16 research outputs found

    Molecular characterization of atopic dermatitis:a meta-analysis

    Get PDF

    Meta-analysis derived atopic dermatitis (MADAD) transcriptome defines a robust AD signature highlighting the involvement of atherosclerosis and lipid metabolism pathways

    Get PDF
    BACKGROUND: Atopic dermatitis (AD) is a common inflammatory skin disease with limited treatment options. Several microarray experiments have been conducted on lesional/LS and non-lesional/NL AD skin to develop a genomic disease phenotype. Although these experiments have shed light on disease pathology, inter-study comparisons reveal large differences in resulting sets of differentially expressed genes (DEGs), limiting the utility of direct comparisons across studies. METHODS: We carried out a meta-analysis combining 4 published AD datasets to define a robust disease profile, termed meta-analysis derived AD (MADAD) transcriptome. RESULTS: This transcriptome enriches key AD pathways more than the individual studies, and associates AD with novel pathways, such as atherosclerosis signaling (IL-37, selectin E/SELE). We identified wide lipid abnormalities and, for the first time in vivo, correlated Th2 immune activation with downregulation of key epidermal lipids (FA2H, FAR2, ELOVL3), emphasizing the role of cytokines on the barrier disruption in AD. Key AD “classifier genes” discriminate lesional from nonlesional skin, and may evaluate therapeutic responses. CONCLUSIONS: Our meta-analysis provides novel and powerful insights into AD disease pathology, and reinforces the concept of AD as a systemic disease. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s12920-015-0133-x) contains supplementary material, which is available to authorized users

    Genetic Associations for Activated Partial Thromboplastin Time and Prothrombin Time, their Gene Expression Profiles, and Risk of Coronary Artery Disease

    Get PDF
    Activatedpartialthromboplastintime (aPTT) and prothrombintime (PT) are clinical tests commonly used to screen for coagulation-factor deficiencies. One genome-wide association study (GWAS) has been reported previously for aPTT, but no GWAS has been reported for PT. We conducted a GWAS and meta-analysis to identify genetic loci for aPTT and PT. The GWAS for aPTT was conducted in 9,240 individuals of European ancestry from the Atherosclerosis Risk in Communities (ARIC) study, and the GWAS for PT was conducted in 2,583 participants from the Genetic Study of Three Population Microisolates in South Tyrol (MICROS) and the Lothian Birth Cohorts (LBC) of 1921 and 1936. Replication was assessed in 1,041 to 3,467 individuals. For aPTT, previously reported associations with KNG1, HRG, F11, F12, and ABO were confirmed. A second independent association in ABO was identified and replicated (rs8176704, p = 4.26 × 10−24). Pooling the ARIC and replication data yielded two additional loci in F5 (rs6028, p = 3.22 × 10−9) and AGBL1 (rs2469184, p = 3.61 × 10−8). For PT, significant associations were identified and confirmed in F7 (rs561241, p = 3.71 × 10−56) and PROCR/EDEM2 (rs2295888, p = 5.25 × 10−13). Assessment of existing geneexpression and coronaryarterydisease (CAD) databases identified associations of five of the GWAS loci with altered geneexpression and two with CAD. In summary, eight genetic loci that account for ∌29% of the variance in aPTT and two loci that account for ∌14% of the variance in PT were detected and supported by functional data

    Profiling the Atopic Dermatitis Epidermal Transcriptome by Tape Stripping and BRB-seq

    No full text
    Tape stripping is a non-invasive skin sampling technique, which has recently gained use for the study of the transcriptome of atopic dermatitis (AD), a common inflammatory skin disorder characterized by a defective epidermal barrier and perturbated immune response. Here, we performed BRB-seq—a low cost, multiplex-based, transcriptomic profiling technique—on tape-stripped skin from 30 AD patients and 30 healthy controls to evaluate the methods’ ability to assess the epidermal AD transcriptome. An AD signature consisting of 91 differentially expressed genes, specific for skin barrier and inflammatory response, was identified. The gene expression in the outermost layers, stratum corneum and stratum granulosum, of the skin showed highest correlation between tape-stripped skin and matched full-thickness punch biopsies. However, we observed that low and highly variable transcript counts, probably due to low RNA yield and RNA degradation in the tape-stripped skin samples, were a limiting factor for epidermal transcriptome profiling as compared to punch biopsies. We conclude that deep BRB-seq of tape-stripped skin is needed to counteract large between-sample RNA yield variation and highly zero-inflated data in order to apply this protocol for population-wide screening of the epidermal transcriptome in inflammatory skin diseases

    Identification of novel immune and barrier genes in atopic dermatitis by means of laser capture microdissection

    No full text
    BACKGROUND: The molecular signature of atopic dermatitis/AD lesions is associated with Th2 and Th22 activation, and epidermal alterations. However, the epidermal and dermal AD transcriptomes and their respective contributions to abnormalities in respective immune and barrier phenotypes are unknown. OBJECTIVE: To establish the genomic profile of the epidermal and dermal compartments of lesional/LS and non-lesional/NL AD, as compared with normal skin. METHODS: Laser capture micro-dissection/LCM was performed to separate epidermis and dermis of LS and NL skin from AD patients and normal skin from healthy volunteers followed by gene expression (microarrays and RT-PCR) and immunostaining studies. RESULTS: Our study identified novel immune and barrier genes, including the IL-34 cytokine and claudins 4 and 8, and showed increased detection of key AD genes usually undetectable on arrays (i.e. IL-22, TSLP, CCL22, and CCL26). Overall, the combined epidermal and dermal transcriptomes enlarged the AD transcriptome adding 674 up-regulated and 405 down-regulated differentially expressed genes between LS and NL skin to the AD transcriptome. We were also able to localize individual transcripts as primarily epidermal (DEFB4A) or dermal (IL-22, CTLA4, and CCR7), and link their expressions to possible cellular sources. CONCLUSIONS: This is the first report that establishes robust epidermal and dermal genomic signatures of LS, NL AD and normal/N skin, as compared with whole tissues. These data establish the utility of LCM to separate different compartments and cellular subsets in AD, allowing localization of key barrier or immune molecules, and enable detection of gene products usually not detected on arrays
    corecore