51
|
Bhattacharya N, Ganguli-Indra G, Indra AK. CTIP2 and lipid metabolism: regulation in skin development and associated diseases. Expert Rev Proteomics 2021; 18:1009-1017. [PMID: 34739354 PMCID: PMC9119322 DOI: 10.1080/14789450.2021.2003707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/02/2021] [Indexed: 12/25/2022]
Abstract
INTRODUCTION COUP-TF INTERACTING PROTEIN 2 (CTIP2) is a crucial transcription factor exhibiting its control through coupled modulation of epigenetic modification and transcriptional regulation of key genes related to skin, immune, and nervous system development. Previous studies have validated the essential role of CTIP2 in skin development and maintenance, propagating its effects in epidermal permeability barrier (EPB) homeostasis, wound healing, inflammatory diseases, and epithelial cancers. Lipid metabolism dysregulation, on the other hand, has also established its independent emerging role over the years in normal skin development and various skin-associated ailments. This review focuses on the relatively unexplored connections between CTIP2-mediated control of lipid metabolism and alteration of EPB homeostasis, delayed wound healing, inflammatory diseases exacerbation, and cancer promotion and progression. AREAS COVERED Here we have discussed the intricate interplay of various endogenous lipids and lipoproteins accompanying skin development and associated disease processes and the possible link to CTIP2-mediated regulation of lipid metabolism. EXPERT OPINION Establishing the link between CTIP2 and lipid metabolism alterations in the context of skin morphogenesis and diverse types of skin diseases including cancer can help us identify novel targets for effective therapeutic intervention.
Collapse
Affiliation(s)
- Nilika Bhattacharya
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR, USA
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
- Department of Biochemistry and Biophysics, OSU, Corvallis, OR, USA
- Linus Pauling Science Center, OSU, Corvallis, OR, USA
- Department of Dermatology, OHSU, Portland, OR, USA
| |
Collapse
|
52
|
Murai‐Yamamura M, Garcet S, Yamamura K, Gonzalez J, Miura S, Li X, Hur H, Guttman‐Yassky E, Krueger JG. T H 2 cytokines and Staphylococcus aureus cooperatively induce atopic dermatitis-like transcriptomes. Allergy 2021; 76:3534-3537. [PMID: 34358351 DOI: 10.1111/all.15035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/29/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Mika Murai‐Yamamura
- Laboratory of Investigative Dermatology The Rockefeller University New York NY USA
| | - Sandra Garcet
- Laboratory of Investigative Dermatology The Rockefeller University New York NY USA
| | - Kazuhiko Yamamura
- Laboratory of Investigative Dermatology The Rockefeller University New York NY USA
| | - Juana Gonzalez
- Laboratory of Investigative Dermatology The Rockefeller University New York NY USA
| | - Shunsuke Miura
- Laboratory of Investigative Dermatology The Rockefeller University New York NY USA
| | - Xuan Li
- Laboratory of Investigative Dermatology The Rockefeller University New York NY USA
| | - Hong Hur
- Laboratory of Investigative Dermatology The Rockefeller University New York NY USA
| | - Emma Guttman‐Yassky
- Department of Dermatology Icahn School of Medicine at the Mount Sinai Medical Center New York NY USA
| | - James G. Krueger
- Laboratory of Investigative Dermatology The Rockefeller University New York NY USA
| |
Collapse
|
53
|
Klopot A, Baida G, Kel A, Tsoi LC, Perez White BE, Budunova I. Transcriptome analysis reveals intrinsic pro-inflammatory signaling in healthy African American skin. J Invest Dermatol 2021; 142:1360-1371.e15. [PMID: 34757068 PMCID: PMC9038646 DOI: 10.1016/j.jid.2021.09.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022]
Abstract
Differences in morphology and physiology of darkly pigmented compared to lightly pigmented skin are well recognized. There are also disparities in prevalence and clinical features for many inflammatory skin diseases including atopic dermatitis and psoriasis; however, the underlying mechanisms are largely unknown. We compared the baseline gene expression in full thickness skin biopsies from healthy individuals self-reporting as African American (AA) or White Non-Hispanic (WNH). Extensively validated RNA-Seq analysis identified 570 differentially expressed genes (DEG) in AA skin including immunoglobulins and their receptors such as FCER1G; pro-inflammatory genes such as TNFα, IL-32; EDC (epidermal differentiation cluster) and keratin genes. DEGs were functionally enriched for inflammatory responses, keratinization, cornified envelope formation. RNA-seq analysis of 3D human skin equivalents (HSE) made from AA and WNH primary keratinocytes revealed 360 DEGs (some shared with skin) which were enriched by similar functions. AA HSE appeared more responsive to TNFα pro-inflammatory effects. Finally, AA-specific DEGs in skin and HSE significantly overlapped with molecular signatures of skin in AD and psoriasis patients. Overall, these findings suggest the existence of intrinsic pro-inflammatory circuits in AA keratinocytes/skin that may account for disease disparities and will help to build a foundation for the development of targeted skin disease prevention.
Collapse
Affiliation(s)
- Anna Klopot
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Gleb Baida
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Alexander Kel
- geneXplain GmbH, Wolfenbüttel, Germany; Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Bethany E Perez White
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Irina Budunova
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
54
|
Villani AP, Pavel AB, Wu J, Fernandes M, Maari C, Saint‐Cyr Proulx E, Jack C, Glickman J, Choi S, He H, Ungar B, Estrada Y, Kameyama N, Zhang N, Gonzales J, Tardif J, Krueger JG, Bissonnette R, Guttman‐Yassky E. Vascular inflammation in moderate-to-severe atopic dermatitis is associated with enhanced Th2 response. Allergy 2021; 76:3107-3121. [PMID: 33866573 DOI: 10.1111/all.14859] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/15/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND In atopic dermatitis (AD), some studies have shown an association with increased cardiovascular disease in certain populations. However, other investigations found modest or no association. Despite conflicting results, molecular profiling studies in both AD skin and blood have demonstrated upregulation of atherosclerosis and cardiovascular risk-related markers. However, the underlying mechanisms connecting AD to vascular inflammation/atherosclerosis are unknown. In this study, we aim to determine factors associated with vascular inflammation/atherosclerosis in AD patients. METHODS We used 18-FDG PET-CT to characterize vascular inflammation in AD patients and healthy subjects. In parallel, we assessed their skin and blood immune profiles to determine AD-related immune biomarkers associated with vascular inflammation. We also assessed levels of circulating microparticles, which are known to be associated with increased cardiovascular risk. RESULTS We found significant correlations between vascular inflammation and Th2-related products in skin and blood of AD patients as well as atherosclerosis-related markers that were modulated by dupilumab. Circulating levels of endothelial microparticles were significantly higher in severe AD patients and tended to correlate with vascular inflammation assessed by PET-CT. CONCLUSION Vascular inflammation in AD is associated with enhanced Th2 response and clinical severity, which may explain cardiovascular comorbidities observed in select AD populations. Larger prospective studies are needed to further evaluate vascular inflammation and cardiovascular events and mortality in AD patients. Finally, as dupilumab treatment demonstrated significant modulation of atherosclerosis-related genes in AD patients compared to placebo, these data suggest that modulation of vascular inflammation with systemic treatment should be explored in patients with AD.
Collapse
Affiliation(s)
- Axel P. Villani
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Ana B. Pavel
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
- Department of Biomedical Engineering University of Mississippi University MS USA
| | - Jianni Wu
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
- College of Medicine State University of New York Downstate Medical Center Brooklyn New York USA
| | - Marie Fernandes
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | | | | | | | - Jacob Glickman
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Seulah Choi
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Helen He
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Benjamin Ungar
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Yeriel Estrada
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Naoya Kameyama
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Ning Zhang
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Juana Gonzales
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Jean‐Claude Tardif
- Montreal Heart Institute Faculty of Medicine Université de Montréal Montreal QC Canada
| | - James G. Krueger
- Laboratory for Investigative Dermatology The Rockefeller University New York NY USA
| | | | - Emma Guttman‐Yassky
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| |
Collapse
|
55
|
Transcriptomic Profiling of Tape-Strips From Moderate to Severe Atopic Dermatitis Patients Treated With Dupilumab. Dermatitis 2021; 32:S71-S80. [PMID: 34405829 DOI: 10.1097/der.0000000000000764] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Tape-strips are a minimally invasive approach to characterize skin biomarkers in atopic dermatitis (AD). However, they have not yet been used for tracking gene expression changes with systemic treatment. OBJECTIVE The aim of the study was to evaluate gene expression changes and therapeutic response biomarkers in AD patients before and after dupilumab (interleukin 4Rα antibody) treatment using tape-strips to obtain epidermal tissue for analysis. METHODS Lesional and nonlesional tape-stripped skin was sampled from 18 AD patients before and after dupilumab treatment and from 17 healthy subjects and analyzed by RNA-seq. RESULTS At baseline, we detected 6745 and 4859 differentially expressed genes between lesional and nonlesional skin versus normal, respectively, whereas 841 and 977 genes were differentially expressed after treatment, respectively (fold change >1.5 and false discovery rate <0.05). Tape-strips captured significant modulation with dupilumab in key AD immune (eg, C-C motif chemokine ligand 13 [CCL13], CCL17, CCL18) and barrier (eg, periplakin, FA2H) biomarkers. Changes in biomarkers (CCL20, interleukin 34, FABP7) were also significantly correlated with clinical disease improvements (Eczema Area and Severity Index; R > 0.5 or R < -0.4, P < 0.05). CONCLUSIONS This real-life study represents the first comprehensive RNA-seq molecular profiling of tape-strips from moderate to severe AD patients after dupilumab therapy. Analysis of tape strip specimens detected significant gene expression changes in key AD biomarkers with dupilumab treatment, suggesting that this approach may be useful to monitor therapeutic responses in inflammatory skin diseases.
Collapse
|
56
|
Lang CCV, Renert-Yuval Y, Del Duca E, Pavel AB, Wu J, Zhang N, Dubin C, Obi A, Chowdhoury M, Kim M, Estrada YD, Krueger JG, Kaderbhai H, Semango G, Schmid-Grendelmeier P, Brüggen MC, Masenga JE, Guttman-Yassky E. Immune and barrier characterization of atopic dermatitis skin phenotype in Tanzanian patients. Ann Allergy Asthma Immunol 2021; 127:334-341. [PMID: 33975024 PMCID: PMC11344219 DOI: 10.1016/j.anai.2021.04.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/13/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is a common disease, with particularly high prevalence found in Africa. It is increasingly recognized that patients with AD of different ethnic backgrounds have unique molecular signatures in the skin, potentially accounting for treatment response variations. Nevertheless, the skin profile of patients with AD from Africa is unknown, hindering development of new treatments targeted to this patient population. OBJECTIVE To characterize the skin profile of patients with AD from Africa. METHODS Gene expression studies, including RNA sequencing (using threshold of fold change of >2 and false discovery rate of <0.05) and real-time polymerase chain reaction, were performed on skin biopsies of Tanzanian patients with moderate-to-severe AD and controls. RESULTS Tanzanian AD skin presented robust up-regulations of multiple key mediators of both T helper 2 (TH2) (interleukin 13 [IL-13], IL-10, IL-4R, CCL13,CCL17,CCL18,CCL26) and TH22 (IL22, S100As) pathways. Markers related to TH17 and IL-23 (IL-17A, IL-23A, IL-12, PI3, DEFB4B) and TH1 (interferon gamma, CXCL9,CXCL10,CXCL11) were also significantly overexpressed in AD tissues (FDR<.05), albeit to a lesser extent. IL-36 isoforms revealed substantial up-regulations in African skin. The barrier fingerprint of Tanzanian AD revealed no suppression of hallmark epidermal barrier differentiation genes, such as filaggrin, loricrin, and periplakin, with robust attenuation of lipid metabolism genes (ie, AWAT1). CONCLUSION The skin phenotype of Tanzanian patients with AD is consistent with that of African Americans, exhibiting dominant TH2 and TH22 skewing, minimal dysregulation of terminal differentiation, and even broader attenuation of lipid metabolism-related products. These data highlight the unique characteristic of AD in Black individuals and the need to develop unique treatments targeting patients with AD from these underrepresented populations.
Collapse
Affiliation(s)
- Claudia C V Lang
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - Yael Renert-Yuval
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York; Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Ester Del Duca
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Dermatology, University of Magna Graecia, Catanzaro, Italy
| | - Ana B Pavel
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Biomedical Engineering, University of Mississippi, Oxford, Mississippi
| | - Jianni Wu
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ning Zhang
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Celina Dubin
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ashley Obi
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mashkura Chowdhoury
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Madeline Kim
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yeriel D Estrada
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Hashim Kaderbhai
- Department of Dermatology, M.P. Shah Hospital, Nairobi, Kenya; Department of Dermatology, Regional Dermatology Training Center, Moshi, Tanzania
| | - George Semango
- Department of Dermatology, Regional Dermatology Training Center, Moshi, Tanzania
| | | | - Marie-Charlotte Brüggen
- Department of Dermatology, University Hospital Zürich, Zürich, Switzerland; Hochgebirgsklinik Davos, Davos, Switzerland
| | - John E Masenga
- Department of Dermatology, Regional Dermatology Training Center, Moshi, Tanzania
| | - Emma Guttman-Yassky
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
57
|
Konger RL, Derr-Yellin E, Zimmers TA, Katona T, Xuei X, Liu Y, Zhou HM, Simpson ER, Turner MJ. Epidermal PPARγ Is a Key Homeostatic Regulator of Cutaneous Inflammation and Barrier Function in Mouse Skin. Int J Mol Sci 2021; 22:ijms22168634. [PMID: 34445339 PMCID: PMC8395473 DOI: 10.3390/ijms22168634] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 01/10/2023] Open
Abstract
Both agonist studies and loss-of-function models indicate that PPARγ plays an important role in cutaneous biology. Since PPARγ has a high level of basal activity, we hypothesized that epidermal PPARγ would regulate normal homeostatic processes within the epidermis. In this current study, we performed mRNA sequencing and differential expression analysis of epidermal scrapings from knockout mice and wildtype littermates. Pparg-/-epi mice exhibited a 1.5-fold or greater change in the expression of 11.8% of 14,482 identified transcripts. Up-regulated transcripts included those for a large number of cytokines/chemokines and their receptors, as well as genes associated with inflammasome activation and keratinization. Several of the most dramatically up-regulated pro-inflammatory genes in Pparg-/-epi mouse skin included Igfl3, 2610528A11Rik, and Il1f6. RT-PCR was performed from RNA obtained from non-lesional full-thickness skin and verified a marked increase in these transcripts, as well as transcripts for Igflr1, which encodes the receptor for Igfl3, and the 2610528A11Rik receptor (Gpr15). Transcripts for Il4 were detected in Pparg-/-epi mouse skin, but transcripts for Il17 and Il22 were not detected. Down-regulated transcripts included sebaceous gland markers and a number of genes associated with lipid barrier formation. The change in these transcripts correlates with an asebia phenotype, increased transepidermal water loss, alopecia, dandruff, and the appearance of spontaneous inflammatory skin lesions. Histologically, non-lesional skin showed hyperkeratosis, while inflammatory lesions were characterized by dermal inflammation and epidermal acanthosis, spongiosis, and parakeratosis. In conclusion, loss of epidermal Pparg alters a substantial set of genes that are associated with cutaneous inflammation, keratinization, and sebaceous gland function. The data indicate that epidermal PPARγ plays an important role in homeostatic epidermal function, particularly epidermal differentiation, barrier function, sebaceous gland development and function, and inflammatory signaling.
Collapse
Affiliation(s)
- Raymond L. Konger
- Department of Pathology & Laboratory Medicine, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA; (E.D.-Y.); (T.K.)
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- The Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (T.A.Z.); (Y.L.)
- Correspondence: ; Tel.: +1-317-274-4154
| | - Ethel Derr-Yellin
- Department of Pathology & Laboratory Medicine, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA; (E.D.-Y.); (T.K.)
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Teresa A. Zimmers
- The Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (T.A.Z.); (Y.L.)
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Terrence Katona
- Department of Pathology & Laboratory Medicine, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA; (E.D.-Y.); (T.K.)
| | - Xiaoling Xuei
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Yunlong Liu
- The Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (T.A.Z.); (Y.L.)
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
| | - Hong-Ming Zhou
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.-M.Z.); (M.J.T.)
| | - Ed Ronald Simpson
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.-M.Z.); (M.J.T.)
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Matthew J. Turner
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.-M.Z.); (M.J.T.)
- Department of Dermatology, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
58
|
Mikhaylov D, Del Duca E, Guttman-Yassky E. Proteomic signatures of inflammatory skin diseases: a focus on atopic dermatitis. Expert Rev Proteomics 2021; 18:345-361. [PMID: 34033497 DOI: 10.1080/14789450.2021.1935247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Atopic dermatitis (AD) is a chronic inflammatory skin condition characterized by cutaneous and systemic inflammation and barrier abnormalities. Over the past few decades, proteomic studies have been increasingly applied to AD research to compliment transcriptomic evaluations. Proteomic analyses helped identify new biomarkers of AD, allowing investigation of both the cutaneous AD profile and the systemic inflammation associated with the disease.Areas covered: This review discusses key studies that utilized various proteomic technologies to analyze AD skin and/or blood, which facilitated discovery of biomarkers related to pathogenesis, disease severity, systemic inflammation, and therapeutic response. Moreover, this review summarizes proteomic studies that helped define various AD endotypes/phenotypes. A literature search was conducted by querying Scopus, Google Scholar, PubMed/Medline, and Clinicaltrials.gov up to January 2021.Expert opinion: Use of proteomics in AD has allowed for identification of novel AD-related protein biomarkers. This approach continues to evolve and is becoming increasingly common for the study of AD, in conjunction with other -omics platforms, as proteomics shifts to quicker and more sensitive methods for detection of potential protein biomarkers. Although many biomarkers have been identified thus far, future larger studies are necessary to further correlate these markers with clinical parameters.
Collapse
Affiliation(s)
- Daniela Mikhaylov
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ester Del Duca
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Dermatology, University of Magna Graecia, Catanzaro, Italy
| | - Emma Guttman-Yassky
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
59
|
Guttman-Yassky E, Diaz A, Pavel AB, Fernandes M, Lefferdink R, Erickson T, Canter T, Rangel S, Peng X, Li R, Estrada Y, Xu H, Krueger JG, Paller AS. Use of Tape Strips to Detect Immune and Barrier Abnormalities in the Skin of Children With Early-Onset Atopic Dermatitis. JAMA Dermatol 2021; 155:1358-1370. [PMID: 31596431 DOI: 10.1001/jamadermatol.2019.2983] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Importance Molecular profiling of skin biopsies is the criterion standard for evaluating the cutaneous atopic dermatitis (AD) phenotype. However, skin biopsies are not always feasible in children. A reproducible minimally invasive approach that can track cutaneous disease in pediatric longitudinal studies or clinical trials is lacking. Objective To assess a minimally invasive approach using tape strips to identify skin biomarkers that may serve as a surrogate to biomarkers identified using whole-tissue biopsies. Design, Setting, and Participants This cross-sectional study of 51 children younger than 5 years recruited children with moderate to severe AD and children without AD from the dermatology outpatient clinics at a children's hospital. Sixteen tape strips were serially collected from the nonlesional and lesional skin of 21 children who had AD and were less than 6 months from disease initiation and from the normal skin of 30 children who did not have AD between January 22, 2016, and April 20, 2018. Main Outcomes and Measures Gene and protein expression were evaluated using quantitative real-time polymerase chain reaction and immunohistochemistry. Results A total of 51 children younger than 5 years were included in the study; 21 children had moderate to severe AD with less than 6 months of disease duration, and 30 children did not have AD. Of the 21 children with AD, the mean (SD) age was 1.7 (1.7) years, and most were male (15 [71.4%] and white (15 [71.4%]). Of the 30 children without AD, the mean (SD) age was 1.8 (2.0) years, and most were female (20 [66.7%]) and white (22 [73.3%]). Seventy-seven of 79 evaluated immune and barrier gene products were detected (gene detection rate, 97%) in 70 of 71 tape strips (sample detection rate, 99%), with 53 of 79 markers differentiating between children with lesional and/or nonlesional AD from children without AD. Many cellular markers of T cells (CD3), AD-related dendritic cells (Fc ε RI and OX40 ligand receptors), and key inflammatory (matrix metallopeptidase 12), innate (interleukin 8 [IL-8] and IL-6), helper T cell 2 (TH2; IL-4, IL-13, and chemokines CCL17 and CCL26), and TH17/TH22 (IL-19, IL-36G, and S100A proteins) genes were significantly increased in lesional and nonlesional AD compared with tape strips from normal skin. For example, IL-4 mean (SE) for lesional was -15.2 (0.91) and normal was -19.5 (0.48); P < .001. Parallel decreases occurred in epidermal barrier gene products (FLG, CLDN23, and FA2H) and negative immune regulators (IL-34 and IL-37). For example, the decrease for FLG lesional was mean (SE) -2.9 (0.42) and for normal was 2.2 (0.45); P < .001. Associations were found between disease severity or transepidermal water loss and TH2 (IL-33 and IL-4R) and TH17/TH22 (IL-36G and S100As) products in lesional and nonlesional AD skin (evaluated using the SCORing Atopic Dermatitis, Eczema Area and Severity Index, and Pruritus Atopic Dermatitis Quickscore tools). Conclusions and Relevance In this study, tape strips provide a minimally invasive alternative for serially evaluating AD-associated cutaneous biomarkers and may prove useful for tracking pediatric AD therapeutic response and predicting future course and comorbidities.
Collapse
Affiliation(s)
- Emma Guttman-Yassky
- Department of Dermatology and Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York
| | - Aisleen Diaz
- Department of Dermatology and Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York.,School of Medicine, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Ana B Pavel
- Department of Dermatology and Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York
| | - Marie Fernandes
- Department of Dermatology and Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York
| | - Rachel Lefferdink
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Taylor Erickson
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Talia Canter
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Stephanie Rangel
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiangyu Peng
- Department of Dermatology and Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York
| | - Randall Li
- Department of Dermatology and Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York
| | - Yeriel Estrada
- Department of Dermatology and Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York
| | - Hui Xu
- Department of Dermatology and Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Amy S Paller
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
60
|
Ottman N, Barrientos‐Somarribas M, Fyhrquist N, Alexander H, Wisgrill L, Olah P, Tsoka S, Greco D, Levi‐Schaffer F, Soumelis V, Schröder JM, Kere J, Nestle FO, Barker J, Ranki A, Lauerma A, Homey B, Andersson B, Alenius H. Microbial and transcriptional differences elucidate atopic dermatitis heterogeneity across skin sites. Allergy 2021; 76:1173-1187. [PMID: 33001460 PMCID: PMC8246754 DOI: 10.1111/all.14606] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/03/2020] [Accepted: 09/06/2020] [Indexed: 12/29/2022]
Abstract
It is well established that different sites in healthy human skin are colonized by distinct microbial communities due to different physiological conditions. However, few studies have explored microbial heterogeneity between skin sites in diseased skin, such as atopic dermatitis (AD) lesions. To address this issue, we carried out deep analysis of the microbiome and transcriptome in the skin of a large cohort of AD patients and healthy volunteers, comparing two physiologically different sites: upper back and posterior thigh. Microbiome samples and biopsies were obtained from both lesional and nonlesional skin to identify changes related to the disease process. Transcriptome analysis revealed distinct disease-related gene expression profiles depending on anatomical location, with keratinization dominating the transcriptomic signatures in posterior thigh, and lipid metabolism in the upper back. Moreover, we show that relative abundance of Staphylococcus aureus is associated with disease severity in the posterior thigh, but not in the upper back. Our results suggest that AD may select for similar microbes in different anatomical locations-an "AD-like microbiome," but distinct microbial dynamics can still be observed when comparing posterior thigh to upper back. This study highlights the importance of considering the variability across skin sites when studying the development of skin inflammation.
Collapse
Affiliation(s)
- Noora Ottman
- Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
| | | | - Nanna Fyhrquist
- Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
- Human Microbiome Research Program University of Helsinki Helsinki Finland
| | - Helen Alexander
- St John's Institute of Dermatology Guy's and St Thomas' NHS Foundation Trust and King's College London London UK
| | - Lukas Wisgrill
- Division of Neonatology Pediatric Intensive Care and Neuropediatrics Medical University of Vienna Vienna Austria
| | - Peter Olah
- Department of Dermatology University Hospital Duesseldorf Duesseldorf Germany
- Department of Dermatology, Venereology and Oncodermatology University of Pécs Pécs Hungary
| | - Sophia Tsoka
- Department of Informatics Faculty of Natural and Mathematical Sciences King’s College London London UK
| | - Dario Greco
- Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
- Institute of Biomedical Technology University of Tampere Tampere Finland
- Institute of Biotechnology University of Helsinki Helsinki Finland
| | - Francesca Levi‐Schaffer
- Pharmacology Unit School of Pharmacy Faculty of Medicine The Institute for Drug Research The Hebrew University of Jerusalem Jerusalem Israel
| | | | - Jens M. Schröder
- Department of Dermatology University Hospital Schleswig‐Holstein Kiel Germany
| | - Juha Kere
- Department of Biosciences and Nutrition Karolinska Institutet Stockholm Sweden
- School of Basic and Medical Biosciences King’s College London London UK
| | - Frank O. Nestle
- Cutaneous Medicine Unit St. John’s Institute of Dermatology and Biomedical Research Centre Faculty of Life Sciences and Medicine King’s College London London UK
| | - Jonathan Barker
- St John’s Institute of Dermatology Division of Genetics and Molecular Medicine Faculty of Life Sciences and Medicine Kings College London London UK
| | - Annamari Ranki
- Department of Dermatology, Allergology and Venereology Inflammation Centre University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Antti Lauerma
- Department of Dermatology, Allergology and Venereology Inflammation Centre University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Bernhard Homey
- Department of Dermatology University Hospital Duesseldorf Duesseldorf Germany
| | - Björn Andersson
- Department of Cell and Molecular Biology Karolinska Institutet Stockholm Sweden
| | - Harri Alenius
- Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
- Human Microbiome Research Program University of Helsinki Helsinki Finland
| |
Collapse
|
61
|
Kim YW, Ko EA, Jung SC, Lee D, Seo Y, Kim S, Kim JH, Bang H, Zhou T, Ko JH. Transcriptomic insight into the translational value of two murine models in human atopic dermatitis. Sci Rep 2021; 11:6616. [PMID: 33758305 PMCID: PMC7988112 DOI: 10.1038/s41598-021-86049-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
This study sought to develop a novel diagnostic tool for atopic dermatitis (AD). Mouse transcriptome data were obtained via RNA-sequencing of dorsal skin tissues of CBA/J mice affected with contact hypersensitivity (induced by treatment with 1-chloro-2,4-dinitrobenzene) or brush stimulation-induced AD-like skin condition. Human transcriptome data were collected from German, Swedish, and American cohorts of AD patients from the Gene Expression Omnibus database. edgeR and SAM algorithms were used to analyze differentially expressed murine and human genes, respectively. The FAIME algorithm was then employed to assign pathway scores based on KEGG pathway database annotations. Numerous genes and pathways demonstrated similar dysregulation patterns in both the murine models and human AD. Upon integrating transcriptome information from both murine and human data, we identified 36 commonly dysregulated differentially expressed genes, which were designated as a 36-gene signature. A severity score (AD index) was applied to each human sample to assess the predictive power of the 36-gene AD signature. The diagnostic power and predictive accuracy of this signature were demonstrated for both AD severity and treatment outcomes in patients with AD. This genetic signature is expected to improve both AD diagnosis and targeted preclinical research.
Collapse
Affiliation(s)
- Young-Won Kim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, 06974, Korea
| | - Eun-A Ko
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 63243, Korea
| | - Sung-Cherl Jung
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 63243, Korea
| | - Donghee Lee
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, 06974, Korea
| | - Yelim Seo
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, 06974, Korea
| | - Seongtae Kim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, 06974, Korea
| | - Jung-Ha Kim
- Department of Family Medicine, College of Medicine, Chung-Ang University Hospital, Seoul, 06973, Korea
| | - Hyoweon Bang
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, 06974, Korea
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA.
| | - Jae-Hong Ko
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, 06974, Korea.
| |
Collapse
|
62
|
Martin P, Goldstein JD, Mermoud L, Diaz-Barreiro A, Palmer G. IL-1 Family Antagonists in Mouse and Human Skin Inflammation. Front Immunol 2021; 12:652846. [PMID: 33796114 PMCID: PMC8009184 DOI: 10.3389/fimmu.2021.652846] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 12/23/2022] Open
Abstract
Interleukin (IL)-1 family cytokines initiate inflammatory responses, and shape innate and adaptive immunity. They play important roles in host defense, but excessive immune activation can also lead to the development of chronic inflammatory diseases. Dysregulated IL-1 family signaling is observed in a variety of skin disorders. In particular, IL-1 family cytokines have been linked to the pathogenesis of psoriasis and atopic dermatitis. The biological activity of pro-inflammatory IL-1 family agonists is controlled by the natural receptor antagonists IL-1Ra and IL-36Ra, as well as by the regulatory cytokines IL-37 and IL-38. These four anti-inflammatory IL-1 family members are constitutively and highly expressed at steady state in the epidermis, where keratinocytes are a major producing cell type. In this review, we provide an overview of the current knowledge concerning their regulatory roles in skin biology and inflammation and their therapeutic potential in human inflammatory skin diseases. We further highlight some common misunderstandings and less well-known observations, which persist in the field despite recent extensive interest for these cytokines.
Collapse
Affiliation(s)
- Praxedis Martin
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jérémie D. Goldstein
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Loïc Mermoud
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alejandro Diaz-Barreiro
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
63
|
Renert-Yuval Y, Del Duca E, Pavel AB, Fang M, Lefferdink R, Wu J, Diaz A, Estrada YD, Canter T, Zhang N, Wagner A, Chamlin S, Krueger JG, Guttman-Yassky E, Paller AS. The molecular features of normal and atopic dermatitis skin in infants, children, adolescents, and adults. J Allergy Clin Immunol 2021; 148:148-163. [PMID: 33453290 DOI: 10.1016/j.jaci.2021.01.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/01/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Although atopic dermatitis (AD) often presents in infancy and persists into adulthood, comparative characterization of AD skin among different pediatric age groups is lacking. OBJECTIVE We sought to define skin biopsy profiles of lesional and nonlesional AD across different age groups (0-5-year-old infants with disease duration <6 months, 6-11-year-old children, 12-17-year-old adolescents, ≥18-year-old adults) versus age-appropriate controls. METHODS We performed gene expression analyses by RNA-sequencing and real-time PCR (RT-PCR) and protein expression analysis using immunohistochemistry. RESULTS TH2/TH22 skewing, including IL-13, CCL17/thymus and activation-regulated chemokine, IL-22, and S100As, characterized the common AD signature, with a global pathway-level enrichment across all ages. Nevertheless, specific cytokines varied widely. For example, IL-33, IL-1RL1/IL-33R, and IL-9, often associated with early atopic sensitization, showed greatest upregulations in infants. TH17 inflammation presented a 2-peak curve, with highest increases in infants (including IL-17A and IL-17F), followed by adults. TH1 polarization was uniquely detected in adults, even when compared with adolescents, with significant upregulation in adults of IFN-γ and CXCL9/CXCL10/CXCL11. Although all AD age groups had barrier abnormalities, only adults had significant decreases in filaggrin expression. Despite the short duration of the disease, infant AD presented robust downregulations of multiple barrier-related genes in both lesional and nonlesional skin. Clinical severity scores significantly correlated with TH2/TH22-related markers in all pediatric age groups. CONCLUSIONS The shared signature of AD across ages is TH2/TH22-skewed, yet differential expression of specific TH2/TH22-related genes, other TH pathways, and barrier-related genes portray heterogenetic, age-specific molecular fingerprints.
Collapse
Affiliation(s)
- Yael Renert-Yuval
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ester Del Duca
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Dermatology, University Magna Graecia, Catanzaro, Italy
| | - Ana B Pavel
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Biomedical Engineering, University of Mississippi, Oxford, Miss
| | - Milie Fang
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Rachel Lefferdink
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Jianni Wu
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Aisleen Diaz
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yeriel D Estrada
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Talia Canter
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Ning Zhang
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Annette Wagner
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Sarah Chamlin
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| | - Emma Guttman-Yassky
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Amy S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| |
Collapse
|
64
|
Tape strips detect distinct immune and barrier profiles in atopic dermatitis and psoriasis. J Allergy Clin Immunol 2021; 147:199-212. [DOI: 10.1016/j.jaci.2020.05.048] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/21/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022]
|
65
|
Lossius AH, Berents TL, Saetre F, Nilsen HR, Bradley M, Asad S, Haraldsen G, Sundnes O, Holm JØ. Early transcriptional changes after UVB treatment in atopic dermatitis include inverse regulation of IL-36γ and IL-37. Exp Dermatol 2020; 30:249-261. [PMID: 33067891 DOI: 10.1111/exd.14217] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/26/2020] [Accepted: 10/12/2020] [Indexed: 12/22/2022]
Abstract
Phototherapy with narrow-band Ultraviolet B (nb-UVB) is a major therapeutic option in atopic dermatitis (AD), yet knowledge of the early molecular responses to this treatment is lacking. The objective of this study was to map the early transcriptional changes in AD skin in response to nb-UVB treatment. Adult patients (n = 16) with AD were included in the study and scored with validated scoring tools. AD skin was irradiated with local nb-UVB on day 0, 2 and 4. Skin biopsies were taken before and after treatment (day 0 and 7) and analysed for genome-wide modulation of transcription. When examining the early response after three local UVB treatments, gene expression analysis revealed 77 significantly modulated transcripts (30 down- and 47 upregulated). Among them were transcripts related to the inflammatory response, melanin synthesis, keratinization and epidermal structure. Interestingly, the pro-inflammatory cytokine IL-36γ was reduced after treatment, while the anti-inflammatory cytokine IL-37 increased after treatment with nb-UVB. There was also a modulation of several other mediators involved in inflammation, among them defensins and S100 proteins. This is the first study of early transcriptomic changes in AD skin in response to nb-UVB. We reveal robust modulation of a small group of inflammatory and anti-inflammatory targets, including the IL-1 family members IL36γ and IL-37, which is evident before any detectable changes in skin morphology or immune cell infiltrates. These findings provide important clues to the molecular mechanisms behind the treatment response and shed light on new potential treatment targets.
Collapse
Affiliation(s)
- Astrid H Lossius
- Institute of clinical medicine, University of Oslo, Oslo, Norway.,Department of Dermatology, Oslo University Hospital, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Teresa L Berents
- Department of Dermatology, Oslo University Hospital, Oslo, Norway
| | - Frank Saetre
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Hogne R Nilsen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Maria Bradley
- Division of Dermatology and Venereology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Samina Asad
- Division of Dermatology and Venereology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Guttorm Haraldsen
- Institute of clinical medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Olav Sundnes
- Department of Dermatology, Oslo University Hospital, Oslo, Norway
| | - Jan-Øivind Holm
- Institute of clinical medicine, University of Oslo, Oslo, Norway.,Department of Dermatology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
66
|
Rojahn TB, Vorstandlechner V, Krausgruber T, Bauer WM, Alkon N, Bangert C, Thaler FM, Sadeghyar F, Fortelny N, Gernedl V, Rindler K, Elbe-Bürger A, Bock C, Mildner M, Brunner PM. Single-cell transcriptomics combined with interstitial fluid proteomics defines cell type-specific immune regulation in atopic dermatitis. J Allergy Clin Immunol 2020; 146:1056-1069. [PMID: 32344053 DOI: 10.1016/j.jaci.2020.03.041] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/10/2020] [Accepted: 03/27/2020] [Indexed: 02/09/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is the most common chronic inflammatory skin disease, but its complex pathogenesis is only insufficiently understood, resulting in still limited treatment options. OBJECTIVE We sought to characterize AD on both transcriptomic and proteomic levels in humans. METHODS We used skin suction blistering, a painless and nonscarring procedure that can simultaneously sample skin cells and interstitial fluid. We then compared results with conventional biopsies. RESULTS Suction blistering captured epidermal and most immune cells equally well as biopsies, except for mast cells and nonmigratory CD163+ macrophages that were only present in biopsy isolates. Using single-cell RNA sequencing, we found comparable transcriptional profiles of key inflammatory pathways between blister and biopsy AD, but suction blistering was superior in cell-specific resolution for high-abundance transcripts (KRT1/KRT10, KRT16/KRT6A, S100A8/S100A9), which showed some background signals in biopsy isolates. Compared with healthy controls, we found characteristic upregulation of AD-typical cytokines such as IL13 and IL22 in Th2 and Th22 cells, respectively, but we also discovered these mediators in proliferating T cells and natural killer T cells, that also expressed the antimicrobial cytokine IL26. Overall, not T cells, but myeloid cells were most strongly enriched in AD, and we found dendritic cell (CLEC7A, amphiregulin/AREG, EREG) and macrophage products (CCL13) among the top upregulated proteins in AD blister fluid proteomic analyses. CONCLUSION These data show that by using cutting-edge technology, suction blistering offers several advantages over conventional biopsies, including better transcriptomic resolution of skin cells, combined with proteomic information from interstitial fluid, unraveling novel inflammatory players that shape the cellular and proteomic microenvironment of AD.
Collapse
Affiliation(s)
- Thomas B Rojahn
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Vera Vorstandlechner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Department of Surgery, Research Laboratory for Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Wolfgang M Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Natalia Alkon
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christine Bangert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Felix M Thaler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Farzaneh Sadeghyar
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Victoria Gernedl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Katharina Rindler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Patrick M Brunner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
67
|
Uluçkan Ö, Jiménez M, Roediger B, Schnabl J, Díez-Córdova LT, Troulé K, Weninger W, Wagner EF. Cutaneous Immune Cell-Microbiota Interactions Are Controlled by Epidermal JunB/AP-1. Cell Rep 2020; 29:844-859.e3. [PMID: 31644908 PMCID: PMC6856727 DOI: 10.1016/j.celrep.2019.09.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 07/01/2019] [Accepted: 09/13/2019] [Indexed: 12/16/2022] Open
Abstract
Atopic dermatitis (AD) is a multi-factorial skin disease with a complex inflammatory signature including type 2 and type 17 activation. Although colonization by S. aureus is common in AD, the mechanisms rendering an organism prone to dysbiosis, and the role of IL-17A in the control of S. aureus-induced skin inflammation, are not well understood. Here, we show several pathological aspects of AD, including type 2/type 17 immune responses, elevated IgE, barrier dysfunction, pruritus, and importantly, spontaneous S. aureus colonization in JunBΔep mice, with a large transcriptomic overlap with AD. Additionally, using Rag1−/− mice, we demonstrate that adaptive immune cells are necessary for protection against S. aureus colonization. Prophylactic antibiotics, but not antibiotics after established dysbiosis, reduce IL-17A expression and skin inflammation, examined using Il17a-eGFP reporter mice. Mechanistically, keratinocytes lacking JunB exhibit higher MyD88 levels in vitro and in vivo, previously shown to regulate S. aureus colonization. In conclusion, our data identify JunB as an upstream regulator of microbiota-immune cell interactions and characterize the IL-17A response upon spontaneous dysbiosis. JunBΔep mice display several defining features of atopic dermatitis Skin of JunBΔep mice are colonized by human-derived S. aureus Adaptive immune system is necessary for protection against S. aureus JunB is an upstream regulator of the microbiota-immune cell interactions
Collapse
Affiliation(s)
- Özge Uluçkan
- Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain.
| | - Maria Jiménez
- Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Ben Roediger
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Jakob Schnabl
- Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Lucía T Díez-Córdova
- Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Kevin Troulé
- Bioinformatics Unit, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Wolfgang Weninger
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia; Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Erwin F Wagner
- Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; Department of Dermatology and Department of Laboratory Medicine, Medical University of Vienna, Lazarettgasse 14a, 1090 Vienna, Austria.
| |
Collapse
|
68
|
Guerrero-Aspizua S, Carretero M, Conti CJ, Del Río M. The importance of immunity in the development of reliable animal models for psoriasis and atopic dermatitis. Immunol Cell Biol 2020; 98:626-638. [PMID: 32479655 DOI: 10.1111/imcb.12365] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 01/17/2023]
Abstract
Psoriasis (PS) and atopic dermatitis (AD) are common inflammatory skin diseases characterized by an imbalance in specific T-cell subsets, resulting in a specific cytokine profile in patients. Obtaining models closely resembling both pathologies along with a relevant clinical impact is crucial for the development of new therapies because of the high prevalence of these diseases. Single-gene mouse models developed until now do not fully reflect the complexity of these disorders, in part not only because of inherent differences between mice and humans but also because of the multifactorial nature of these pathologies. The skin-humanized mouse model developed by our group, based on a tissue engineering approach, has been used to test therapeutic strategies, although this methodology is still technically challenging and not widely available. The skin-humanized mouse models for PS and AD reproduce human skin phenotypes, providing valuable tools for drug development and testing in the preclinical setting. The tissue engineering approach allows the development of personalized medicine, covering the broad genotypic spectrum of these pathologies. This review highlights the main differences between available murine models focusing on the tissue-specific immunity of PS and AD. We discuss their contribution to unravel the complex pathophysiology of these diseases and to translate this knowledge into more accurate therapies.
Collapse
Affiliation(s)
- Sara Guerrero-Aspizua
- Department of Bioengineering, Universidad Carlos III de Madrid, Leganés, 28911, Spain.,Hospital Fundación Jiménez Díaz e Instituto de Investigación FJD, Madrid, 28040, Spain.,Epithelial Biomedicine Division, CIEMAT, Madrid, 28040, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, U714, Spain
| | - Marta Carretero
- Hospital Fundación Jiménez Díaz e Instituto de Investigación FJD, Madrid, 28040, Spain.,Epithelial Biomedicine Division, CIEMAT, Madrid, 28040, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, U714, Spain
| | - Claudio J Conti
- Department of Bioengineering, Universidad Carlos III de Madrid, Leganés, 28911, Spain.,Hospital Fundación Jiménez Díaz e Instituto de Investigación FJD, Madrid, 28040, Spain
| | - Marcela Del Río
- Department of Bioengineering, Universidad Carlos III de Madrid, Leganés, 28911, Spain.,Hospital Fundación Jiménez Díaz e Instituto de Investigación FJD, Madrid, 28040, Spain.,Epithelial Biomedicine Division, CIEMAT, Madrid, 28040, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, U714, Spain
| |
Collapse
|
69
|
Möbus L, Rodriguez E, Harder I, Stölzl D, Boraczynski N, Gerdes S, Kleinheinz A, Abraham S, Heratizadeh A, Handrick C, Haufe E, Werfel T, Schmitt J, Weidinger S. Atopic dermatitis displays stable and dynamic skin transcriptome signatures. J Allergy Clin Immunol 2020; 147:213-223. [PMID: 32615169 DOI: 10.1016/j.jaci.2020.06.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Skin transcriptome studies in atopic dermatitis (AD) showed broad dysregulation as well as "improvement" under therapy. These observations were mainly made in trials and based on microarray data. OBJECTIVES Our aim was to explore the skin transcriptome and the impact of systemic treatment in patients of the TREATgermany registry. METHODS Biopsy specimens from 59 patients with moderate-to-severe AD before and 30 patients 12 weeks after start of systemic treatment (dupilumab [n = 22] or cyclosporine [n = 8]) and from 31 healthy controls were subjected to mRNA sequencing. Differential expression, pathway enrichment, correlation, and coexpression network analysis were conducted. RESULTS Both lesional and nonlesional skin showed a stable "core" signature characterized by disturbed epidermal differentiation and activation of IL-31/IL-1 signaling. A second dynamic signature showed progressive enrichment for type 2 inflammation, TH17 signaling, and natural killer cell function. Markers correlated with disease activity have functions in epidermal barrier properties and immune modulation. IL4RA was among the top 3 central dysregulated genes. Cyclosporine led to a more pronounced global transcriptome reversion and normalized TH17 cell/IL23 signaling, whereas dupilumab led to a stronger increase in level of epidermal differentiation markers. Both treatments strongly decreased levels of type 2 markers, but overall the residual profile was still profoundly different from that of healthy skin. Lower levels of IL4RA and IL13 and high IL36A expression were related to a stronger clinical response to dupilumab. CONCLUSION The AD core signature is characterized by dysregulation of genes related to keratinocyte differentiation and itch signaling. A dynamic signature reflects progressive immune responses dominated by type 2 cytokines with an additional role of TH17 and natural killer cell signaling.
Collapse
Affiliation(s)
- Lena Möbus
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Elke Rodriguez
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Inken Harder
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Dora Stölzl
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Nicole Boraczynski
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Sascha Gerdes
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | - Susanne Abraham
- University Allergy Centre, Carl Gustav Carus University Medical Centre, TU Dresden, Dresden, Germany
| | - Annice Heratizadeh
- Division of Immunodermatology and Allergy Research, Department of Dermatology, Allergy, and Venereology, Hannover Medical School, Hannover, Germany
| | - Christiane Handrick
- Practice for Dermatology and Venereology, Christiane Handrick, MD, Berlin, Germany
| | - Eva Haufe
- Center for Evidence-Based Health Care, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology, Allergy, and Venereology, Hannover Medical School, Hannover, Germany
| | - Jochen Schmitt
- Center for Evidence-Based Health Care, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| |
Collapse
|
70
|
Nomura T, Wu J, Kabashima K, Guttman-Yassky E. Endophenotypic Variations of Atopic Dermatitis by Age, Race, and Ethnicity. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 8:1840-1852. [DOI: 10.1016/j.jaip.2020.02.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/11/2020] [Accepted: 02/25/2020] [Indexed: 12/21/2022]
|
71
|
Guttman-Yassky E, Gallo RL, Pavel AB, Nakatsuji T, Li R, Zhang N, Messersmith E, Maeda-Chubachi T. A Nitric Oxide-Releasing Topical Medication as a Potential Treatment Option for Atopic Dermatitis through Antimicrobial and Anti-Inflammatory Activity. J Invest Dermatol 2020; 140:2531-2535.e2. [PMID: 32428543 DOI: 10.1016/j.jid.2020.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/26/2020] [Accepted: 04/26/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | - Richard L Gallo
- Department of Dermatology, UC San Diego School of Medicine, San Diego, California, USA
| | - Ana B Pavel
- Department of Dermatology, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | - Teruaki Nakatsuji
- Department of Dermatology, UC San Diego School of Medicine, San Diego, California, USA
| | - Randall Li
- Department of Dermatology, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | - Ning Zhang
- Department of Dermatology, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | | | | |
Collapse
|
72
|
Glickman JW, Dubin C, Renert-Yuval Y, Dahabreh D, Kimmel GW, Auyeung K, Estrada YD, Singer G, Krueger JG, Pavel AB, Guttman-Yassky E. Cross-sectional study of blood biomarkers of patients with moderate to severe alopecia areata reveals systemic immune and cardiovascular biomarker dysregulation. J Am Acad Dermatol 2020; 84:370-380. [PMID: 32376430 DOI: 10.1016/j.jaad.2020.04.138] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/23/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Although there is increased understanding of the alopecia areata (AA) pathogenesis based on studies in scalp tissues, little is known about its systemic profile. OBJECTIVE To evaluate the blood proteomic signature of AA and determine biomarkers associated with increased disease severity. METHODS In a cross-sectional study, we assessed 350 inflammatory and cardiovascular proteins using OLINK high-throughput proteomics in patients with moderate to severe AA (n = 35), as compared with healthy individuals (n = 36), patients with moderate to severe psoriasis (n = 19), and those with atopic dermatitis (n = 49). RESULTS Seventy-four proteins were significantly differentially expressed between AA and control individuals (false discovery rate, <.05) including innate immunity (interleukin [IL] 6/IL-8), T helper (Th) type 1 (interferon [IFN] γ/CXCL9/CXCL10/CXCL11), Th2 (CCL13/CCL17/CCL7), Th17 (CCL20/PI3/S100A12), and cardiovascular-risk proteins (OLR1/OSM/MPO/PRTN3). Eighty-six biomarkers correlated with AA clinical severity (P < .05), including Th1/Th2, and cardiovascular/atherosclerosis-related proteins, including SELP/PGLYRP1/MPO/IL-18/OSM (P < .05). Patients with AA totalis/universalis showed the highest systemic inflammatory tone, including cardiovascular risk biomarkers, compared to control individuals and even to patients with atopic dermatitis and those with psoriasis. The AA profile showed some Th1/Th2 differences in the setting of concomitant atopy. LIMITATIONS Our analysis was limited to 350 proteins. CONCLUSION This study defined the abnormalities of moderate to severe AA and associated circulatory biomarkers. It shows that AA has systemic immune, cardiovascular, and atherosclerosis biomarker dysregulation, suggesting the need for systemic treatment approaches.
Collapse
Affiliation(s)
- Jacob W Glickman
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Celina Dubin
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yael Renert-Yuval
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Dante Dahabreh
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Grace W Kimmel
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kelsey Auyeung
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yeriel D Estrada
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Giselle Singer
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Ana B Pavel
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Emma Guttman-Yassky
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
73
|
Pavel AB, Zhou L, Diaz A, Ungar B, Dan J, He H, Estrada YD, Xu H, Fernandes M, Renert-Yuval Y, Krueger JG, Guttman-Yassky E. The proteomic skin profile of moderate-to-severe atopic dermatitis patients shows an inflammatory signature. J Am Acad Dermatol 2020; 82:690-699. [DOI: 10.1016/j.jaad.2019.10.039] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/16/2019] [Accepted: 10/22/2019] [Indexed: 01/08/2023]
|
74
|
Review-Current Concepts in Inflammatory Skin Diseases Evolved by Transcriptome Analysis: In-Depth Analysis of Atopic Dermatitis and Psoriasis. Int J Mol Sci 2020; 21:ijms21030699. [PMID: 31973112 PMCID: PMC7037913 DOI: 10.3390/ijms21030699] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
During the last decades, high-throughput assessment of gene expression in patient tissues using microarray technology or RNA-Seq took center stage in clinical research. Insights into the diversity and frequency of transcripts in healthy and diseased conditions provide valuable information on the cellular status in the respective tissues. Growing with the technique, the bioinformatic analysis toolkit reveals biologically relevant pathways which assist in understanding basic pathophysiological mechanisms. Conventional classification systems of inflammatory skin diseases rely on descriptive assessments by pathologists. In contrast to this, molecular profiling may uncover previously unknown disease classifying features. Thereby, treatments and prognostics of patients may be improved. Furthermore, disease models in basic research in comparison to the human disease can be directly validated. The aim of this article is not only to provide the reader with information on the opportunities of these techniques, but to outline potential pitfalls and technical limitations as well. Major published findings are briefly discussed to provide a broad overview on the current findings in transcriptomics in inflammatory skin diseases.
Collapse
|
75
|
Tang HHF, Sly PD, Holt PG, Holt KE, Inouye M. Systems biology and big data in asthma and allergy: recent discoveries and emerging challenges. Eur Respir J 2020; 55:13993003.00844-2019. [PMID: 31619470 DOI: 10.1183/13993003.00844-2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022]
Abstract
Asthma is a common condition caused by immune and respiratory dysfunction, and it is often linked to allergy. A systems perspective may prove helpful in unravelling the complexity of asthma and allergy. Our aim is to give an overview of systems biology approaches used in allergy and asthma research. Specifically, we describe recent "omic"-level findings, and examine how these findings have been systematically integrated to generate further insight.Current research suggests that allergy is driven by genetic and epigenetic factors, in concert with environmental factors such as microbiome and diet, leading to early-life disturbance in immunological development and disruption of balance within key immuno-inflammatory pathways. Variation in inherited susceptibility and exposures causes heterogeneity in manifestations of asthma and other allergic diseases. Machine learning approaches are being used to explore this heterogeneity, and to probe the pathophysiological patterns or "endotypes" that correlate with subphenotypes of asthma and allergy. Mathematical models are being built based on genomic, transcriptomic and proteomic data to predict or discriminate disease phenotypes, and to describe the biomolecular networks behind asthma.The use of systems biology in allergy and asthma research is rapidly growing, and has so far yielded fruitful results. However, the scale and multidisciplinary nature of this research means that it is accompanied by new challenges. Ultimately, it is hoped that systems medicine, with its integration of omics data into clinical practice, can pave the way to more precise, personalised and effective management of asthma.
Collapse
Affiliation(s)
- Howard H F Tang
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia .,Cambridge Baker Systems Genomics Initiative, Dept of Public Health and Primary Care, University of Cambridge, Cambridge, UK.,School of BioSciences, The University of Melbourne, Parkville, Australia
| | - Peter D Sly
- Queensland Children's Medical Research Institute, The University of Queensland, Brisbane, Australia.,Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Patrick G Holt
- Queensland Children's Medical Research Institute, The University of Queensland, Brisbane, Australia.,Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Kathryn E Holt
- Dept of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Australia.,London School of Hygiene and Tropical Medicine, London, UK
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia.,Cambridge Baker Systems Genomics Initiative, Dept of Public Health and Primary Care, University of Cambridge, Cambridge, UK.,School of BioSciences, The University of Melbourne, Parkville, Australia.,The Alan Turing Institute, London, UK
| |
Collapse
|
76
|
Pan Y, Wen X, Hao D, Wang Y, Wang L, He G, Jiang X. The role of IL-37 in skin and connective tissue diseases. Biomed Pharmacother 2019; 122:109705. [PMID: 31918276 DOI: 10.1016/j.biopha.2019.109705] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/12/2019] [Accepted: 11/24/2019] [Indexed: 02/05/2023] Open
Abstract
IL-37 was discovered as an anti-inflammatory and immunosuppressive cytokine of the IL-1 family. Significant advancements in the understanding of signaling pathways associated with IL-37 have been made in recent years. IL-37 binds to IL-18R and recruits IL-1R8 to form the IL-37/IL-1R8/IL-18Rα complex. Capase-1 plays a key role in the nuclear transduction of IL-37 signal, processing precursor IL-37 into the mature isoform, and interacting with Smad3. IL-37 exerts its role by activating anti-inflammation pathways including AMPK, PTEN, Mer, STAT3 and p62, and promoting tolerogenic dendritic cells and Tregs. In addition, IL-37 inhibits pro-inflammatory cytokines such as IL-1, IL-6, IL-8, IL-17, IL-23, TNF-α, and IFN-γ, and suppresses Fyn, MAPK, TAK1, NFκB, and mTOR signaling. The final effects of IL-37 depend on the interaction among IL-18R, IL-1R8, IL-37 and IL-18BP. Previous studies have deciphered the role of IL-37 in the development and pathogenesis of autoimmune diseases, chronic infections and cancer. In this review, we discuss the role of IL-37 in psoriasis, atopic dermatitis, Behcet's diseases, systemic lupus erythematosus, and other skin and connective tissue diseases.
Collapse
Affiliation(s)
- Yu Pan
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xiang Wen
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Dan Hao
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yujia Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lian Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Gu He
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, PR China.
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
77
|
Fung WT, Wu JT, Chan WMM, Chan HH, Pang H. Pathway-based meta-analysis for partially paired transcriptomics analysis. Res Synth Methods 2019; 11:123-133. [PMID: 31682084 DOI: 10.1002/jrsm.1381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/27/2019] [Accepted: 09/16/2019] [Indexed: 11/09/2022]
Abstract
Pathway-based differential expression analysis allows the incorporation of biological domain knowledge into transcriptomics analysis to enhance our understanding of disease mechanisms. To integrate information among multiple studies at the pathway level, pathway-based meta-analysis can be performed. Paired or partially paired samples are common in biomedical research. However, there are currently no existing pathway-based meta-analysis methods appropriate for paired or partially paired study designs. In this study, we developed a pathway-based meta-analysis approach for paired or partially paired samples. Meta-analysis on the transcriptomics profiles were conducted using p-value-based, rank-based, and effect size-based algorithms. The application of our approach was demonstrated using partially paired data from psoriasis transcriptomics studies. Upon combining six transcriptomics studies, genes related to the cell cycle and DNA replication pathways are found to be highly perturbed in psoriatic lesional skin samples. Results were validated externally with independent RNA-Seq data. Comparison with existing pathway meta-analysis methods revealed consistent results, with our method showing higher detection power. This study demonstrated the utility of our newly developed pathway-based meta-analysis that allows the incorporation of partially paired or paired samples. The proposed framework can be applied to omics data including but not limited to transcriptomics data.
Collapse
Affiliation(s)
- Wing Tung Fung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Joseph T Wu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wai Man Mandy Chan
- Division of Dermatology, Department of Medicine, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Henry H Chan
- Division of Dermatology, Department of Medicine, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Herbert Pang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
78
|
Crisaborole and atopic dermatitis skin biomarkers: An intrapatient randomized trial. J Allergy Clin Immunol 2019; 144:1274-1289. [DOI: 10.1016/j.jaci.2019.06.047] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/15/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022]
|
79
|
He H, Li R, Choi S, Zhou L, Pavel A, Estrada YD, Krueger JG, Guttman-Yassky E. Increased cardiovascular and atherosclerosis markers in blood of older patients with atopic dermatitis. Ann Allergy Asthma Immunol 2019; 124:70-78. [PMID: 31622668 DOI: 10.1016/j.anai.2019.10.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/02/2019] [Accepted: 10/06/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is associated with increased systemic inflammation and cardiovascular risk. Although previous studies have found increased inflammatory proteins in the blood of patients with AD, detailed comparison among patients with AD of different ages is unavailable. OBJECTIVE To characterize the blood proteomic signature of patients with AD as a function of age. METHODS We used the OLINK high-throughput proteomic assay to measure serum inflammatory and cardiovascular risk proteins in 71 patients with moderate to severe AD from 3 age groups (18-40 years old [n = 26], 41-60 years old [n = 24], and >60 years old [n = 21]) compared with 37 age-matched controls. Total and allergen-specific serum IgEs were also measured. RESULTS When we compared patients with AD from 3 different age groups with their respective controls, we identified a total of 172 differentially expressed proteins. TH2 chemokines (CCL13, CCL17) were consistently elevated in patients with AD across all ages (P < .05), whereas TH1 (CXCL10) and TH17 (KYNU, CCL20) markers incrementally increased with age in both patients with AD and healthy subjects. Elderly patients with AD (>60 years old) exhibited striking upregulation of key proinflammatory proteins, including markers of atherosclerosis (CCL4, CCL7, SORT1), cardiovascular risk (GDF15, MPO, ST2), cell adhesion (CDH3), and apoptosis (FAS; all P < .05) compared with younger patients with AD and age-matched controls. We also found that total and allergen-specific serum IgEs decreased significantly with age in patients with AD (P < .05). CONCLUSION Elderly patients with AD had increased levels of systemic inflammatory markers, including those associated with cardiovascular and atherosclerosis risk, which may explain their increased incidence of cardiovascular disease. This finding suggests that older patients with AD may benefit from cardiovascular disease screening and prevention.
Collapse
Affiliation(s)
- Helen He
- Department of Dermatology, Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Randall Li
- Department of Dermatology, Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Seulah Choi
- Department of Dermatology, Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lisa Zhou
- Department of Dermatology, Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ana Pavel
- Department of Dermatology, Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yeriel D Estrada
- Department of Dermatology, Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Emma Guttman-Yassky
- Department of Dermatology, Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
80
|
Oral Janus kinase/SYK inhibition (ASN002) suppresses inflammation and improves epidermal barrier markers in patients with atopic dermatitis. J Allergy Clin Immunol 2019; 144:1011-1024. [DOI: 10.1016/j.jaci.2019.07.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/26/2019] [Accepted: 07/18/2019] [Indexed: 12/26/2022]
|
81
|
Smith L, Gatault S, Casals-Diaz L, Kelly PA, Camerer E, Métais C, Knaus UG, Eissner G, Steinhoff M. House dust mite-treated PAR2 over-expressor mouse: A novel model of atopic dermatitis. Exp Dermatol 2019; 28:1298-1308. [PMID: 31487753 DOI: 10.1111/exd.14030] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/14/2019] [Accepted: 08/28/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is a complex skin disease involving causative effects from both intrinsic and extrinsic sources. Murine models of the disease often fall short in one of these components and, as a result, do not fully encapsulate these disease mechanisms. OBJECTIVE We aimed to determine whether the protease-activated receptor 2 over-expressor mouse (PAR2OE) with topical house dust mite (HDM) application is a more comprehensive and clinically representative AD model. METHODS Following HDM extract application to PAR2OE mice and controls, AD clinical scoring, itching behaviour, skin morphology and structure, barrier function, immune cell infiltration and inflammatory markers were assessed. Skin morphology was analysed using haematoxylin and eosin staining, and barrier function was assessed by transepidermal water loss measurements. Immune infiltrate was characterised by histological and immunofluorescence staining. Finally, an assessment of AD-related gene expression was performed using quantitative RT-PCR. RESULTS PAR2OE mice treated with HDM displays all the characteristic clinical symptoms including erythema, dryness and oedema, skin morphology, itch and inflammation typically seen in patients with AD. There is a significant influx of mast cells (P < .01) and eosinophils (P < .0001) into the dermis of these mice. Furthermore, the PAR2OE + HDM mice exhibit similar expression patterns of key differentially expressed genes as seen in human AD. CONCLUSION The PAR2OE + HDM mouse presents with a classic AD pathophysiology and is a valuable model in terms of reproducibility and overall disease representation to study the condition and potential therapeutic approaches.
Collapse
Affiliation(s)
- Leila Smith
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Solene Gatault
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Laura Casals-Diaz
- Skin Biology and Pharmacology, Almirall R&D Centre, Sant Feliu de Llobregat, Barcelona, Spain
| | - Pamela A Kelly
- Department of Veterinary Pathology, School of Veterinary Medicine, University College Dublin, Dublin 4, Ireland
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France
| | - Charles Métais
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Ulla G Knaus
- Conway Institute, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Günther Eissner
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Martin Steinhoff
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland.,Department of Dermatology, Hamad Medical Corporation and Qatar University, Doha, Qatar
| |
Collapse
|
82
|
Minzaghi D, Pavel P, Dubrac S. Xenobiotic Receptors and Their Mates in Atopic Dermatitis. Int J Mol Sci 2019; 20:E4234. [PMID: 31470652 PMCID: PMC6747412 DOI: 10.3390/ijms20174234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/19/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023] Open
Abstract
Atopic dermatitis (AD) is the most common inflammatory skin disease worldwide. It is a chronic, relapsing and pruritic skin disorder which results from epidermal barrier abnormalities and immune dysregulation, both modulated by environmental factors. AD is strongly associated with asthma and allergic rhinitis in the so-called 'atopic march.' Xenobiotic receptors and their mates are ligand-activated transcription factors expressed in the skin where they control cellular detoxification pathways. Moreover, they regulate the expression of genes in pathways involved in AD in epithelial cells and immune cells. Activation or overexpression of xenobiotic receptors in the skin can be deleterious or beneficial, depending on context, ligand and activation duration. Moreover, their impact on skin might be amplified by crosstalk among xenobiotic receptors and their mates. Because they are activated by a broad range of endogenous molecules, drugs and pollutants owing to their promiscuous ligand affinity, they have recently crystalized the attention of researchers, including in dermatology and especially in the AD field. This review examines the putative roles of these receptors in AD by critically evaluating the conditions under which the proteins and their ligands have been studied. This information should provide new insights into AD pathogenesis and ways to develop new therapeutic interventions.
Collapse
Affiliation(s)
- Deborah Minzaghi
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Petra Pavel
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
83
|
Messad F, Louveau I, Koffi B, Gilbert H, Gondret F. Investigation of muscle transcriptomes using gradient boosting machine learning identifies molecular predictors of feed efficiency in growing pigs. BMC Genomics 2019; 20:659. [PMID: 31419934 PMCID: PMC6697907 DOI: 10.1186/s12864-019-6010-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 07/30/2019] [Indexed: 01/09/2023] Open
Abstract
Background Improving feed efficiency (FE) is a major challenge in pig production. This complex trait is characterized by a high variability. Therefore, the identification of predictors of FE may be a relevant strategy to reduce phenotyping efforts in breeding and selection programs. The aim of this study was to investigate the suitability of expressed muscle genes in prediction of FE traits in growing pigs. The approach considered different transcriptomics experiments to cover a large range of FE values and identify reliable predictors. Results Microarrays data were obtained from longissimus muscles of two lines divergently selected for residual feed intake (RFI). Pigs (n = 71) from three experiments belonged to generations 6 to 8 of selection, were fed either a diet with a standard composition or a diet rich in fiber and lipids, received feed ad libitum or at restricted level, and weighed between 80 and 115 kg at slaughter. For each pig, breeding value for RFI was estimated (RFI-BV), and feed conversion ratio (FCR) and energy-based feed conversion ratio (FCRe) were calculated during the test periods. Gradient boosting algorithms were used on the merged muscle transcriptomes to identify very important predictors of FE traits. About 20,405 annotated molecular probes were commonly expressed in longissimus muscle across experiments. Six to 267 expressed muscle genes covering a variety of biological processes were found as important predictors for RFI-BV (R2 = 0.63–0.65), FCR (R2 = 0.61–0.70) and FCRe (R2 = 0.49–0.52). The error of prediction was less than 8% for FCR. Altogether, 56 predictors were common to RFI-BV and FCR. Expression levels of 24 target genes were further measured by qPCR. Linear regression confirmed the good accuracy of combining mRNA levels of these genes to fit FE traits (RFI-BV: R2 = 0.73, FRC: R2 = 0.76; FCRe: R2 = 0.75). Stepwise regression procedure highlighted 10 genes (FKBP5, MUM1, AKAP12, FYN, TMED3, PHKB, TGF, SOCS6, ILR4, and FRAS1) in a linear combination predicting FCR and FCRe. In addition, FKBP5 and expression levels of five other genes (IGF2, SERINC3, CSRNP3, EZR and RPL16) significantly contributed to RFI-BV. Conclusion It was possible to identify few genes expressed in muscle that might be reliable predictors of feed efficiency. Electronic supplementary material The online version of this article (10.1186/s12864-019-6010-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Farouk Messad
- Pegase, INRA, Agrocampus Ouest, 35590, Saint-Gilles, France
| | | | - Basile Koffi
- Pegase, INRA, Agrocampus Ouest, 35590, Saint-Gilles, France
| | | | | |
Collapse
|
84
|
Bhattacharya N, Ganguli-Indra G, Indra AK. Transcriptional control and transcriptomic analysis of lipid metabolism in skin barrier formation and atopic dermatitis (AD). Expert Rev Proteomics 2019; 16:627-645. [PMID: 31322970 DOI: 10.1080/14789450.2019.1646128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Atopic dermatitis (AD) is a multifactorial ailment associated with barrier breach and intense systemic inflammation. Several studies over the years have shown the complex interplay of a large number of factors in governing the progression and outcome of AD. In addition to the diverse types of AD resulting due to variation in the intrinsic mechanisms giving rise to AD such as single nucleotide polymorphisms (SNPs), epigenetic alterations or transcriptional changes, extrinsic factors such as age, ancestry, ethnicity, immunological background of the subject, the interactions of the subject with environmental stimuli and existing microbiome in the periphery surrounding the subject account for further heterogeneity in the clinical manifestations of the disease. Areas covered: Here we have selectively discussed transcriptional regulation of genes associated with skin lipid metabolism in the context of AD. Transcriptional control and transcriptomic changes are just one face of this multifaceted disease known to affect humans and a detailed study concerning those will enable us to develop targeted therapies to deal with the disease. Expert opinion: Large-scale integration of different omics approaches (genomics, epigenomics, transcriptomics, lipidomics, proteomics, metabolomics, effect of exposome) will help identify the potential candidate gene(s) associated with the development of various endotypes of AD.
Collapse
Affiliation(s)
- Nilika Bhattacharya
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU , Corvallis , OR , USA
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU , Corvallis , OR , USA.,Knight Cancer Institute, Oregon Health & Science University (OHSU) , Portland , OR , USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU , Corvallis , OR , USA.,Knight Cancer Institute, Oregon Health & Science University (OHSU) , Portland , OR , USA.,Department of Biochemistry and Biophysics, OSU , Corvallis , OR , USA.,Linus Pauling Science Center, Oregon State University (OSU) , Corvallis , OR , USA.,Department of Dermatology, Oregon Health & Science University (OHSU) , Portland , OR , USA
| |
Collapse
|
85
|
Rogerson C, O'Shaughnessy RFL. Protein kinases involved in epidermal barrier formation: The AKT family and other animals. Exp Dermatol 2019; 27:892-900. [PMID: 29845670 DOI: 10.1111/exd.13696] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2018] [Indexed: 12/20/2022]
Abstract
Formation of a stratified epidermis is required for the performance of the essential functions of the skin; to act as an outside-in barrier against the access of microorganisms and other external factors, to prevent loss of water and solutes via inside-out barrier functions and to withstand mechanical stresses. Epidermal barrier function is initiated during embryonic development and is then maintained throughout life and restored after injury. A variety of interrelated processes are required for the formation of a stratified epidermis, and how these processes are both temporally and spatially regulated has long been an aspect of dermatological research. In this review, we describe the roles of multiple protein kinases in the regulation of processes required for epidermal barrier formation.
Collapse
Affiliation(s)
- Clare Rogerson
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, UK
| | - Ryan F L O'Shaughnessy
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
86
|
Moyle M, Cevikbas F, Harden JL, Guttman‐Yassky E. Understanding the immune landscape in atopic dermatitis: The era of biologics and emerging therapeutic approaches. Exp Dermatol 2019; 28:756-768. [PMID: 30825336 PMCID: PMC6850480 DOI: 10.1111/exd.13911] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/28/2019] [Accepted: 02/14/2019] [Indexed: 12/15/2022]
Abstract
Atopic dermatitis (AD) is a chronic, systemic, inflammatory disease that affects the skin and is characterized by persistent itch and marked redness. AD is associated with an increased risk of skin infections and a reduced quality of life. Most AD treatment options to date were not designed to selectively target disease-causing pathways that have been established for this indication. Topical therapies have limited efficacy in moderate-to-severe disease, and systemic agents such as corticosteroids and immunosuppressants present with tolerability issues. Advances in the understanding of AD pathobiology have made possible a new generation of more disease-specific AD therapies. AD is characterized by the inappropriate activation of type 2 T helper (Th2) cells and type 2 innate lymphoid (ILC2) cells, with a predominant increase in type 2 cytokines in the skin, including interleukin (IL)-13 and IL-4. Both cytokines are implicated in tissue inflammation and epidermal barrier dysfunction, and monoclonal antibodies targeting each of these interleukins or their receptors are in clinical development in AD. In March 2017, dupilumab, a human anti-IL-4Rα antibody, became the first biologic to receive approval in the United States for the treatment of moderate-to-severe AD. The anti-IL-13 monoclonal antibodies lebrikizumab and tralokinumab, which bind different IL-13 epitopes with potentially different effects, are currently in advanced-stage trials. Here, we briefly review the underlying pathobiology of AD, the scientific basis for current AD targets, and summarize current clinical studies of these agents, including new research to develop both predictive and response biomarkers to further advance AD therapy in the era of precision medicine.
Collapse
|
87
|
Litman T. Personalized medicine-concepts, technologies, and applications in inflammatory skin diseases. APMIS 2019; 127:386-424. [PMID: 31124204 PMCID: PMC6851586 DOI: 10.1111/apm.12934] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/31/2019] [Indexed: 12/19/2022]
Abstract
The current state, tools, and applications of personalized medicine with special emphasis on inflammatory skin diseases like psoriasis and atopic dermatitis are discussed. Inflammatory pathways are outlined as well as potential targets for monoclonal antibodies and small-molecule inhibitors.
Collapse
Affiliation(s)
- Thomas Litman
- Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
- Explorative Biology, Skin ResearchLEO Pharma A/SBallerupDenmark
| |
Collapse
|
88
|
Kori M, Gov E, Arga KY. Novel Genomic Biomarker Candidates for Cervical Cancer As Identified by Differential Co-Expression Network Analysis. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 23:261-273. [PMID: 31038390 DOI: 10.1089/omi.2019.0025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cervical cancer is the second most common malignancy and the third reason for mortality among women in developing countries. Although infection by the oncogenic human papilloma viruses is a major cause, genomic contributors are still largely unknown. Network analyses, compared with candidate gene studies, offer greater promise to map the interactions among genomic loci contributing to cervical cancer risk. We report here a differential co-expression network analysis in five gene expression datasets (GSE7803, GSE9750, GSE39001, GSE52903, and GSE63514, from the Gene Expression Omnibus) in patients with cervical cancer and healthy controls. Kaplan-Meier Survival and principle component analyses were employed to evaluate prognostic and diagnostic performances of biomarker candidates, respectively. As a result, seven distinct co-expressed gene modules were identified. Among these, five modules (with sizes of 9-45 genes) presented high prognostic and diagnostic capabilities with hazard ratios of 2.28-11.3, and diagnostic odds ratios of 85.2-548.8. Moreover, these modules were associated with several key biological processes such as cell cycle regulation, keratinization, neutrophil degranulation, and the phospholipase D signaling pathway. In addition, transcription factors ETS1 and GATA2 were noted as common regulatory elements. These genomic biomarker candidates identified by differential co-expression network analysis offer new prospects for translational cancer research, not to mention personalized medicine to forecast cervical cancer susceptibility and prognosis. Looking into the future, we also suggest that the search for a molecular basis of common complex diseases should be complemented by differential co-expression analyses to obtain a systems-level understanding of disease phenotype variability.
Collapse
Affiliation(s)
- Medi Kori
- 1 Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey
| | - Esra Gov
- 2 Department of Bioengineering, Faculty of Engineering, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| | - Kazım Yalçın Arga
- 1 Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey
| |
Collapse
|
89
|
Guttman-Yassky E, Pavel AB, Zhou L, Estrada YD, Zhang N, Xu H, Peng X, Wen HC, Govas P, Gudi G, Ca V, Fang H, Salhi Y, Back J, Reddy V, Bissonnette R, Maari C, Grossman F, Wolff G. GBR 830, an anti-OX40, improves skin gene signatures and clinical scores in patients with atopic dermatitis. J Allergy Clin Immunol 2019; 144:482-493.e7. [PMID: 30738171 DOI: 10.1016/j.jaci.2018.11.053] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 11/12/2018] [Accepted: 11/16/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND GBR 830 is a humanized mAb against OX40, a costimulatory receptor on activated T cells. OX40 inhibition might have a therapeutic role in T cell-mediated diseases, including atopic dermatitis (AD). OBJECTIVE This exploratory phase 2a study investigated the safety, efficacy, and tissue effects of GBR 830 in patients with AD. METHODS Patients with moderate-to-severe AD (affected body surface area, ≥10%; Eczema Area and Severity Index score, ≥12; and inadequate response to topical treatments) were randomized 3:1 to 10 mg/kg intravenous GBR 830 or placebo on day 1 (baseline) and day 29. Biopsy specimens were collected (n = 40) at days 1, 29, and 71. Primary end points included treatment-emergent adverse events (TEAEs) and changes from baseline in biomarkers (epidermal hyperplasia/cytokines) at days 29 and 71. RESULTS GBR 830 was well tolerated, with equal TEAE distribution (GBR 830, 63.0% [29/46]; placebo, 63.0% [10/16]). One serious TEAE in the GBR 830 group was deemed unrelated to study drug. At day 71, the proportion of intent-to-treat subjects achieving 50% or greater improvement in Eczema Area and Severity Index score was greater with GBR 830 (76.9% [20/26]) versus placebo (37.5% [3/8]). GBR 830 induced significant progressive reductions in TH1 (IFN-γ/CXCL10), TH2 (IL-31/CCL11/CCL17), and TH17/TH22 (IL-23p19/IL-8/S100A12) mRNA expression in lesional skin. Significant progressive reductions until day 71 in the drug group were seen in OX40+ T cells and OX40L+ dendritic cells (P < .001). Hyperplasia measures (thickness/keratin 16/Ki67) showed greater reductions with GBR 830 (P < .001). CONCLUSIONS Two doses of GBR 830 administered 4 weeks apart were well tolerated and induced significant progressive tissue and clinical changes until day 71 (42 days after the last dose), highlighting the potential of OX40 targeting in patients with AD.
Collapse
Affiliation(s)
| | - Ana B Pavel
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Lisa Zhou
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Ning Zhang
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hui Xu
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Xiangyu Peng
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Huei-Chi Wen
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | - Vinu Ca
- Glenmark Pharmaceuticals, Ltd, Mumbai, India
| | - Hui Fang
- Glenmark Pharmaceuticals, Inc, Paramus, NJ
| | | | - Jonathan Back
- Glenmark Pharmaceuticals, SA, La Chaux-de-Fonds, Switzerland
| | | | | | | | | | | |
Collapse
|
90
|
Czarnowicki T, He H, Krueger JG, Guttman-Yassky E. Atopic dermatitis endotypes and implications for targeted therapeutics. J Allergy Clin Immunol 2019; 143:1-11. [DOI: 10.1016/j.jaci.2018.10.032] [Citation(s) in RCA: 238] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/02/2018] [Accepted: 10/11/2018] [Indexed: 01/28/2023]
|
91
|
Tham EH, Leung DY. Mechanisms by Which Atopic Dermatitis Predisposes to Food Allergy and the Atopic March. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2019; 11:4-15. [PMID: 30479073 PMCID: PMC6267189 DOI: 10.4168/aair.2019.11.1.4] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 08/13/2018] [Indexed: 12/23/2022]
Abstract
The Atopic march denotes the progression from atopic dermatitis (AD) to the development of other allergic disorders such as immunoglobulin (Ig) E-mediated food allergy, allergic rhinitis and asthma in later childhood. There is increasing evidence from prospective birth cohort studies that early-onset AD is a risk factor for other allergic diseases or is found in strong association with them. Animal studies now provide mechanistic insights into the pathways that may be responsible for triggering the progression from the skin barrier dysfunction seen in AD to epicutaneous sensitization, food allergy and allergic airway disorders. Recent large randomized controlled trials have demonstrated the efficacy of early interventions targeted at AD and food allergy prevention. These show great promise for research into future strategies aimed at prevention of the atopic march.
Collapse
Affiliation(s)
- Elizabeth Huiwen Tham
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore
- Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Donald Ym Leung
- Department of Pediatrics, National Jewish Health, Denver, CO, USA
- Department of Pediatrics, University of Colorado at Denver Health Sciences Center, Aurora, CO, USA.
| |
Collapse
|
92
|
Atopic and Contact Dermatitis. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00044-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
93
|
Brunner PM, Pavel AB, Khattri S, Leonard A, Malik K, Rose S, Jim On S, Vekaria AS, Traidl-Hoffmann C, Singer GK, Baum D, Gilleaudeau P, Sullivan-Whalen M, Fuentes-Duculan J, Li X, Zheng X, Estrada Y, Garcet S, Wen HC, Gonzalez J, Coats I, Cueto I, Neumann AU, Lebwohl MG, Krueger JG, Guttman-Yassky E. Baseline IL-22 expression in patients with atopic dermatitis stratifies tissue responses to fezakinumab. J Allergy Clin Immunol 2019; 143:142-154. [PMID: 30121291 DOI: 10.1016/j.jaci.2018.07.028] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 07/06/2018] [Accepted: 07/27/2018] [Indexed: 12/23/2022]
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.
Collapse
Affiliation(s)
- Patrick M Brunner
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Ana B Pavel
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Saakshi Khattri
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexandra Leonard
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kunal Malik
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sharon Rose
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Shelbi Jim On
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anjali S Vekaria
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Claudia Traidl-Hoffmann
- Institute of Environmental Medicine, University Center for Health Sciences at the Klinikum Augsburg, Technical University Munich and Helmholtz Zentrum München-German Research Center for Environmental Health, Augsburg, Germany; Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Giselle K Singer
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Danielle Baum
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | | | - Xuan Li
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Xiuzhong Zheng
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Yeriel Estrada
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sandra Garcet
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Huei-Chi Wen
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Juana Gonzalez
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Israel Coats
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Inna Cueto
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Avidan U Neumann
- Institute of Environmental Medicine, University Center for Health Sciences at the Klinikum Augsburg, Technical University Munich and Helmholtz Zentrum München-German Research Center for Environmental Health, Augsburg, Germany
| | - Mark G Lebwohl
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - James G Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Emma Guttman-Yassky
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
94
|
Sanyal RD, Pavel AB, Glickman J, Chan TC, Zheng X, Zhang N, Cueto I, Peng X, Estrada Y, Fuentes-Duculan J, Alexis AF, Krueger JG, Guttman-Yassky E. Atopic dermatitis in African American patients is T H2/T H22-skewed with T H1/T H17 attenuation. Ann Allergy Asthma Immunol 2019; 122:99-110.e6. [PMID: 30223113 DOI: 10.1016/j.anai.2018.08.024] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 08/28/2018] [Accepted: 08/31/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND African Americans (AA) are disproportionately impacted by atopic dermatitis (AD), with increased prevalence and therapeutic challenges unique to this population. Molecular profiling data informing development of targeted therapeutics for AD are derived primarily from European American (EA) patients. These studies are absent in AA, hindering development of effective treatments for this population. OBJECTIVE We sought to characterize the global molecular profile of AD in the skin of AA patients as compared with that of EA AD and healthy controls. METHODS We performed RNA-Seq with reverse transcription polymerase chain reaction validation and immunohistochemistry studies in lesional and nonlesional skin of AA and EA AD patients vs healthy controls. RESULTS African American AD lesions were characterized by greater infiltration of dendritic cells (DCs) marked by the high-affinity immunoglobulin E (IgE) receptor (FcεR1+) compared with EA AD (P < .05). Both AD cohorts showed similarly robust up-regulation of Th2-related (CCL17/18/26) and Th22-related markers (interleukin [IL]-22, S100A8/9/12), but AA AD featured decreased expression of innate immune (tumor necrosis factor [TNF], IL-1β), Th1-related (interferon gamma [IFN-γ], MX1, IL-12RB1), and Th17-related markers (IL-23p19, IL-36G, CXCL1) vs EA AD (P < .05). The Th2 (IL-13) and Th22-related products (IL-22, S100A8/9/12) and serum IgE were significantly correlated with clinical severity (Scoring of Atopic Dermatitis [SCORAD]) in AA. Fillagrin (FLG) was exclusively down-regulated in EA AD, whereas loricrin (LOR) was down-regulated in both AD cohorts and negatively correlated with SCORAD in AA. CONCLUSION The molecular phenotype of AA AD skin is characterized by attenuated Th1 and Th17 but similar Th2/Th22-skewing to EA AD. Our data encourages a personalized medicine approach accounting for phenotype-specific characteristics in future development of targeted therapeutics and clinical trial design for AD.
Collapse
Affiliation(s)
- Riana D Sanyal
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ana B Pavel
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jacob Glickman
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Tom C Chan
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan; The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Xiuzhong Zheng
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Ning Zhang
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Inna Cueto
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Xiangyu Peng
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yeriel Estrada
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Judilyn Fuentes-Duculan
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Andrew F Alexis
- Department of Dermatology, Mount Sinai St. Luke's and Mount Sinai West, New York, New York
| | - James G Krueger
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Emma Guttman-Yassky
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
95
|
Ghosh D, Bernstein JA, Khurana Hershey GK, Rothenberg ME, Mersha TB. Leveraging Multilayered "Omics" Data for Atopic Dermatitis: A Road Map to Precision Medicine. Front Immunol 2018; 9:2727. [PMID: 30631320 PMCID: PMC6315155 DOI: 10.3389/fimmu.2018.02727] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022] Open
Abstract
Atopic dermatitis (AD) is a complex multifactorial inflammatory skin disease that affects ~280 million people worldwide. About 85% of AD cases begin in childhood, a significant portion of which can persist into adulthood. Moreover, a typical progression of children with AD to food allergy, asthma or allergic rhinitis has been reported (“allergic march” or “atopic march”). AD comprises highly heterogeneous sub-phenotypes/endotypes resulting from complex interplay between intrinsic and extrinsic factors, such as environmental stimuli, and genetic factors regulating cutaneous functions (impaired barrier function, epidermal lipid, and protease abnormalities), immune functions and the microbiome. Though the roles of high-throughput “omics” integrations in defining endotypes are recognized, current analyses are primarily based on individual omics data and using binary clinical outcomes. Although individual omics analysis, such as genome-wide association studies (GWAS), can effectively map variants correlated with AD, the majority of the heritability and the functional relevance of discovered variants are not explained or known by the identified variants. The limited success of singular approaches underscores the need for holistic and integrated approaches to investigate complex phenotypes using trans-omics data integration strategies. Integrating omics layers (e.g., genome, epigenome, transcriptome, proteome, metabolome, lipidome, exposome, microbiome), which often have complementary and synergistic effects, might provide the opportunity to capture the flow of information underlying AD disease manifestation. Overlapping genes/candidates derived from multiple omics types include FLG, SPINK5, S100A8, and SERPINB3 in AD pathogenesis. Overlapping pathways include macrophage, endothelial cell and fibroblast activation pathways, in addition to well-known Th1/Th2 and NFkB activation pathways. Interestingly, there was more multi-omics overlap at the pathway level than gene level. Further analysis of multi-omics overlap at the tissue level showed that among 30 tissue types from the GTEx database, skin and esophagus were significantly enriched, indicating the biological interconnection between AD and food allergy. The present work explores multi-omics integration and provides new biological insights to better define the biological basis of AD etiology and confirm previously reported AD genes/pathways. In this context, we also discuss opportunities and challenges introduced by “big omics data” and their integration.
Collapse
Affiliation(s)
- Debajyoti Ghosh
- Division of Immunology, Allergy & Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Jonathan A Bernstein
- Division of Immunology, Allergy & Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Tesfaye B Mersha
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
96
|
Guttman‐Yassky E, Hanifin JM, Boguniewicz M, Wollenberg A, Bissonnette R, Purohit V, Kilty I, Tallman AM, Zielinski MA. The role of phosphodiesterase 4 in the pathophysiology of atopic dermatitis and the perspective for its inhibition. Exp Dermatol 2018; 28:3-10. [DOI: 10.1111/exd.13808] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/12/2018] [Accepted: 10/12/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Emma Guttman‐Yassky
- Department of DermatologyIcahn School of Medicine at Mount Sinai New York New York
| | - Jon M. Hanifin
- Department of DermatologyOregon Health and Science University Portland Oregon
| | | | - Andreas Wollenberg
- Department of Dermatology and AllergyLudwig Maximilian University Munich Germany
| | | | | | | | | | | |
Collapse
|
97
|
Integrating the skin and blood transcriptomes and serum proteome in hidradenitis suppurativa reveals complement dysregulation and a plasma cell signature. PLoS One 2018; 13:e0203672. [PMID: 30265680 PMCID: PMC6162087 DOI: 10.1371/journal.pone.0203672] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 08/24/2018] [Indexed: 01/10/2023] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic skin disease of the pilo-sebaceous apocrine unit characterized by significant inflammation and an impaired quality of life. The pathogenesis of HS remains unclear. To determine the HS skin and blood transcriptomes and HS blood proteome, patient data from previously published studies were analysed and integrated from a cohort of patients with moderate to severe HS (n = 17) compared to healthy volunteers (n = 10). The analysis utilized empirical Bayes methods to determine differentially expressed genes (DEGs) (fold change (FCH) >2.0 and false discovery rate (FDR) <0.05), and differentially expressed proteins (DEPs) (FCH>1.5, FDR<0.05). In the HS skin transcriptome (lesional skin compared to non-lesional skin), there was an abundance of immunoglobulins, antimicrobial peptides, and an interferon signature. Gene-sets related to Notch signalling and Interferon pathways were differentially activated in lesional compared to non-lesional skin. CIBERSORT analysis of the HS skin transcriptome revealed a significantly increased proportion of plasma cells in lesional skin. In the HS skin and blood transcriptomes and HS blood proteome, gene-sets related to the complement system changed significantly (FDR<0.05), with dysregulation of complement-specific DEGs and DEPs. These data point towards an exaggerated immune response in lesional skin that may be responding to commensal cutaneous bacterial presence and raise the possibility that this may be an important driver of HS disease progression.
Collapse
|
98
|
Guttman-Yassky E, Bissonnette R, Ungar B, Suárez-Fariñas M, Ardeleanu M, Esaki H, Suprun M, Estrada Y, Xu H, Peng X, Silverberg JI, Menter A, Krueger JG, Zhang R, Chaudhry U, Swanson B, Graham NMH, Pirozzi G, Yancopoulos GD, D Hamilton JD. Dupilumab progressively improves systemic and cutaneous abnormalities in patients with atopic dermatitis. J Allergy Clin Immunol 2018; 143:155-172. [PMID: 30194992 DOI: 10.1016/j.jaci.2018.08.022] [Citation(s) in RCA: 451] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 07/19/2018] [Accepted: 08/14/2018] [Indexed: 01/16/2023]
Abstract
BACKGROUND Dupilumab is an IL-4 receptor α mAb inhibiting signaling of IL-4 and IL-13, key drivers of type 2-driven inflammation, as demonstrated by its efficacy in patients with atopic/allergic diseases. OBJECTIVE This placebo-controlled, double-blind trial (NCT01979016) evaluated the efficacy, safety, and effects of dupilumab on molecular/cellular lesional and nonlesional skin phenotypes and systemic type 2 biomarkers of patients with moderate-to-severe atopic dermatitis (AD). METHODS Skin biopsy specimens and blood were evaluated from 54 patients randomized 1:1 to weekly subcutaneous doses of 200 mg of dupilumab or placebo for 16 weeks. RESULTS Dupilumab (vs placebo) significantly improved clinical signs and symptoms of AD, was well tolerated, and progressively shifted the lesional transcriptome toward a nonlesional phenotype (weeks 4-16). Mean improvements in a meta-analysis-derived AD transcriptome (genes differentially expressed between lesional and nonlesional skin) were 68.8% and 110.8% with dupilumab and -10.5% and 55.0% with placebo (weeks 4 and 16, respectively; P < .001). Dupilumab significantly reduced expression of genes involved in type 2 inflammation (IL13, IL31, CCL17, CCL18, and CCL26), epidermal hyperplasia (keratin 16 [K16] and MKi67), T cells, dendritic cells (ICOS, CD11c, and CTLA4), and TH17/TH22 activity (IL17A, IL-22, and S100As) and concurrently increased expression of epidermal differentiation, barrier, and lipid metabolism genes (filaggrin [FLG], loricrin [LOR], claudins, and ELOVL3). Dupilumab reduced lesional epidermal thickness versus placebo (week 4, P = .001; week 16, P = .0002). Improvements in clinical and histologic measures correlated significantly with modulation of gene expression. Dupilumab also significantly suppressed type 2 serum biomarkers, including CCL17, CCL18, periostin, and total and allergen-specific IgEs. CONCLUSION Dupilumab-mediated inhibition of IL-4/IL-13 signaling through IL-4 receptor α blockade significantly and progressively improved disease activity, suppressed cellular/molecular cutaneous markers of inflammation and systemic measures of type 2 inflammation, and reversed AD-associated epidermal abnormalities.
Collapse
Affiliation(s)
- Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY.
| | | | - Benjamin Ungar
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Mayte Suárez-Fariñas
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Hitokazu Esaki
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Maria Suprun
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yeriel Estrada
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hui Xu
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Xiangyu Peng
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jonathan I Silverberg
- Department of Dermatology, Preventive Medicine and Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Alan Menter
- Department of Dermatology, Baylor University Medical Center, Dallas, Tex
| | - James G Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Rick Zhang
- Regeneron Pharmaceuticals, Tarrytown, NY
| | | | | | | | | | | | | |
Collapse
|
99
|
Mills KJ, Robinson MK, Sherrill JD, Schnell DJ, Xu J. Analysis of gene expression profiles of multiple skin diseases identifies a conserved signature of disrupted homeostasis. Exp Dermatol 2018; 27:1000-1008. [DOI: 10.1111/exd.13694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Kevin J. Mills
- The Procter and Gamble Company; Mason Business Center; Mason OH USA
| | | | | | - Daniel J. Schnell
- Department of Biomedical Informatics; Cincinnati Children's Hospital Medical Center; Cincinnati OH USA
| | - Jun Xu
- Calico, LLC; South San Francisco CA USA
| |
Collapse
|
100
|
Cho BA, Yoo SK, Seo JS. Signatures of photo-aging and intrinsic aging in skin were revealed by transcriptome network analysis. Aging (Albany NY) 2018; 10:1609-1626. [PMID: 30021930 PMCID: PMC6075446 DOI: 10.18632/aging.101496] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/06/2018] [Indexed: 12/18/2022]
Abstract
There are various factors that alter physiological characteristics in skin. Elucidating the underlying mechanism of transcriptional alterations by intrinsic and extrinsic factors may lead us to understand the aging process of skin. To identify the transcriptomic changes of the aging skin, we analyzed publicly available RNA sequencing data from Genotype-Tissue Expression (GTEx) project. GTEx provided RNA sequencing data of suprapubic (n=228) and lower leg (n=349) skins, which are photo-protected and photo-damaged. Using differentially expressed gene analysis and weighted gene co-expression network analysis, we characterized transcriptomic changes due to UV exposure and aging. Genes involved in skin development such as epidermal differentiation complex component (SPRR and LCE families), vasculature development (TGFBR1, TGFBR2, TGFBR3, KDR, FGF2, and VEGFC), and matrix metalloproteinase (MMP2, MMP3, MMP8, MMP10, and MMP13) were up-regulated by UV exposure. Also, down-regulated lipid metabolism and mitochondrial biogenesis were observed in photo-damaged skin. Moreover, wound healing process was universally down-regulated in suprapubic and lower leg with aging and further down-regulation of lipid metabolism and up-regulation of vasculature development were found as photo-aging signatures. In this study, dynamic transcriptomic alterations were observed in aged skin. Hence, our findings may help to discover a potential therapeutic target for skin rejuvenation.
Collapse
Affiliation(s)
- Byuri Angela Cho
- Gong Wu Genomic Medicine Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
- These authors contributed equally to this work
| | - Seong-Keun Yoo
- Genomic Institute, Macrogen Inc., Seoul, Republic of Korea
- These authors contributed equally to this work
| | - Jeong-Sun Seo
- Gong Wu Genomic Medicine Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
- Genomic Institute, Macrogen Inc., Seoul, Republic of Korea
| |
Collapse
|