-1ra levels in skin and serum, while blockade of IL-6 completely inhibited IL-1ra expression induced by autoantibodies to COL7. In line, treatment of mice with EBA with recombinant IL-6 induced IL-1ra concentrations exceeding those of untreated animals with EBA, and IL-1ra (anakinra) administration significantly impaired experimental EBA induction. We here identified a novel anti-inflammatory pathway in an organ-specific autoimmune disease. Modulation of this IL-1ra pathway by classical IL-6 signalling demonstrates anti-inflammatory and protective activities of IL-6 in vivo.


Subject(s)
Autoimmune Diseases/prevention & control , Epidermolysis Bullosa Acquisita/prevention & control , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-6/pharmacology , Animals , Apoptosis , Autoantibodies/immunology , Autoimmune Diseases/blood , Collagen Type VII/immunology , Cytokines/blood , Disease Models, Animal , Epidermolysis Bullosa Acquisita/blood , Epidermolysis Bullosa Acquisita/immunology , Humans , Inflammation , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction/immunology , Skin/metabolism , Tissue Inhibitor of Metalloproteinase-1/immunology
13.
J Pathol ; 228(1): 8-19, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22430937

ABSTRACT

Autoantibody-mediated diseases are clinically heterogeneous and often fail conventional therapeutic strategies. Gene expression profiling has helped to identify new molecular pathways in these diseases, although their potential as treatment targets largely remains to be functionally validated. Based on weighted gene co-expression network analysis, we determined the transcriptional network in experimental epidermolysis bullosa acquisita (EBA), a paradigm of an antibody-mediated organ-specific autoimmune disease characterized by autoantibodies directed against type VII collagen. We identified 33 distinct and differentially expressed modules, including Fcγ receptor (FcγR) IV and components of the neutrophil-associated enzyme system in autoantibody transfer-induced EBA. Validation experiments, including functional analysis, demonstrated that FcγRIV expression on neutrophils crucially contributes to autoantibody-induced tissue injury in the transfer model of EBA. Mice lacking the common γ-chain of activating FcγRs, deficient in FcγRIV or treated with FcγRIV function blocking antibody, but not mice deficient in FcγRI, FcγRIIB, FcγRIII or both FcγRI and FcγRIII, were effectively protected from EBA. Skin disease was restored in γ-chain-deficient mice locally reconstituted with neutrophils from wild-type, but not from γ-chain-deficient, mice. Our findings both genetically and functionally identify a novel disease-related molecule, FcγRIV, in an autoantibody-mediated disorder, which may be of importance for the development of novel targeted therapies.


Subject(s)
Autoimmune Diseases/genetics , Epidermolysis Bullosa Acquisita/genetics , Genetic Predisposition to Disease , Receptors, IgG/genetics , Animals , Antibodies, Blocking/pharmacology , Autoantibodies/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Collagen Type VII/immunology , Disease Models, Animal , Epidermolysis Bullosa Acquisita/drug therapy , Epidermolysis Bullosa Acquisita/immunology , Epidermolysis Bullosa Acquisita/pathology , Gene Expression Profiling , Immunoglobulin G/immunology , Immunoglobulin Isotypes , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Organ Specificity , Receptors, IgG/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL