Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Eur J Dermatol ; 29(2): 167-173, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30882351

ABSTRACT

Dermatitis herpetiformis (DH) and celiac disease (CD) are considered to be autoimmune diseases that share a specific trigger (gluten) and a common genetic background (HLA-DQ2/DQ8). However, the pathogenesis of DH is not yet fully understood and no data are available regarding a possible role of fibroblasts in this disease. The aim of this study was to assess baseline DNA damage in fibroblasts in DH-diagnosed patients vs. fibroblasts of controls without DH or CD. Primary fibroblast cultures were derived from dermal biopsies from DH patients and controls (without DH or CD). In vitro genotoxic damage was investigated using the comet assay and ɣH2AX test after different treatments (with 33mer peptide and digested gliadin [DG]) in order to investigate a correlation between oxidative stress (evaluated by reactive oxygen species formation) and glutathione content. Our results demonstrate a difference in baseline DNA damage between cutaneous fibroblasts of controls and DH patients, moreover, DNA damage significantly increased after exposure to gluten (DG and 33mer peptide) in fibroblasts from DH patients. DNA damage in fibroblasts from patients under dapsone treatment was similar to that of the control group. Our data indicate that oxidative stress and DNA damage may be characteristics of fibroblasts from DH patients who are not treated with dapsone, particularly after exposure to gliadin peptides.


Subject(s)
Celiac Disease/genetics , DNA Damage , Dermatitis Herpetiformis/genetics , Fibroblasts/cytology , Adult , Aged , Celiac Disease/immunology , Comet Assay , Dermatitis Herpetiformis/immunology , Female , Gliadin/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Histones/metabolism , Humans , Male , Middle Aged , Reactive Oxygen Species/metabolism
2.
Biomed Res Int ; 2017: 5965492, 2017.
Article in English | MEDLINE | ID: mdl-28808661

ABSTRACT

Itch which is one of the major, subjective symptoms in a course of bullous pemphigoid and dermatitis herpetiformis makes those two diseases totally different than other autoimmune blistering diseases. Its pathogenesis is still not fully known. The aim of this research was to assess the role of IL-31 in development of itch as well as to measure its intensity. Obtained results, as well as literature data, show that lower concentration of IL-31 in patients' serum may be correlated with its role in JAK/STAT signaling pathway which is involved in development of autoimmune blistering disease. Intensity of itch is surprisingly huge problem for the patients and the obtained results are comparable with results presented by atopic patients.


Subject(s)
Dermatitis Herpetiformis/blood , Interleukins/blood , Pemphigoid, Bullous/blood , Pruritus/blood , Aged , Aged, 80 and over , Autoimmune Diseases/blood , Autoimmune Diseases/genetics , Autoimmune Diseases/physiopathology , Dermatitis Herpetiformis/genetics , Dermatitis Herpetiformis/physiopathology , Female , Humans , Interleukins/genetics , Janus Kinases/genetics , Male , Middle Aged , Pemphigoid, Bullous/genetics , Pemphigoid, Bullous/physiopathology , Pruritus/genetics , Pruritus/physiopathology , STAT Transcription Factors/genetics , Signal Transduction
4.
Bioessays ; 38(5): 427-39, 2016 May.
Article in English | MEDLINE | ID: mdl-26990286

ABSTRACT

We propose a biochemical mechanism for celiac disease and non-celiac gluten sensitivity that may rationalize many of the extradigestive disorders not explained by the current immunogenetic model. Our hypothesis is based on the homology between the 33-mer gliadin peptide and a component of the NMDA glutamate receptor ion channel - the human GRINA protein - using BLASTP software. Based on this homology the 33-mer may act as a natural antagonist interfering with the normal interactions of GRINA and its partners. The theory is supported by numerous independent data from the literature, and provides a mechanistic link with otherwise unrelated disorders, such as cleft lip and palate, thyroid dysfunction, restless legs syndrome, depression, ataxia, hearing loss, fibromyalgia, dermatitis herpetiformis, schizophrenia, toxoplasmosis, anemia, osteopenia, Fabry disease, Barret's adenocarcinoma, neuroblastoma, urinary incontinence, recurrent miscarriage, cardiac anomalies, reduced risk of breast cancer, stiff person syndrome, etc. The hypothesis also anticipates better animal models, and has the potential to open new avenues of research.


