Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Am Acad Dermatol ; 86(2): 322-330, 2022 02.
Article in English | MEDLINE | ID: mdl-34339761

ABSTRACT

BACKGROUND: Although hidradenitis suppurativa (HS) shares some transcriptomic and cellular infiltrate features with psoriasis, their skin proteome remains unknown. OBJECTIVE: To define and compare inflammatory protein biomarkers of HS and psoriasis skin. METHODS: We assessed 92 inflammatory biomarkers in HS (n = 13), psoriasis (n = 11), and control skin (n = 11) using Olink high-throughput proteomics. We also correlated HS skin and blood biomarkers using proteomics and RNA sequencing. RESULTS: We identified 57 differentially expressed proteins (DEPs) in lesional psoriasis and 64 DEPs in lesional HS skin, compared to healthy controls. Both HS and psoriasis lesional skin demonstrated a significant upregulation of T helper 1 and T helper 17 proteins. Healthy-appearing perilesional HS skin had 63 DEPs compared to healthy controls. Nonlesional HS and psoriasis skin had 24 and 7 DEPs, respectively, compared to healthy controls. Tumor necrosis factor and 8 other proteins were significantly correlated with clinical severity in perilesional HS skin (2 cm from a nodule). LIMITATIONS: Inclusion of only moderate-to-severe patients and the cohort size. CONCLUSION: HS has a greater inflammatory profile and is more diffusely distributed compared with psoriasis. Proteins correlated with disease severity are potential disease mediators. Perilesional skin is comparably inflamed to lesional skin, suggesting the need to treat beyond skin nodules.


Subject(s)
Hidradenitis Suppurativa , Psoriasis , Biomarkers/metabolism , Hidradenitis Suppurativa/genetics , Humans , Proteome/metabolism , Psoriasis/pathology , Skin/pathology
2.
J Allergy Clin Immunol ; 143(1): 142-154, 2019 01.
Article in English | MEDLINE | ID: mdl-30121291

ABSTRACT

BACKGROUND: IL-22 is potentially a pathogenic cytokine in patients with atopic dermatitis (AD), but the molecular effects of IL-22 antagonism have not been defined in human subjects. OBJECTIVE: We sought to evaluate the cellular and molecular effects of IL-22 blockade in tissues from patients with moderate-to-severe AD. METHODS: We assessed lesional and nonlesional skin from 59 patients with moderate-to-severe AD treated with anti-IL-22 (fezakinumab) versus placebo (2:1) using transcriptomic and immunohistochemistry analyses. RESULTS: Greater reversal of the AD genomic profile was seen with fezakinumab versus placebo, namely 25.3% versus 10.5% at 4 weeks (P = 1.7 × 10-5) and 65.5% versus 13.9% at 12 weeks (P = 9.5 × 10-19), respectively. Because IL-22 blockade showed clinical efficacy only in patients with severe AD, we used baseline median IL-22 mRNA expression to stratify for high (n = 30) and low (n = 29) IL-22 expression groups. Much stronger mean transcriptomic improvements were seen with fezakinumab in the IL-22-high drug-treated group (82.8% and 139.4% at 4 and 12 weeks, respectively) than in the respective IL-22-high placebo-treated group (39.6% and 56.3% at 4 and 12 weeks) or the IL-22-low groups. Significant downregulations of multiple immune pathways, including TH1/CXCL9, TH2/CCL18/CCL22, TH17/CCL20/DEFB4A, and TH22/IL22/S100A's, were restricted to the IL-22-high drug group (P < .05). Consistently, tissue predictors of clinical response were mostly genes involved in T-cell and dendritic cell activation and differentiation. CONCLUSIONS: This is the first report showing a profound effect of IL-22 blockade on multiple inflammatory pathways in AD. These data, supported by robust effects in patients with high IL-22 baseline expression, suggest a central role for IL-22 in AD, indicating the need for a precision medicine approach for improving therapeutic outcomes in patients with AD.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Dermatitis, Atopic/drug therapy , Gene Expression Regulation/drug effects , Interleukins/biosynthesis , Skin/metabolism , Adult , Antibodies, Monoclonal, Humanized , Dermatitis, Atopic/immunology , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Female , Gene Expression Regulation/immunology , Humans , Interleukins/immunology , Male , Middle Aged , Skin/immunology , Skin/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology , Interleukin-22
3.
Exp Dermatol ; 26(1): 28-35, 2017 01.
Article in English | MEDLINE | ID: mdl-27304428

ABSTRACT

Atopic dermatitis (AD) is the most common inflammatory skin disease, but treatment options for moderate-to-severe disease are limited. Ustekinumab is an IL-12/IL-23p40 antagonist that suppresses Th1, Th17 and Th22 activation, commonly used for psoriasis patients. We sought to assess efficacy and safety of ustekinumab in patients with moderate-to-severe AD. In this phase II, double-blind, placebo-controlled study, 33 patients with moderate-to-severe AD were randomly assigned to either ustekinumab (n=16) or placebo (n=17), with subsequent crossover at 16 weeks, and last dose at 32 weeks. Background therapy with mild topical steroids was allowed to promote retention. Study endpoints included clinical (SCORAD50) and biopsy-based measures of tissue structure and inflammation, using protein and gene expression studies. The ustekinumab group achieved higher SCORAD50 responses at 12, 16 (the primary endpoint) and 20 weeks compared to placebo, but the difference between groups was not significant. The AD molecular profile/transcriptome showed early robust gene modulation, with sustained further improvements until 32 weeks in the initial ustekinumab group. Distinct and more robust modulation of Th1, Th17 and Th22 but also Th2-related AD genes was seen after 4 weeks of ustekinumab treatment (i.e. MMP12, IL-22, IL-13, IFN-γ, elafin/PI3, CXCL1 and CCL17; P<.05). Epidermal responses (K16, terminal differentiation) showed faster (4 weeks) and long-term regulation (32 weeks) from baseline in the ustekinumab group. No severe adverse events were observed. Ustekinumab had clear clinical and molecular effects, but clinical outcomes might have been obscured by a profound "placebo" effect, most likely due to background topical glucocorticosteroids and possibly insufficient dosing for AD.


Subject(s)
Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/genetics , Dermatologic Agents/therapeutic use , Ustekinumab/therapeutic use , Adult , Chemokine CCL17/genetics , Chemokine CXCL1/genetics , Cross-Over Studies , Dermatitis, Atopic/pathology , Dermatologic Agents/adverse effects , Double-Blind Method , Elafin/genetics , Female , Gene Expression Profiling , Humans , Interferon-gamma/genetics , Interleukin-13/genetics , Interleukins/genetics , Male , Matrix Metalloproteinase 12/genetics , Severity of Illness Index , Transcriptome/drug effects , Ustekinumab/adverse effects , Interleukin-22
4.
J Allergy Clin Immunol ; 138(1): 169-178, 2016 07.
Article in English | MEDLINE | ID: mdl-26948076

ABSTRACT

BACKGROUND: Topical glucocorticosteroids are considered an efficient treatment option for atopic dermatitis (AD), but a global assessment of glucocorticosteroid responses on key disease circuits upon weeks to months of treatment is currently lacking. OBJECTIVE: We sought to assess short (4 weeks) and long-term (16 weeks) application of topical glucocorticosteroids on AD skin and define response biomarkers. METHODS: The effects of triamcinolone acetonide cream 0.025% were assessed based on gene expression and immunohistochemistry studies at baseline, 4 weeks, and 16 weeks in biopsy specimens from 15 patients with moderate-to-severe AD. RESULTS: At 16 weeks, only 3 patients were clinical responders (by using SCORAD50 criteria), but 6 patients qualified as responders based on histologic criteria. Baseline characteristics indicated more severe disease in nonresponders. While 3 of 15 patients experienced only transient benefit after 4 weeks, others showed progressive improvements toward 16 weeks. Topical glucocorticosteroid use in patients with AD resulted in improvements of the AD genomic signature of 25.6% at 4 weeks and 71.8% at 16 weeks, respectively, and even 123.9% in the histologic responder group. Cytokines (IL-12p40, IL-13, IL-22, CCL17, CCL18, peptidase inhibitor 3 [PI3]/elafin, and S100As) showed consistent decreases from baseline toward 16 weeks with corresponding improvements in epidermal disease hallmarks (keratin 16 and loricrin) in lesional skin from responders (P < .05). Nonresponders largely showed lesser/nonsignificant reductions in key inflammatory and barrier markers (keratin 16, IL-13, IL-22, CCL17, CCL18, PI3/elafin, S100As, and loricrin). The combination of IL-21 and IFN-γ baseline expression closely predicted individual clinical glucocorticosteroid responses at 16 weeks of treatment. CONCLUSION: Our study indicates that even low-potency glucocorticosteroids can broadly affect immune and barrier responses in patients with moderate-to-severe AD, associating higher baseline severity with increased steroid resistance in patients with AD.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Dermatitis, Atopic/drug therapy , Steroids/administration & dosage , Administration, Topical , Adult , Aged , Biopsy , Cluster Analysis , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/genetics , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Male , Middle Aged , Severity of Illness Index , Skin/metabolism , Skin/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Time Factors , Treatment Outcome , Young Adult
5.
J Invest Dermatol ; 134(5): 1418-1427, 2014 May.
Article in English | MEDLINE | ID: mdl-24270662

ABSTRACT

The precise mechanisms governing invasion at the leading edge of squamous cell carcinoma (SCC) and its subsequent metastasis are not fully understood. We aimed to define the cancer-related molecular changes that distinguish noninvasive tumor from invasive SCC. To this end, we combined laser capture microdissection with complementary DNA (cDNA) microarray analysis. We defined invasion-associated genes as those differentially regulated only in invasive SCC nests, but not in actinic keratosis or in situ SCC, compared with normal epidermis. There were 383 upregulated and 354 downregulated genes in the "invasion set." SCC invasion was characterized by aberrant expression of various proteolytic molecules. We noted increased expression of MMP7 and IL-24 in invasive SCC. IL-24 induced the expression of matrix metallopeptidase 7 (MMP7) in SCC cells in culture. In addition, blocking of MMP7 by a specific antibody significantly delayed the migration of SCC cells in culture. These results suggest a possible contribution of IL-24 to SCC invasion via enhancing focal expression of MMP7, although IL-24 has been suggested to have antitumor growth effects in other cancer types. Identification of regional molecular changes that regulate cancer invasion may facilitate the development of new targeted treatments for aggressive cancer.


Subject(s)
Carcinoma, Squamous Cell/genetics , Interleukins/genetics , Matrix Metalloproteinase 7/genetics , Skin Neoplasms/genetics , Transcriptome , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Interleukins/metabolism , Matrix Metalloproteinase 7/metabolism , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Oligonucleotide Array Sequence Analysis , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
6.
Oncotarget ; 4(12): 2237-48, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24163262

ABSTRACT

The pathogenesis of BCC is associated with sonic hedgehog (SHH) signaling. Vismodegib, a smoothened inhibitor that targets this pathway, is now in clinical use for advanced BCC patients, but its efficacy is limited. Therefore, new therapeutic options for this cancer are required. We studied gene expression profiling of BCC tumour tissues coupled with laser capture microdissection to identify tumour specific receptor tyrosine kinase expression that can be targeted by small molecule inhibitors. We found a >250 fold increase (FDR<10-4) of the oncogene, anaplastic lymphoma kinase (ALK) as well as its ligands, pleiotrophin and midkine in BCC compared to microdissected normal epidermis. qRT-PCR confirmed increased expression of ALK (p<0.05). Stronger expression of phosphorylated ALK in BCC tumour nests than normal skin was observed by immunohistochemistry. Crizotinib, an FDA-approved ALK inhibitor, reduced keratinocyte proliferation in culture, whereas a c-Met inhibitor did not. Crizotinib significantly reduced the expression of GLI1 and CCND2 (members of SHH-pathway) mRNA by approximately 60% and 20%, respectively (p<0.01). Our data suggest that ALK may increase GLI1 expression in parallel with the conventional SHH-pathway and promote keratinocyte proliferation. Hence, an ALK inhibitor alone or in combination with targeting SHH-pathway molecules may be a potential treatment for BCC patients.


Subject(s)
Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/enzymology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/enzymology , Anaplastic Lymphoma Kinase , Animals , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Crizotinib , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratinocytes/metabolism , Laser Capture Microdissection , Mice , Mice, Transgenic , Molecular Targeted Therapy , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Zinc Finger Protein GLI1
7.
JAMA Dermatol ; 149(2): 178-86, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23560298

ABSTRACT

OBJECTIVE: To characterize the presence of CD200 and CD200 receptor (CD200R) in the human cutaneous squamous cell carcinoma (SCC) microenvironment and to define a possible role for the CD200 axis in immune evasion by SCC. DESIGN: Gene expression in SCC vs normal skin was studied. Laser capture microdissection was used to determine differential expression of CD200 in normal skin vs actinic keratosis vs SCC in situ vs invasive SCC. Immunofluorescence microscopy was used to define expression of CD200R on macrophages, myeloid dendritic cells, natural killer cells, and T cells in SCC vs normal skin. The effects of SCC supernatant on induction of CD200 in human blood endothelial cells was also examined. SETTING: Academic Medical Center with an established Section of Mohs and Dermatologic Surgery and an active Cutaneous Biology Research Program. PARTICIPANTS: Surgical discard tissue from deidentified patients and samples of normal skin from healthy volunteers were used in this study. MAIN OUTCOME MEASURES: Expression of CD200 on SCC-associated blood vessels; expression of CD200 receptor on SCC-associated macrophages and T cells; and induction of CD200 on endothelial cells by SCC supernatants. RESULTS: CD200 gene and message were upregulated in SCC stroma. Immunostaining revealed a higher number of CD200(+) cells in SCC stroma than in normal dermis (180.8 cells/mm(2) vs 24.6 cells/mm(2)) (P<.01). CD200 was further identified mainly on blood vessel endothelium in SCC. Tumor supernatant was able to induce CD200 expression on human dermal blood endothelial cells in culture. CD200R was identified on macrophages and dendritic cells in SCC microenvironment. CONCLUSIONS: CD200 expression on local blood vessels may promote tumor progression by suppressing CD200R myeloid cells during diapedesis. These data highlight a previously unrecognized mechanism of immune evasion by SCC and may provide guidance for the development of targeted therapy.


Subject(s)
Antigens, CD/genetics , Antigens, Surface/genetics , Carcinoma, Squamous Cell/pathology , Receptors, Cell Surface/genetics , Skin Neoplasms/pathology , Up-Regulation , Academic Medical Centers , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Dendritic Cells/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation, Neoplastic , Humans , Killer Cells, Natural/metabolism , Laser Capture Microdissection , Macrophages/metabolism , Microscopy, Fluorescence , Mohs Surgery , Orexin Receptors , Skin/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/immunology , T-Lymphocytes/metabolism , Transendothelial and Transepithelial Migration , Tumor Microenvironment
8.
PLoS One ; 7(11): e49408, 2012.
Article in English | MEDLINE | ID: mdl-23226210

ABSTRACT

The autoinflammatory disorder, Neonatal-onset Multisystem Inflammatory Disease (NOMID) is the most severe phenotype of disorders caused by mutations in CIAS1 that result in increased production and secretion of active IL-1ß. NOMID patients present with systemic and organ-specific inflammation of the skin, central nervous system and bone, and respond dramatically to treatment with IL-1 blocking agents. We compared the cellular infiltrates and transcriptome of skin biopsies from patients with NOMID (n = 14) before treatment (lesional (LS) and non-lesional (pre-NL) skin) and after treatment (post-NL) with the IL-1 blocker anakinra (recombinant IL-1 receptor antagonist, Kineret®, Swedish Orphan Biovitrum AB, SOBI), to normal skin (n = 5) to assess tissue responses in the context of untreated and treated disease. Abundant neutrophils distinguish LS skin from pre-NL and post-NL skin. CD11c(+) dermal dendritic cells and CD163(+) macrophages expressed activated caspase-1 and are a likely source of cutaneous IL-1 production. Treatment with anakinra led to the disappearance of neutrophils, but CD3(+) T cells and HLA-DR(+) cells remained elevated. Among the upregulated genes IL-6, IL-8, TNF, IL-17A, CCL20, and the neutrophil defensins DEFA1 and DEFA3 were differentially regulated in LS tissues (compared to normal skin). Important significantly downregulated pathways in LS skin included IL-1R/TLR signaling, type I and II cytokine receptor signaling, mitochondrial dysfunction, and antigen presentation. The differential expression and regulation of microRNAs and pathways involved in post-transcriptional modification were suggestive of epigenetic modification in the chronically inflamed tissue. Overall, the dysregulated genes and pathways suggest extensive "adaptive" mechanisms to control inflammation and maintain tissue homeostasis, likely triggered by chronic IL-1 release in the skin of patients with NOMID.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/immunology , Homeostasis/drug effects , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1beta/genetics , Receptors, Interleukin-1/genetics , Skin/immunology , Adolescent , Adult , Antigens, CD/genetics , Antigens, CD/immunology , Biopsy , Caspase 1/genetics , Caspase 1/immunology , Child , Child, Preschool , Cryopyrin-Associated Periodic Syndromes/drug therapy , Cryopyrin-Associated Periodic Syndromes/genetics , Cryopyrin-Associated Periodic Syndromes/pathology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/immunology , Female , Gene Expression/drug effects , Gene Expression/immunology , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Homeostasis/genetics , Homeostasis/immunology , Humans , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/immunology , Langerhans Cells/drug effects , Langerhans Cells/immunology , Langerhans Cells/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , MicroRNAs/genetics , MicroRNAs/immunology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/immunology , Signal Transduction/drug effects , Skin/drug effects , Skin/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
9.
J Invest Dermatol ; 132(6): 1615-26, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22402443

ABSTRACT

Psoriasis vulgaris is a complex disease characterized by alterations in growth and differentiation of epidermal keratinocytes, as well as a marked increase in leukocyte populations. Lesions are known to contain alterations in messenger RNAs encoding more than 1,000 products, but only a very small number of these transcripts has been localized to specific cell types or skin regions. In this study, we used laser capture microdissection (LCM) and gene array analysis to study the gene expression of cells in lesional epidermis (EPI) and dermis, compared with the corresponding non-lesional regions. Using this approach, we detected >1,800 differentially expressed gene products in the EPI or dermis of psoriasis lesions. These results established sets of genes that are differentially expressed between epidermal and dermal compartments, as well as between non-lesional and lesional psoriasis skin. One of our findings involved the local production of CCL19, a lymphoid-organizing chemokine, and its receptor CCR7 in psoriatic dermal aggregates, along with the presence of gene products LAMP3/DC-LAMP and CD83, which typify mature dendritic cells (DCs). Gene expression patterns obtained with LCM and microarray analysis along with T-cell and DC detection by immune staining suggest a possible mechanism for lymphoid organization via CCL19/CCR7 in diseased skin.


Subject(s)
Dermis/pathology , Laser Capture Microdissection/methods , Oligonucleotide Array Sequence Analysis/methods , Psoriasis/genetics , Psoriasis/pathology , Dermis/immunology , Fluorescent Antibody Technique/methods , Fluorescent Antibody Technique/standards , Gene Expression Profiling/methods , Gene Expression Profiling/standards , Humans , Immunohistochemistry/methods , Immunohistochemistry/standards , Laser Capture Microdissection/standards , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Oligonucleotide Array Sequence Analysis/standards , Psoriasis/immunology , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...