Subject(s)
Celiac Disease/metabolism , Gliadin/metabolism , Models, Genetic , Receptors, N-Methyl-D-Aspartate/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Ataxia/genetics , Ataxia/metabolism , Ataxia/pathology , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Celiac Disease/chemically induced , Celiac Disease/genetics , Celiac Disease/pathology , Cleft Lip/genetics , Cleft Lip/metabolism , Cleft Lip/pathology , Cleft Palate/genetics , Cleft Palate/metabolism , Cleft Palate/pathology , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Dermatitis Herpetiformis/genetics , Dermatitis Herpetiformis/metabolism , Dermatitis Herpetiformis/pathology , Gene Expression Regulation , Gliadin/genetics , Glutens/adverse effects , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Binding , Protein Multimerization , Proteins/genetics , Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Sequence Homology, Amino Acid , Signal Transduction , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Thyroiditis/genetics , Thyroiditis/metabolism , Thyroiditis/pathology , Trans-Activators
7.
PLoS One ; 9(7): e101428, 2014.
Article in English | MEDLINE | ID: mdl-24999842

ABSTRACT

We performed a genome-wide association study (GWAS) of 1550 North American celiac disease cases and 3084 controls. Twelve SNPs, distributed across four regions (3p21.31, 4q27, 6q15, 6q25), were significantly associated with disease (p-value <1.0×10-7), and a further seven SNPs, across four additional regions (1q24.3, 10p15.1, 6q22.31, 17q21.32) had suggestive evidence (1.0×10-7 < p-value < 1.0×10-6). This study replicated a previous suggestive association within FRMD4B (3p14.1), confirming it as a celiac disease locus. All four regions with significant associations and two regions with suggestive results (1q24.3, 10p15.1) were known disease loci. The 6q22.31 and 10p11.23 regions were not replicated. A total of 410 SNPs distributed across the eight significant and suggestive regions were tested for association with dermatitis herpetiformis and microscopic colitis. Preliminary, suggestive statistical evidence for association with the two traits was found at chromosomes 3p21.31, 6q15, 6q25, 1q24.3 and 10p11.23, with future studies being required to validate the reported associations.


Subject(s)
Celiac Disease/genetics , Genetic Loci/genetics , Genome-Wide Association Study , Americas/epidemiology , Celiac Disease/complications , Celiac Disease/epidemiology , Colitis, Microscopic/complications , Colitis, Microscopic/genetics , Dermatitis Herpetiformis/complications , Dermatitis Herpetiformis/genetics , Humans , Polymorphism, Single Nucleotide
11.
G Ital Dermatol Venereol ; 148(2): 197-201, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23588145

ABSTRACT

Regulatory T cells (Tregs) play a crucial role by maintaining the peripheral tolerance and inhibiting autoimmunity. In recent years, numerous autoimmune and immune-mediated diseases have been shown to present significant number depletion and/or function impairment of this subset. In the present study, we present a brief overview of the results obtained by our group in association with the centers belonging to the Italian Immunopathology Group, as to the expression levels and biological significance of circulating regulatory CD4+CD25+brightFOXP3+ T cells in a variety of immune-mediated skin diseases (such as psoriasis, scleroderma, bullous pemphigoid and GvHD), together with preliminary results achieved in patients with inflammatory bowel disease-related dermatoses. This review shows that this series of different cutaneous diseases characterised by an immune-mediated pathogenesis, share a significant down-regulation of circulating FOXP3+ Treg cells, whilst the treatment and the achievement of clinical response are generally associated with an opposite phenomenon with up-regulation of Treg cells. Future studies are mandatory to identify the effective role of these modifications in the disease pathogenesis as well as its relationship with the clinical response.


Subject(s)
Dermatitis Herpetiformis/immunology , Flow Cytometry , T-Lymphocytes, Regulatory/immunology , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/immunology , Dermatitis Herpetiformis/genetics , Dermatitis Herpetiformis/metabolism , Dermatitis Herpetiformis/pathology , Down-Regulation/genetics , Flow Cytometry/methods , Forkhead Transcription Factors/genetics , Humans , Pemphigoid, Bullous/immunology , Psoriasis/immunology , Scleroderma, Systemic/immunology , T-Lymphocyte Subsets/immunology , Up-Regulation/genetics
12.
Tissue Antigens ; 80(6): 488-93, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23075394

ABSTRACT

Homozygosity for a nonsense mutation in the fucosyltransferase 2 (FUT2) gene (rs601338G>A) leads to the absence of ABH blood groups (FUT2 non-secretor status) in body fluids. As the secretor status has been shown to be a major determinant for the gut microbial spectrum, assumed to be important in the gut immune homeostasis, we studied the association of rs601338-FUT2 with celiac disease (CelD) and inflammatory bowel disease (IBD) in the Finnish population. Rs601338 was genotyped in CelD (n = 909), dermatitis herpetiformis (DH) (n = 116), ulcerative colitis (UC) (n = 496) and Crohn's disease (CD) (n = 280) patients and healthy controls (n = 2738). CelD showed significant genotypic [P = 0.0074, odds ratio (OR): 1.28] and recessive (P = 0.015, OR: 1.28) association with the rs601338-AA genotype. This was also found in the combined CelD+DH dataset (genotype association: P = 0.0060, OR: 1.28; recessive association: P < 0.011, OR: 1.28). The A allele of rs601338 showed nominal association with dominant protection from UC (P = 0.044, OR: 0.82) and UC+CD (P = 0.035, OR: 0.84). The frequency of non-secretors (rs601338-GG) in controls, CelD, DH, UC and CD datasets was 14.7%, 18%, 18.1%, 14.3% and 16.1%, respectively. No association was evident in the DH or CD datasets alone. In conclusion, FUT2 non-secretor status is associated with CelD susceptibility and FUT2 secretor status may also play a role in IBD in the Finnish population.


Subject(s)
Celiac Disease/enzymology , Celiac Disease/genetics , Fucosyltransferases/genetics , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/genetics , Alleles , Base Sequence , Case-Control Studies , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/genetics , Crohn Disease/enzymology , Crohn Disease/genetics , DNA Primers/genetics , Dermatitis Herpetiformis/enzymology , Dermatitis Herpetiformis/genetics , Finland , Genes, Recessive , Genetic Association Studies , Genotype , Humans , Polymorphism, Single Nucleotide , Risk Factors , Galactoside 2-alpha-L-fucosyltransferase
13.
Clin Dev Immunol ; 2012: 198956, 2012.
Article in English | MEDLINE | ID: mdl-22991566

ABSTRACT

Dermatitis herpetiformis (DH) is an autoimmune blistering skin disease associated with gluten-sensitive enteropathy (CD). In order to investigate the pathogenesis of skin lesions at molecular level, we analysed the gene expression profiles in skin biopsies from 6 CD patients with DH and 6 healthy controls using Affymetrix HG-U133A 2.0 arrays. 486 genes were differentially expressed in DH skin compared to normal skin: 225 were upregulated and 261 were downregulated. Consistently with the autoimmune origin of DH, functional classification of the differentially expressed genes (DEGs) indicates a B- and T-cell immune response (LAG3, TRAF5, DPP4, and NT5E). In addition, gene modulation provides evidence for a local inflammatory response (IL8, PTGFR, FSTL1, IFI16, BDKRD2, and NAMPT) with concomitant leukocyte recruitment (CCL5, ENPP2), endothelial cell activation, and neutrophil extravasation (SELL, SELE). DEGs also indicate overproduction of matrix proteases (MMP9, ADAM9, and ADAM19) and proteolytic enzymes (CTSG, ELA2, CPA3, TPSB2, and CMA1) that may contribute to epidermal splitting and blister formation. Finally, we observed modulation of genes involved in cell growth inhibition (CGREF1, PA2G4, and PPP2R1B), increased apoptosis (FAS, TNFSF10, and BASP1), and reduced adhesion at the dermal epidermal junction (PLEC1, ITGB4, and LAMA5). In conclusion, our results identify genes that are involved in the pathogenesis of DH skin lesions.


Subject(s)
Dermatitis Herpetiformis/genetics , Gene Expression Profiling , Skin/immunology , Skin/metabolism , Adult , Apoptosis/genetics , B-Lymphocytes/immunology , Cell Adhesion/genetics , Cell Proliferation , Cells, Cultured , Dermatitis Herpetiformis/immunology , Dermatitis Herpetiformis/pathology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Humans , Inflammation , Leukocytes/immunology , Leukocytes/metabolism , Lymphocyte Activation , Male , Matrix Metalloproteinases/biosynthesis , Middle Aged , Neutrophils/immunology , Neutrophils/metabolism , Peptide Hydrolases/biosynthesis , Skin/pathology , T-Lymphocytes/immunology
14.
Clin Dev Immunol ; 2012: 239691, 2012.
Article in English | MEDLINE | ID: mdl-22778763

ABSTRACT

Dermatitis herpetiformis (DH) is a rare autoimmune disease linked to gluten sensitivity with a chronic-relapsing course. It is currently considered to be the specific cutaneous manifestation of celiac disease (CD). Both conditions are mediated by the IgA class of autoantibodies, and the diagnosis of DH is dependent on the detection of granular deposits of IgA in the skin. There is an underlying genetic predisposition to the development of DH, but environmental factors are also important. This paper describes these different factors and discusses the known mechanism that lead to the development of skin lesions.


Subject(s)
Dermatitis Herpetiformis/genetics , Dermatitis Herpetiformis/pathology , Animals , Dermatitis Herpetiformis/immunology , Environment , Gene-Environment Interaction , Humans , Immunoglobulin A/immunology , Transglutaminases/immunology
15.
Clin Dev Immunol ; 2012: 562168, 2012.
Article in English | MEDLINE | ID: mdl-22778765

ABSTRACT

We reviewed all 91 Japanese dermatitis herpetiformis (DH) patients reported over the last 35 years. The male-to-female ratio was 2 : 1. The mean age at onset was 43.8, and 13 years earlier for female patients. More than half of these Japanese DH patients showed granular IgA deposition in the papillary dermis, and another one-third showed fibrillar IgA deposition. The male patients with granular IgA deposition were 10 years older than those with fibrillar deposition. Whereas patients with granular IgA deposition showed typical distribution of the skin lesions, the predilection sites of DH tended to be spared in patients with fibrillar IgA deposition. Only 3 patients had definite gluten-sensitive enteropathy. There was a statistical difference in the frequency of human leukocyte antigen (HLA)-DR9 between the granular group and controls among Japanese. No patients had HLA-DQ2 or -DQ8, which is frequently found in Caucasian DH patients. The absence of HLA-DQ2/DQ8, the inability to identify celiac disease in most cases, the predominance of fibrillar IgA, and the unusual distribution of clinical lesions in Japanese patients suggest that Japanese DH may be a subset of DH patients and have a pathogenesis which is different from that currently proposed in Caucasian DH patients.


Subject(s)
Asian People , Dermatitis Herpetiformis/immunology , Dermatitis Herpetiformis/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Dermatitis Herpetiformis/genetics , Female , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , Humans , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Infant , Japan , Male , Middle Aged , Young Adult
16.
Semin Immunopathol ; 34(4): 497-511, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22572887

ABSTRACT

The initial development and maintenance of tolerance to dietary antigens is a complex process that, when prevented or interrupted, can lead to human disease. Understanding the mechanisms by which tolerance to specific dietary antigens is attained and maintained is crucial to our understanding of the pathogenesis of diseases related to intolerance of specific dietary antigens. Two diseases that are the result of intolerance to a dietary antigen are celiac disease (CD) and dermatitis herpetiformis (DH). Both of these diseases are dependent upon the ingestion of gluten (the protein fraction of wheat, rye, and barley) and manifest in the gastrointestinal tract and skin, respectively. These gluten-sensitive diseases are two examples of how devastating abnormal immune responses to a ubiquitous food can be. The well-recognized risk genotype for both is conferred by either of the HLA class II molecules DQ2 or DQ8. However, only a minority of individuals who carry these molecules will develop either disease. Also of interest is that the age at diagnosis can range from infancy to 70-80 years of age. This would indicate that intolerance to gluten may potentially be the result of two different phenomena. The first would be that, for various reasons, tolerance to gluten never developed in certain individuals, but that for other individuals, prior tolerance to gluten was lost at some point after childhood. Of recent interest is the concept of non-celiac gluten sensitivity, which manifests as chronic digestive or neurologic symptoms due to gluten, but through mechanisms that remain to be elucidated. This review will address how animal models of gluten-sensitive disorders have substantially contributed to a better understanding of how gluten intolerance can arise and cause disease.


Subject(s)
Celiac Disease/immunology , Dermatitis Herpetiformis/immunology , Disease Models, Animal , Glutens/immunology , Animals , Autoimmunity/genetics , CD4-Positive T-Lymphocytes/immunology , Celiac Disease/genetics , Celiac Disease/metabolism , Celiac Disease/therapy , Dermatitis Herpetiformis/genetics , Dermatitis Herpetiformis/metabolism , Dermatitis Herpetiformis/therapy , Epithelial Cells/immunology , Genetic Predisposition to Disease , Histocompatibility Antigens Class II/immunology , Humans , Immune Tolerance , Interleukin-15/immunology , Interleukin-15/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/microbiology , Lymphocyte Activation , Metagenome , T-Lymphocytes, Regulatory/immunology , Transglutaminases/metabolism
17.
Immunol Allergy Clin North Am ; 32(2): 263-74, vi, 2012 May.
Article in English | MEDLINE | ID: mdl-22560139

ABSTRACT

Dermatitis herpetiformis (DH) is an autoimmune blistering skin disease in which antigen presentation in the gastrointestinal mucosa results in cutaneous IgA deposition and distinct, neutrophil-driven cutaneous lesions. Our findings suggest that the qualitatively different immune response to gluten in the intestinal mucosa of patients with DH results in minimal clinical symptoms, allowing the continued ingestion of gluten and the eventual development of DH. Our model may provide a new way to understand the pathogenesis of other skin diseases associated with gastrointestinal inflammation such as pyoderma gangrenosum or erythema nodosum, or explain association of seronegative inflammatory arthritis with inflammatory bowel disease.


Subject(s)
Celiac Disease/immunology , Dermatitis Herpetiformis/immunology , Skin/immunology , Animals , Celiac Disease/genetics , Celiac Disease/pathology , Dermatitis Herpetiformis/genetics , Dermatitis Herpetiformis/pathology , Diet, Gluten-Free , Humans , Immunity, Mucosal , Immunoglobulin A/immunology , Inflammation Mediators/metabolism , Interleukin-8/metabolism , Models, Biological , Skin/pathology , Tumor Necrosis Factor-alpha/metabolism
19.
J Am Acad Dermatol ; 64(6): 1017-24; quiz 1025-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21571167

ABSTRACT

Dermatitis herpetiformis (DH) is an autoimmune disease that is linked to gluten sensitivity and has a clear relationship to celiac disease. Both conditions are mediated by the IgA class of autoantibodies and the diagnosis of DH is dependent on detection of granular deposits of IgA in the skin. There is an underlying genetic predisposition to the development of DH but environmental factors are also important. Typically, young adults present with excoriations only, as the severe pruritus effectively destroys any primary lesions. Based upon our experience with DH and a comprehensive literature review, we provide an update of DH epidemiology, pathophysiology, and clinical presentation.


Subject(s)
Dermatitis Herpetiformis/physiopathology , Animals , Celiac Disease/epidemiology , Comorbidity , Dermatitis Herpetiformis/diagnosis , Dermatitis Herpetiformis/epidemiology , Dermatitis Herpetiformis/genetics , Dermatitis Herpetiformis/immunology , Diagnosis, Differential , Genetic Predisposition to Disease , Humans , Hypothyroidism/epidemiology , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Lymphoma, Non-Hodgkin/epidemiology
20.
J Am Acad Dermatol ; 63(5): 892-5, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739096

ABSTRACT

Dermatitis herpetiformis (DH) is an immune-mediated cutaneous disease occasionally associated with celiac disease, but rarely associated with systemic lupus erythematosus (SLE). The combination of DH and SLE is immunologically mediated and suggests a relationship between the two conditions. We describe a woman with DH and SLE with a novel HLA phenotype.


Subject(s)
Dermatitis Herpetiformis/complications , Dermatitis Herpetiformis/genetics , Histocompatibility Testing , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/genetics , Adult , Biopsy , Dermatitis Herpetiformis/pathology , Female , Haplotypes , Humans , Lupus Erythematosus, Systemic/pathology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL