1
|
Chen YC, Cheng YP, Liu CY, Wang WC, Wang TM, Lai CY, Guo JW. Sakuranetin modulates IL-17A-related inflammation and enhances skin barrier function in an imiquimod-induced psoriasis model. Biomed Pharmacother 2025; 186:118035. [PMID: 40209307 DOI: 10.1016/j.biopha.2025.118035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025] Open
Abstract
Sakuranetin, a methoxylated flavanone with known anti-inflammatory and antioxidant properties, has yet to be explored for its therapeutic potential in psoriasis treatment. This study evaluates its efficacy in an imiquimod-induced psoriasis-like murine model, focusing on both immune modulation and skin barrier restoration. Topical administration of sakuranetin significantly attenuated psoriatic inflammation, with high-dose treatment achieving comparable efficacy to desoximetasone. RNA sequencing and qPCR validation revealed that sakuranetin modulates keratinization, cornified envelope formation, and kallikrein-related peptidase activity, suggesting its role in epidermal homeostasis. Notably, sakuranetin selectively downregulated IL-17A while maintaining IL-17F expression, supporting a targeted immunomodulatory effect. Additionally, sakuranetin upregulated KRT1, KRT16, KLK7, Sprr2f, and Wfdc18 while downregulating Sprr2k and Sprr3, indicating a refined regulation of skin barrier proteins. The observed upregulation of Tslp suggests a potential role in epidermal repair mechanisms. These findings highlight sakuranetin as a novel, plant-derived therapeutic candidate with dual anti-inflammatory and skin barrier-enhancing properties, providing a potential corticosteroid-sparing approach for long-term psoriasis management.
Collapse
Affiliation(s)
- Yung-Chuan Chen
- Department of Pediatrics, Cathay General Hospital, Taipei 10630, Taiwan.
| | - Yu-Ping Cheng
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan; Department of Dermatology, Cathay General Hospital, Taipei 10630, Taiwan.
| | - Chih-Yi Liu
- Division of Pathology, Sijhih Cathay General Hospital, Taipei 22174, Taiwan.
| | - Wei-Chin Wang
- Department of Pediatrics, Cathay General Hospital, Taipei 10630, Taiwan.
| | - Tzu-Ming Wang
- Department of Pediatrics, Cathay General Hospital, Taipei 10630, Taiwan.
| | - Chien-Yu Lai
- Department of Pediatrics, Cathay General Hospital, Taipei 10630, Taiwan.
| | - Jiun-Wen Guo
- Department of Medical Research, Cathay General Hospital, Taipei 10630, Taiwan.
| |
Collapse
|
2
|
Rybka M, Mazurek Ł, Jurak J, Laskowska A, Zajdel M, Czuwara J, Sulejczak D, Szudzik M, Samborowska E, Schwartz RA, Dziadek M, Salagierski S, Drapała A, Ufnal M, Konop M. Keratin-TMAO dressing accelerates full-thickness skin wound healing in diabetic rats via M2-macrophage polarization and the activation of PI3K/AKT/mTOR signaling pathway. Int J Biol Macromol 2025; 310:143313. [PMID: 40274140 DOI: 10.1016/j.ijbiomac.2025.143313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Affiliation(s)
- Mateusz Rybka
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research (CePT), Medical University of Warsaw, Warsaw, Poland.
| | - Łukasz Mazurek
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Jan Jurak
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Anna Laskowska
- Department of Pharmaceutical Microbiology and Bioanalysis, Centre for Preclinical Research and Technology (CePT), Faculty of Pharmacy, Medical University of Warsaw, Poland
| | - Mikołaj Zajdel
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Joanna Czuwara
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Mateusz Szudzik
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Emilia Samborowska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Robert A Schwartz
- Department of Dermatology and Pathology, Rutgers New Jersey Medical School, Newark, United States of America
| | - Michał Dziadek
- Department of Glass Technology and Amorphous Coatings, AGH University of Krakow, Krakow, Poland; Department of Materials Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Szymon Salagierski
- Department of Glass Technology and Amorphous Coatings, AGH University of Krakow, Krakow, Poland
| | - Adrian Drapała
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Marcin Ufnal
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Marek Konop
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research (CePT), Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
3
|
Sarkar S, Mandal D, Ghosh A, Chattopadhyay D. Biopolymers in Wound Dressing. ACS SYMPOSIUM SERIES 2024:207-234. [DOI: 10.1021/bk-2024-1487.ch009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Sresha Sarkar
- Department of Polymer Science and Technology, University of Calcutta, 92 A.P.C. Road, Kolkata -700 009, India
| | - Debashmita Mandal
- Center for Research in Nanoscience and Nanotechnology, Acharya Prafulla Chandra Roy Sikhsha Prangan, University of Calcutta, JD-2, Sector-III, Salt Lake City, Kolkata-700098, India
| | - Adrija Ghosh
- Department of Polymer Science and Technology, University of Calcutta, 92 A.P.C. Road, Kolkata -700 009, India
| | - Dipankar Chattopadhyay
- Department of Polymer Science and Technology, University of Calcutta, 92 A.P.C. Road, Kolkata -700 009, India
- Center for Research in Nanoscience and Nanotechnology, Acharya Prafulla Chandra Roy Sikhsha Prangan, University of Calcutta, JD-2, Sector-III, Salt Lake City, Kolkata-700098, India
| |
Collapse
|
4
|
Kelly-Scumpia KM, Archang MM, Purbey PK, Yokota T, Wu R, McCourt J, Li S, Crosbie RH, Scumpia PO, Deb A. Modulating the extracellular matrix to treat wound healing defects in Ehlers-Danlos syndrome. iScience 2024; 27:110676. [PMID: 39262784 PMCID: PMC11389543 DOI: 10.1016/j.isci.2024.110676] [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: 12/15/2023] [Revised: 05/13/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024] Open
Abstract
Classic Ehlers-Danlos syndrome (cEDS) is a genetic disorder of the connective tissue that is characterized by mutations in genes coding type V collagen. Wound healing defects are characteristic of cEDS and no therapeutic strategies exist. Herein we describe a murine model of cEDS that phenocopies wound healing defects seen in humans. Our model features mice with conditional loss of Col5a1 in Col1a2 + fibroblasts (Col5a1CKO). This model shows that an abnormal extracellular matrix (ECM) characterized by fibrillar disarray, altered mechanical properties, and decreased collagen deposition contribute to the wound healing defect. The cEDS animals exhibit decreased expression of epidermal genes and increased inflammation. Finally, we demonstrate that inhibiting mechanosensitive integrin signaling or by injecting wild-type (WT) fibroblasts into cEDS animals enhances epidermal gene expression, decreases inflammation, and augments wound closure. These findings suggest that cell delivery and/or blocking integrin signaling are potentially therapeutic strategies to rescue wound healing defects in cEDS.
Collapse
Affiliation(s)
- Kindra M Kelly-Scumpia
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Maani M Archang
- Bioengineering Department, University of California, Los Angeles, Los Angeles, CA, USA
- Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Dermatology, VA Greater Los Angeles Healthcare System-West Los Angeles, Los Angeles, CA 90073, USA
| | - Prabhat K Purbey
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tomohiro Yokota
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rimao Wu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jackie McCourt
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rachelle H Crosbie
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Philip O Scumpia
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Dermatology, VA Greater Los Angeles Healthcare System-West Los Angeles, Los Angeles, CA 90073, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Groh N, Magin TM. Pseudomonas-Derived Pyocyanin Links Oxidative Stress and Keratin 6 Expression to Wound Healing. J Invest Dermatol 2023; 143:1865-1867. [PMID: 37318404 DOI: 10.1016/j.jid.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 06/16/2023]
Affiliation(s)
- Niclas Groh
- Division of Cell and Developmental Biology, Institute of Biology, Leipzig University, Leipzig, Germany
| | - Thomas M Magin
- Division of Cell and Developmental Biology, Institute of Biology, Leipzig University, Leipzig, Germany.
| |
Collapse
|
6
|
Pfisterer K, Wielscher M, Samardzic D, Weinzettl P, Symmank D, Shaw LE, Campana R, Huang HJ, Farlik M, Bangert C, Vrtala S, Valenta R, Weninger W. Non-IgE-reactive allergen peptides deteriorate the skin barrier in house dust mite-sensitized atopic dermatitis patients. Front Cell Dev Biol 2023; 11:1240289. [PMID: 37675143 PMCID: PMC10478000 DOI: 10.3389/fcell.2023.1240289] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by type 2 cytokine-driven skin inflammation and epithelial barrier dysfunction. The latter is believed to allow the increased penetration of chemicals, toxins, and allergens into the skin. House dust mite allergens, particularly Der p 2, are important triggers in sensitized individuals with AD; the precise actions of these allergens in epithelial biology remain, however, incompletely understood. In this study, we compared the effects of the protein allergen Der p 2 and a mix of non-IgE-reactive Der p 2 peptides on skin cells using patch tests in AD patients and healthy participants. We then analyzed mRNA expression profiles of keratinocytes by single-cell RNA-sequencing. We report that existing barrier deficiencies in the non-lesional skin of AD patients allow deep penetration of Der p 2 and its peptides, leading to local microinflammation. Der p 2 protein specifically upregulated genes involved in the innate immune system, stress, and danger signals in suprabasal KC. Der p 2 peptides further downregulated skin barrier genes, in particular the expression of genes involved in cell-matrix and cell-cell adhesion. Peptides also induced genes involved in hyperproliferation and caused disturbances in keratinocyte differentiation. Furthermore, inflammasome-relevant genes and IL18 were overexpressed, while KRT1 was downregulated. Our data suggest that Der p 2 peptides contribute to AD initiation and exacerbation by augmenting hallmark features of AD, such as skin inflammation, barrier disruption, and hyperplasia of keratinocytes.
Collapse
Affiliation(s)
- Karin Pfisterer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Wielscher
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - David Samardzic
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Pauline Weinzettl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Dorte Symmank
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Lisa E. Shaw
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Raffaela Campana
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Huey-Jy Huang
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christine Bangert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Susanne Vrtala
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Heryanto YD, Imoto S. Identifying Key Regulators of Keratinization in Lung Squamous Cell Cancer Using Integrated TCGA Analysis. Cancers (Basel) 2023; 15:cancers15072066. [PMID: 37046726 PMCID: PMC10092975 DOI: 10.3390/cancers15072066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/25/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Keratinization is one of lung squamous cell cancer’s (LUSC) hallmark histopathology features. Epithelial cells produce keratin to protect their integrity from external harmful substances. In addition to their roles as cell protectors, recent studies have shown that keratins have important roles in regulating either normal cell or tumor cell functions. The objective of this study is to identify the genes and microRNAs (miRNAs) that act as key regulators of the keratinization process in LUSC. To address this goal, we classified LUSC samples from GDC-TCGA databases based on their keratinization molecular signatures. Then, we performed differential analyses of genes, methylation, and miRNA expression between high keratinization and low keratinization samples. By reconstruction and analysis of the differentially expressed genes (DEGs) network, we found that TP63 and SOX2 were the hub genes that were highly connected to other genes and displayed significant correlations with several keratin genes. Methylation analysis showed that the P63, P73, and P53 DNA-binding motif sites were significantly enriched for differentially methylated probes. We identified SNAI2, GRHL3, TP63, ZNF750, and FOXE1 as the top transcription factors associated with these binding sites. Finally, we identified 12 miRNAs that influence the keratinization process by using miRNA–mRNA correlation analysis.
Collapse
Affiliation(s)
- Yusri Dwi Heryanto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Correspondence:
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Laboratory of Sequence Analysis, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
8
|
Zhang X, Yu H, Wang N, Li C. Comprehensive analysis of long noncoding RNAs and lncRNA-mRNA networks in snakehead (Channa argus) response to Nocardia seriolae infection. FISH & SHELLFISH IMMUNOLOGY 2023; 133:108558. [PMID: 36690266 DOI: 10.1016/j.fsi.2023.108558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 06/17/2023]
Abstract
Evidence has been demonstrated that lncRNAs are involved in a variety of immune responses in vertebrate. It has been demonstrated that immune-related lncRNAs play vital functions in immune regulation against infections in teleost. Nocardia seriolae, as one of the Gram-positive bacteria, can cause chronic systemic granulomatous disease for snakehead (Channa argus). However, how lncRNAs function in the immune regulation process once snakehead was infected with N. seriolae infection has not been studied so far. Accordingly, transcription landscapes of lncRNAs and mRNAs in snakehead were investigated. A total of 1,991 lncRNA were obtained. Totally, we predicted 57,584 co-expression and 16,047 co-location lncRNA-mRNA pairs. To further analyze the potential function of these lncRNAs, GO enrichment analysis and KEGG signal pathways were performed on the target mRNAs of these differently expressed lncRNAs, suggesting that lncRNAs may play essential roles in modulating mRNA expression levels, and subsequently trigger downstream immune signaling pathways to regulate the immune response in snakehead. In addition, 9 DEmRNA and 3 lncRNAs were randomly selected for qRT-PCR analyzed, which confirmed the accuracy of transcriptome data. These results can provide novel knowledge about lncRNAs in immune responses process in snakehead, and can serve as important resources for further investigating the roles of lncRNAs during pathogen infections in teleost.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Haohui Yu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ningning Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
9
|
Raza A, Schulz BL, Nouwens A, Naseem MN, Kamran M, Mantilla Valdivieso EF, Kerr ED, Constantinoiu C, Jonsson NN, James P, Tabor AE. Application of quantitative proteomics to discover biomarkers for tick resistance in cattle. Front Immunol 2023; 14:1091066. [PMID: 36793724 PMCID: PMC9924087 DOI: 10.3389/fimmu.2023.1091066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/04/2023] [Indexed: 01/31/2023] Open
Abstract
Introduction Breeding for tick resistance is a sustainable alternative to control cattle ticks due to widespread resistance to acaricidal drugs and the lack of a protective vaccine. The most accurate method used to characterise the phenotype for tick resistance in field studies is the standard tick count, but this is labour-intensive and can be hazardous to the operator. Efficient genetic selection requires reliable phenotyping or biomarker(s) for accurately identifying tick-resistant cattle. Although breed-specific genes associated with tick resistance have been identified, the mechanisms behind tick resistance have not yet been fully characterised. Methods This study applied quantitative proteomics to examine the differential abundance of serum and skin proteins using samples from naïve tick-resistant and -susceptible Brangus cattle at two-time points following tick exposure. The proteins were digested into peptides, followed by identification and quantification using sequential window acquisition of all theoretical fragment ion mass spectrometry. Results Resistant naïve cattle had a suite of proteins associated with immune response, blood coagulation and wound healing that were significantly (adjusted P < 10- 5) more abundant compared with susceptible naïve cattle. These proteins included complement factors (C3, C4, C4a), alpha-1-acid glycoprotein (AGP), beta-2-glycoprotein-1, keratins (KRT1 & KRT3) and fibrinogens (alpha & beta). The mass spectrometry findings were validated by identifying differences in the relative abundance of selected serum proteins with ELISA. The proteins showing a significantly different abundance in resistant cattle following early and prolonged tick exposures (compared to resistant naïve) were associated with immune response, blood coagulation, homeostasis, and wound healing. In contrast, susceptible cattle developed some of these responses only after prolonged tick exposure. Discussion Resistant cattle were able to transmigrate immune-response related proteins towards the tick bite sites, which may prevent tick feeding. Significantly differentially abundant proteins identified in this research in resistant naïve cattle may provide a rapid and efficient protective response to tick infestation. Physical barrier (skin integrity and wound healing) mechanisms and systemic immune responses were key contributors to resistance. Immune response-related proteins such as C4, C4a, AGP and CGN1 (naïve samples), CD14, GC and AGP (post-infestation) should be further investigated as potential biomarkers for tick resistance.
Collapse
Affiliation(s)
- Ali Raza
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Benjamin L Schulz
- The University of Queensland, School of Chemistry and Molecular Biosciences, St. Lucia, QLD, Australia
| | - Amanda Nouwens
- The University of Queensland, School of Chemistry and Molecular Biosciences, St. Lucia, QLD, Australia
| | - Muhammad Noman Naseem
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Muhammad Kamran
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Emily F Mantilla Valdivieso
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Edward D Kerr
- The University of Queensland, School of Chemistry and Molecular Biosciences, St. Lucia, QLD, Australia
| | - Constantin Constantinoiu
- College of Public Health, Medical & Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Nicholas N Jonsson
- Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Peter James
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Ala E Tabor
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia.,The University of Queensland, School of Chemistry and Molecular Biosciences, St. Lucia, QLD, Australia
| |
Collapse
|
10
|
Kim N, Lee S, Lee S, Kang J, Choi Y, Park J, Park C, Khang D, Kim S. Portable Cold Atmospheric Plasma Patch-Mediated Skin Anti-Inflammatory Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202800. [PMID: 36180414 PMCID: PMC9731685 DOI: 10.1002/advs.202202800] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/27/2022] [Indexed: 05/29/2023]
Abstract
Although plasma is a promising technology in various fields, its clinical application is restricted by several limitations. A cold atmospheric plasma (CAP) patch is fabricated to help overcome hurdles, especially when treating skin diseases. This patch has surface dielectric barrier discharge, which generates reactive oxygen species (ROS) and reactive nitrogen species (RNS) on a flexible polymer film surface on which the embedded electrode induces a locally strong electric field. The effect of the CAP patch on psoriasis is also evaluated. The distinct characteristics of psoriasis between the lesion and non-lesion area allow the CAP patch to be suitable for only lesion area for its treatment. The CAP patch induces the opening of calcium channels in keratinocytes, thereby restoring abnormal keratinocyte differentiation and the collapse of the tight junction; thus, alleviating psoriatic symptoms. In addition, the favorable effect is due to the induction of ROS/RNS by the CAP patch, not the electric field generated during plasma generation. The findings indicate that the proposed portable CAP patch can help treat inflammatory skin disorders, especially psoriasis. As this can be used easily as a combination therapy with existing drugs, it may help reduce side effects caused by existing drugs.
Collapse
Affiliation(s)
- Namkyung Kim
- Cell & Matrix Research InstituteDepartment of PharmacologySchool of MedicineKyungpook National UniversityDaegu41944South Korea
| | - Seunghun Lee
- Department of Nano‐Bio ConvergenceNano Surface Materials DivisionKorea Institute of Materials ScienceChangwon51508South Korea
| | - Soyoung Lee
- Immunoregulatory Materials Research CenterKorea Research Institute of Bioscience and BiotechnologyJeongeup56212South Korea
| | - Jinjoo Kang
- Cell & Matrix Research InstituteDepartment of PharmacologySchool of MedicineKyungpook National UniversityDaegu41944South Korea
| | - Young‐Ae Choi
- Cell & Matrix Research InstituteDepartment of PharmacologySchool of MedicineKyungpook National UniversityDaegu41944South Korea
| | - Jeongsu Park
- Department of PhysiologySchool of MedicineGachon UniversityIncheon21999South Korea
| | - Chul‐Kyu Park
- Department of PhysiologySchool of MedicineGachon UniversityIncheon21999South Korea
| | - Dongwoo Khang
- Department of PhysiologySchool of MedicineGachon UniversityIncheon21999South Korea
| | - Sang‐Hyun Kim
- Cell & Matrix Research InstituteDepartment of PharmacologySchool of MedicineKyungpook National UniversityDaegu41944South Korea
| |
Collapse
|
11
|
Shutova MS, Boehncke WH. Mechanotransduction in Skin Inflammation. Cells 2022; 11:2026. [PMID: 35805110 PMCID: PMC9265324 DOI: 10.3390/cells11132026] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
In the process of mechanotransduction, the cells in the body perceive and interpret mechanical stimuli to maintain tissue homeostasis and respond to the environmental changes. Increasing evidence points towards dysregulated mechanotransduction as a pathologically relevant factor in human diseases, including inflammatory conditions. Skin is the organ that constantly undergoes considerable mechanical stresses, and the ability of mechanical factors to provoke inflammatory processes in the skin has long been known, with the Koebner phenomenon being an example. However, the molecular mechanisms and key factors linking mechanotransduction and cutaneous inflammation remain understudied. In this review, we outline the key players in the tissue's mechanical homeostasis, the available data, and the gaps in our current understanding of their aberrant regulation in chronic cutaneous inflammation. We mainly focus on psoriasis as one of the most studied skin inflammatory diseases; we also discuss mechanotransduction in the context of skin fibrosis as a result of chronic inflammation. Even though the role of mechanotransduction in inflammation of the simple epithelia of internal organs is being actively studied, we conclude that the mechanoregulation in the stratified epidermis of the skin requires more attention in future translational research.
Collapse
Affiliation(s)
- Maria S. Shutova
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| |
Collapse
|
12
|
Luo Y, Zhu Z, Li B, Bai X, Fang H, Qiao P, Chen J, Zhang C, Zhi D, Dang E, Wang G. Keratin 17 Promotes T Cell Response in Allergic Contact Dermatitis by Upregulating C-C Motif Chemokine Ligand 20. Front Immunol 2022; 13:764793. [PMID: 35178048 PMCID: PMC8845002 DOI: 10.3389/fimmu.2022.764793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/14/2022] [Indexed: 12/04/2022] Open
Abstract
Allergic contact dermatitis (ACD) is a delayed-type hypersensitivity response to skin contact allergens in which keratinocytes are critical in the initiation of early responses. Keratin 17 (K17) is a cytoskeletal protein inducible under stressful conditions and regulates multiple cellular processes, especially in skin inflammatory diseases; however, knowledge regarding its contribution to ACD pathogenesis remains ill defined. In the present study, we clarified the proinflammatory role of K17 in an oxazolone (OXA)-induced contact hypersensitivity (CHS) murine model and identified the underlying molecular mechanisms. Our results showed that K17 was highly expressed in the lesional skin of ACD patients and OXA-induced CHS mice. Mice lacking K17 exhibited alleviated OXA-induced skin inflammation, including milder ear swelling, a reduced frequency of T cell infiltration, and decreased inflammatory cytokine levels. In vitro, K17 stimulated and activated human keratinocytes to produce plenty of proinflammatory mediators, especially the chemokine CCL20, and promoted keratinocyte-mediated T cell trafficking. The neutralization of CCL20 with a CCL20-neutralizing monoclonal antibody significantly alleviated OXA-induced skin inflammation in vivo. Moreover, K17 could translocate into the nucleus of activated keratinocytes through a process dependent on the nuclear-localization signal (NLS) and nuclear-export signal (NES) sequences, thus facilitating the activation and nuclear translocation of signal transducer and activator of transcription 3 (STAT3), further promoting the production of CCL20 and T cell trafficking to the lesional skin. Taken together, these results highlight the novel roles of K17 in driving allergen-induced skin inflammation and suggest targeting K17 as a potential strategy for ACD.
Collapse
Affiliation(s)
- Yixin Luo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhenlai Zhu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Bing Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaocui Bai
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Pei Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiaoling Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chen Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dalong Zhi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
13
|
Wang ML, Rajpar I, Ruggiero NA, Fertala J, Steplewski A, Beredjiklian PK, Rivlin MR, Chen Y, Feldman GJ, Fertala A, Tomlinson RE. Circulating inflammatory cytokines alter transcriptional activity within fibrotic tissue of Dupuytren's disease patients. J Orthop Res 2022; 40:738-749. [PMID: 33913534 DOI: 10.1002/jor.25059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/31/2021] [Accepted: 04/19/2021] [Indexed: 02/04/2023]
Abstract
Dupuytren's disease is a benign fibroproliferative disorder of the hand that results in disabling digital contractures that impair function and diminish the quality of life. The incidence of this disease has been correlated with chronic inflammatory states, but any direct association between inflammatory cytokines and Dupuytren's disease is not known. We hypothesized that advanced fibroproliferation is associated with increased levels of circulating inflammatory cytokines. Blood and fibrotic cord tissue were collected preoperatively from patients with severe contracture and control patients. Blood plasma concentrations of known inflammatory cytokines were evaluated using a multiplex immunoassay. Proteins from the cord tissue were analyzed by RNA sequencing and immunohistochemistry. Moreover, collagen-rich cords were analyzed using Fourier-transform infrared spectroscopy. The results indicate that patients exhibited significantly elevated circulating inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin (IL)-2, and IL-12p70, as compared with controls. Similarly, IL-4 and IL-13 were detected significantly more frequently in Dupuytren's disease as compared with control. RNA sequencing revealed 5311 differentially expressed genes and distinct clustering between diseased and control samples. In addition to increased expression of genes associated with fibroproliferation, we also observed upregulation of transcripts activated by inflammatory cytokines, including prolactin inducible protein and keratin intermediate filaments. IL-2, but not TNF-α, was detected in fibrotic cord tissue by immunohistochemistry. Finally, spectroscopic assays revealed a significant reduction of the collagen content and alterations of collagen cross-linking within the Dupuytren's disease tissues. In total, our results illustrate that patients with severe Dupuytren's disease exhibit substantially elevated circulating inflammatory cytokines that may drive fibroproliferation. Clinical Significance: The results from this study establish the basis for a specific cytokine profile that may be useful for diagnostic testing and therapeutic intervention in Dupuytren's disease.
Collapse
Affiliation(s)
- Mark L Wang
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Division of Hand Surgery, The Rothman Orthopaedic Institute, Philadelphia, Pennsylvania, USA
| | - Ibtesam Rajpar
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Nicholas A Ruggiero
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jolanta Fertala
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Andrzej Steplewski
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Pedro K Beredjiklian
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Division of Hand Surgery, The Rothman Orthopaedic Institute, Philadelphia, Pennsylvania, USA
| | - Michael R Rivlin
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Division of Hand Surgery, The Rothman Orthopaedic Institute, Philadelphia, Pennsylvania, USA
| | - Yong Chen
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, New Jersey, USA
| | - George J Feldman
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Andrzej Fertala
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ryan E Tomlinson
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Konop M, Rybka M, Drapała A. Keratin Biomaterials in Skin Wound Healing, an Old Player in Modern Medicine: A Mini Review. Pharmaceutics 2021; 13:2029. [PMID: 34959311 PMCID: PMC8705570 DOI: 10.3390/pharmaceutics13122029] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/20/2021] [Accepted: 11/26/2021] [Indexed: 12/26/2022] Open
Abstract
Impaired wound healing is a major medical problem. To solve it, researchers around the world have turned their attention to the use of tissue-engineered products to aid in skin regeneration in case of acute and chronic wounds. One of the primary goals of tissue engineering and regenerative medicine is to develop a matrix or scaffold system that mimics the structure and function of native tissue. Keratin biomaterials derived from wool, hair, and bristle have been the subjects of active research in the context of tissue regeneration for over a decade. Keratin derivatives, which can be either soluble or insoluble, are utilized as wound dressings since keratins are dynamically up-regulated and needed in skin wound healing. Tissue biocompatibility, biodegradability, mechanical durability, and natural abundance are only a few of the keratin biomaterials' properties, making them excellent wound dressing materials to treat acute and chronic wounds. Several experimental and pre-clinical studies described the beneficial effects of the keratin-based wound dressing in faster wound healing. This review focuses exclusively on the biomedical application of a different type of keratin biomaterials as a wound dressing in pre-clinical and clinical conditions.
Collapse
Affiliation(s)
- Marek Konop
- Laboratory of Center for Preclinical Research, Department of Experimental Physiology and Pathophysiology, Medical University of Warsaw, 02-106 Warsaw, Poland; (M.R.); (A.D.)
| | | | | |
Collapse
|
15
|
Lee KH, Do HK, Kim DY, Kim W. Impact of chlorogenic acid on modulation of significant genes in dermal fibroblasts and epidermal keratinocytes. Biochem Biophys Res Commun 2021; 583:22-28. [PMID: 34715497 DOI: 10.1016/j.bbrc.2021.10.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 11/25/2022]
Abstract
Chlorogenic acid is one of the most abundant polyphenols found in human diet. It is well-documented that chlorogenic acid has a significant impact on human cells, especially in the regulation of inflammation and metabolic processes. However, its role in regulating skin functions, especially with respect to the dermal collagen network or epidermal skin barrier, has not yet been elucidated. Here, we report that chlorogenic acid treatment can induce production of procollagen type I in human dermal fibroblast, Hs68 cell lines. Moreover, this treatment can stimulate upregulation of skin barrier genes, including the ones encoding filaggrin (FLG), involucrin (IVL), and envoplakin (EVPL), in epidermal keratinocytes. Chlorogenic acid also triggered a multifaceted response in the cytokine profile of keratinocytes. Therefore, we suggest that chlorogenic acid can be used to restore the impaired dermal matrix network as well as the epidermal skin barrier.
Collapse
Affiliation(s)
- Kyung-Ha Lee
- Department of Cosmetic Science and Technology, Daegu Haany University, Gyeongsan, 38610, South Korea
| | - Hwan-Kwon Do
- Department of Physical Medicine and Rehabilitation, Inje University College of Medicine, Haeundae Paik Hospital, Busan, 48108, South Korea
| | - Do-Yeon Kim
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, 41940, South Korea
| | - Wanil Kim
- Department of Biochemistry, Department of Convergence Medical Science, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, South Korea.
| |
Collapse
|
16
|
Guan K, Li H, Chen H, Qi X, Wang R, Ma Y. TMT-based quantitative proteomics analysis reveals the effect of bovine derived MFG-E8 against oxidative stress on rat L6 cells. Food Funct 2021; 12:7310-7320. [PMID: 34169949 DOI: 10.1039/d1fo01135a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sarcopenia is an aging-associated oxidative stress-induced mitochondrial dysfunction characterized by a decline in skeletal muscle mass, strength and function. Milk fat globule-EGF factor 8 (MFG-E8) is a secreted matrix glycoprotein that plays a crucial role in regulating tissue homeostasis and protecting against skeletal muscle injury. To explore the molecular mechanism of MFG-E8 in ameliorating the rotenone (Rot)-induced L6 skeletal muscle cell oxidative stress injury, differential proteomics of inner L6 cells was conducted. Tandem mass tag (TMT) labeling combined with mass spectrometry (MS) was performed to find associations among control, Rot and Rot + MFG-E8 groups. Over 3248 proteins were identified in the L6 cells. A total of 639 significantly differential proteins were identified, including 294 up-regulated proteins (>1.2 fold) and 345 down-regulated proteins (<0.83 fold) after the exogenous intervention of MFG-E8. Based on the analysis of Gene Ontology (GO), STRING and KEGG databases, MFG-E8 relieves oxidative stress induced-L6 cell damage by regulating the expression of these differential proteins mainly via carbon metabolism, glutathione metabolism and mitochondria-mediated metabolic pathways, e.g. carbohydrate, lipid and amino acid metabolism. Furthermore, to verify the protective effect of MFG-E8 on oxidative stress injured L6 cells, the levels of intracellular reactive oxygen species (ROS), nicotinamide adenine dinucleotide/reduced nicotinamide adenine dinucleotide (NAD+/NADH) contents and the protein expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) were detected.
Collapse
Affiliation(s)
- Kaifang Guan
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150090, Heilongjiang, PR China.
| | | | | | | | | | | |
Collapse
|
17
|
Doukas D, Liakou Z, Koukoulis GK, Tontis D. Immunohistochemical Expression of Keratins in Normal Ovine Skin and in Chronic Dermatitis due to Sarcoptes scabiei. J Comp Pathol 2021; 183:63-71. [PMID: 33714434 DOI: 10.1016/j.jcpa.2021.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/30/2020] [Accepted: 01/13/2021] [Indexed: 12/27/2022]
Abstract
Hyperproliferation of epidermal keratinocytes is a major histopathological feature of chronic Sarcoptes scabiei dermatitis. We investigated the immunohistochemical expression of several keratins in scabietic dermatitis in sheep and in the skin of healthy sheep, using a panel of commercially available anti-human antibodies for keratins. Keratins AE1/AE3 and 34BE12 were expressed in all epithelial structures in healthy skin. Keratin MNF116 was expressed in the stratum basale and in the three lowest layers of the stratum spinosum, in follicular epithelium and in apocrine glands. Keratin K5/6 expression was seen in the stratum basale, in the two lowest cell layers of the stratum spinosum, in the outer root sheath of hair follicles and in myoepithelial cells of apocrine glands. K14 expression was observed in the stratum basale, in locally extensive regions of the two lowest cell layers of the stratum spinosum, in the outer root sheath of hair follicles and in sebaceous glands. Immunolabelling of K19 antigen was confined to apocrine glands. In scabietic skin, immunolabelling of keratin 34BE12 was seen in all layers of hyperplastic stratum spinosum and stratum granulosum but was restricted to some locally extensive regions in hyperkeratotic and parakeratotic stratum corneum. Keratin MNF116 was widely labelled in all layers of hyperplastic stratum spinosum and stratum granulosum. There was expansive labelling of K5/6 keratin in all layers of hyperplastic stratum spinosum and in locally extensive regions of stratum granulosum, as well as in hyperkeratotic or parakeratotic stratum corneum. Expansive labelling of K14 keratin was detected in all layers of hyperplastic stratum spinosum and in the layers of the hyperplastic stratum granulosum. K5/6 and K14 keratins were also labelled in the inner root sheath of occasional hair follicles.
Collapse
Affiliation(s)
- Dimitrios Doukas
- Laboratory of Veterinary Pathology, Faculty of Veterinary Science.
| | - Zoi Liakou
- Laboratory of Veterinary Pathology, Faculty of Veterinary Science
| | - George K Koukoulis
- Laboratory of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Karditsa, Larissa, Greece
| | - Dimitrios Tontis
- Laboratory of Veterinary Pathology, Faculty of Veterinary Science
| |
Collapse
|
18
|
Zhang L, Zeng X, Li Y, Chen S, Tang L, Wang N, Yang X, Lin M. Keratin 1 attenuates hypoxic pulmonary artery hypertension by suppressing pulmonary artery media smooth muscle expansion. Acta Physiol (Oxf) 2021; 231:e13558. [PMID: 32920982 DOI: 10.1111/apha.13558] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 09/03/2020] [Accepted: 09/05/2020] [Indexed: 12/13/2022]
Abstract
AIM Abnormally activated vascular smooth muscle cells are key factors in pulmonary artery remodelling (PAR) and pulmonary artery hypertension (PAH). Keratin 1 is involved in inflammatory diseases; however, its role in PAH is unknown. We speculated that keratin 1 could regulate PASMCs and prevent PAH. METHODS Rats were exposed to hypoxia (10% O2 ) or MCT (50 mg/kg, intraperitoneal injection) or treated with AAV6 virus. PAR was measured through HE and Masson staining. PASMC activities were measured using MTS assay, EdU and Western blot analyses after cell knockdown with siRNAs or overexpression with Krt1 vectors. RESULTS 1. Hypoxic PAR was associated with a decrease in keratin 1, especially in PASMCs. 2. Keratin 1 knockdown led to cell proliferation, migration and contraction to synthetic transformation, while keratin 1 overexpression attenuated hypoxia-induced changes in PASMCs. 3. Decreased keratin 1 induced TLR7 upregulation and mediated increases in the inflammatory factors S100a8 and S100a9. 4. Keratin 1 overexpression reduced the inflammatory factor expression induced by TLR7 activation. 5. Further studies demonstrated that keratin 1 expression was negatively correlated with pulmonary vascular pressure following prolonged hypoxia. 6. Pre-treatment with keratin 1 decreased pulmonary artery pressure and the right heart hypertrophy index and alleviated PAR in two model rats. 7. Keratin 1 exhibited a hypermethylation status in hypoxic pulmonary arteries in the sequencing. Hypoxia-induced decrease in keratin 1 expression was associated with Dnmt1 upregulation induced by YY1 downregulation in PASMCs. CONCLUSION This study suggests that keratin 1 regulates PASMC expansion and has a preventive effect on PAH.
Collapse
Affiliation(s)
- Li Zhang
- Department of Physiology & Pathophysiology The School of Basic Medical SciencesFujian Medical University Fuzhou China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases Fujian Medical University Fuzhou China
| | - Xi‐Xi Zeng
- Department of Physiology & Pathophysiology The School of Basic Medical SciencesFujian Medical University Fuzhou China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases Fujian Medical University Fuzhou China
- Department of Clinical Laboratory the Affiliated Hospital of Jiujiang University Jiujiang China
| | - Yu‐Mei Li
- Department of Physiology & Pathophysiology The School of Basic Medical SciencesFujian Medical University Fuzhou China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases Fujian Medical University Fuzhou China
- Fujian Center for Safety Evaluation of New Drug Fujian Medical University Fuzhou China
| | - Shao‐Kun Chen
- Department of Physiology & Pathophysiology The School of Basic Medical SciencesFujian Medical University Fuzhou China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases Fujian Medical University Fuzhou China
| | - Li‐Yu Tang
- Department of Physiology & Pathophysiology The School of Basic Medical SciencesFujian Medical University Fuzhou China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases Fujian Medical University Fuzhou China
| | - Nan Wang
- Department of Physiology & Pathophysiology The School of Basic Medical SciencesFujian Medical University Fuzhou China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases Fujian Medical University Fuzhou China
| | - Xi Yang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases Fujian Medical University Fuzhou China
- Fujian Center for Safety Evaluation of New Drug Fujian Medical University Fuzhou China
| | - Mo‐Jun Lin
- Department of Physiology & Pathophysiology The School of Basic Medical SciencesFujian Medical University Fuzhou China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases Fujian Medical University Fuzhou China
| |
Collapse
|
19
|
Trittmann JK. Keratin 1: A negative regulator of inflammation and potential treatment for pulmonary arterial hypertension. Acta Physiol (Oxf) 2021; 231:e13594. [PMID: 33280251 DOI: 10.1111/apha.13594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jennifer K Trittmann
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
20
|
Bansal R, Khan MM, Dasari S, Verma I, Goodlett DR, Manes NP, Nita-Lazar A, Sharma SP, Kumar A, Singh N, Chakraborti A, Gupta V, Dogra MR, Ram J, Gupta A. Proteomic profile of vitreous in patients with tubercular uveitis. Tuberculosis (Edinb) 2021; 126:102036. [PMID: 33359883 PMCID: PMC11005023 DOI: 10.1016/j.tube.2020.102036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/31/2020] [Accepted: 11/29/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To elucidate disease-specific host protein profile in vitreous fluid of patients with intraocular inflammation due to tubercular uveitis (TBU). METHODS Vitreous samples from 13 patients with TBU (group A), 7 with non-TBU (group B) and 9 with no uveitis (group C) were analysed by shotgun proteomics using Liquid Chromatography Tandem Mass Spectrometry (LC-MS/MS). Differentially expressed proteins (DEPs) were subjected to pathway analysis using WEB-based Gene SeT Analysis Toolkit software. RESULTS Compared to control groups (B + C combined), group A (TBU) displayed 32 (11 upregulated, 21 downregulated) DEPs, which revealed an upregulation of coagulation cascades, complement and classic pathways, and downregulation of metabolism of carbohydrates, gluconeogenesis, glucose metabolism and glycolysis/gluconeogenesis pathways. When compared to group B (non-TBU) alone, TBU displayed 58 DEPs (21 upregulated, 37 downregulated), with an upregulation of apoptosis, KRAS signaling, diabetes pathways, classic pathways, and downregulation of MTORC1 signaling, glycolysis/gluconeogenesis, and glucose metabolism. CONCLUSION This differential protein profile provides novel insights into the molecular mechanisms of TBU and a baseline to explore vitreous biomarkers to differentiate TBU from non-TBU, warranting future studies to identify and validate them as a diagnostic tool in TBU. The enriched pathways generate interesting hypotheses and drive further research.
Collapse
Affiliation(s)
- Reema Bansal
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Mohd M Khan
- University of Maryland, School of Medicine, Baltimore, MD, USA; Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.
| | - Indu Verma
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | | | - Nathan P Manes
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Aleksandra Nita-Lazar
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Surya P Sharma
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Aman Kumar
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Nirbhai Singh
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Anuradha Chakraborti
- Department of Experimental Medicine & Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Vishali Gupta
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - M R Dogra
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Jagat Ram
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Amod Gupta
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
21
|
Zhang X, Yin M, Zhang LJ. Keratin 6, 16 and 17-Critical Barrier Alarmin Molecules in Skin Wounds and Psoriasis. Cells 2019; 8:E807. [PMID: 31374826 PMCID: PMC6721482 DOI: 10.3390/cells8080807] [Citation(s) in RCA: 255] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/17/2022] Open
Abstract
Located at the skin surface, keratinocytes (KCs) are constantly exposed to external stimuli and are the first responders to invading pathogens and injury. Upon skin injury, activated KCs secrete an array of alarmin molecules, providing a rapid and specific innate immune response against danger signals. However, dysregulation of the innate immune response of KCs may lead to uncontrolled inflammation and psoriasis pathogenesis. Keratins (KRT) are the major structural intermediate filament proteins in KCs and are expressed in a highly specific pattern at different differentiation stages of KCs. While KRT14-KRT5 is restricted to basal proliferative KCs, and KRT10-KRT1 is restricted to suprabasal differentiated KCs in normal skin epidermis, the wound proximal KCs downregulate KRT10-K1 and upregulate KRT16/KRT17-KRT6 upon skin injury. Recent studies have recognized KRT6/16/17 as key early barrier alarmins and upregulation of these keratins alters proliferation, cell adhesion, migration and inflammatory features of KCs, contributing to hyperproliferation and innate immune activation of KCs in response to an epidermal barrier breach, followed by the autoimmune activation of T cells that drives psoriasis. Here, we have reviewed how keratins are dysregulated during skin injury, their roles in wound repairs and in initiating the innate immune system and the subsequent autoimmune amplification that arises in psoriasis.
Collapse
Affiliation(s)
- Xiaowei Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Meimei Yin
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Ling-Juan Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
22
|
Menditti D, Laino L, DI Domenico M, Troiano G, Guglielmotti M, Sava S, Mezzogiorno A, Baldi A. Cysts and Pseudocysts of the Oral Cavity: Revision of the Literature and a New Proposed Classification. In Vivo 2018; 32:999-1007. [PMID: 30150421 DOI: 10.21873/invivo.11340] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 06/16/2018] [Accepted: 06/21/2018] [Indexed: 01/19/2023]
Abstract
This article includes a comprehensive and up-to-date review on the cysts of the oral cavity. Several classifications of odontogenic (OC) and non-odontogenic (non-OC) oral cysts and the surrounding regions have been proposed. We suggest a new critical classification based on an established relationship between anatomical area, histological origin and clinical behavior (frequency, rate of recurrence, malignant potential). Moreover, the differential cytokeratin (CKs) expression of the various cysts is reported as epithelium-specific markers of differential diagnosis. Finally, issues related to differential diagnosis and therapeutic approaches of the cysts included in the two groups are described.
Collapse
Affiliation(s)
- Dardo Menditti
- Department of Dentistry, Orthodontics and Oral Surgery, University of Campania, Naples, Italy
| | - Luigi Laino
- Department of Clinical and Experimental Medicine, University of Campania, Naples, Italy
| | - Marina DI Domenico
- Department of General Pathology and Biochemistry, University of Campania, Naples, Italy
| | - Giuseppe Troiano
- Department of Clinical and Experimental Medicine, University of Campania, Naples, Italy
| | - Mario Guglielmotti
- Department of Dentistry, Orthodontics and Oral Surgery, University of Campania, Naples, Italy
| | - Sara Sava
- Department of Dentistry, Orthodontics and Oral Surgery, University of Campania, Naples, Italy
| | - Antonio Mezzogiorno
- Department of Mental Health and Physics, Preventive Medicine, University of Campania, Naples, Italy
| | - Alfonso Baldi
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania, Naples, Italy
| |
Collapse
|
23
|
Castela E, Tulic MK, Rozières A, Bourrat E, Nicolas JF, Kanitakis J, Vabres P, Bessis D, Mazereeuw J, Morice-Picard F, Baty D, Berard F, Lacour JP, Passeron T, Chiaverini C. Epidermolysis bullosa simplex generalized severe induces a T helper 17 response and is improved by apremilast treatment. Br J Dermatol 2018; 180:357-364. [PMID: 29932457 DOI: 10.1111/bjd.16897] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Epidermolysis bullosa simplex generalized severe (EBS-gen sev) is a genetic disorder caused by mutation in the KRT5 or KRT14 genes. Although it is usually considered a mechanical disease, recent data argue for additional inflammatory mechanisms. OBJECTIVES To assess the inflammation in the skin of patients with EBS-gen sev. METHODS A first immunohistochemical retrospective study was performed on frozen skin samples from 17 patients with EBS-gen sev. A second multicentre prospective study was conducted on 10 patients with severe EBS-gen sev. Blister fluid and epidermis were processed for immunochemical analysis and quantitative real-time polymerase chain reaction. Cytokine expression was analysed in blister fluid and compared with that in controls. RESULTS Histological analysis showed a constant dermal perivascular CD4+ lymphocyte infiltrate in skin biopsies of both blister (n = 17) and rubbed skin (n = 5), an epidermal infiltration of neutrophils and eosinophils in 70% of cases, and increased immunostaining for CXCL9 and CXCL10 in blistering skin. High levels of T helper 17 cytokines were detected in lesional skin. Three adult patients with EBS-gen sev were treated with apremilast, with a dramatic improvement of skin blistering and good tolerance. CONCLUSIONS Our study demonstrates the importance of inflammation in patients with EBS-gen sev and underlines the key role for T helper 17 cells in its pathogenesis. In addition, this study provides promising new therapeutic approaches for this disabling disorder.
Collapse
Affiliation(s)
- E Castela
- Department of Dermatology, CHU de Nice, Hôpital Archet 2, 151 Route de Saint Antoine de Ginestière, 06202 Nice CEDEX 2, France.,INSERM U1111-CIRI851, Université Lyon 1, Lyon, France
| | - M K Tulic
- INSERM U1065, Team 12, C3M, Nice, France
| | - A Rozières
- INSERM U1111-CIRI851, Université Lyon 1, Lyon, France
| | - E Bourrat
- MAGEC, Saint-Louis Hospital, Paris, France
| | - J-F Nicolas
- INSERM U1111-CIRI851, Université Lyon 1, Lyon, France.,Department of Allergology and Clinical Immunology, Hospices Civils de Lyon, Lyon, France
| | - J Kanitakis
- Department of Dermatology , Hospices Civils de Lyon, Lyon, France.,Department of Pathology, Hospices Civils de Lyon, Lyon, France
| | - P Vabres
- Department of Dermatology, CHU de Dijon, Dijon, France
| | - D Bessis
- Department of Dermatology, CHU de Montpellier, Montpellier, France
| | | | | | - D Baty
- Scottish Molecular Genetics Consortium, Ninewells Hospital, Dundee, U.K
| | - F Berard
- Department of Allergology and Clinical Immunology, Hospices Civils de Lyon, Lyon, France
| | - J-P Lacour
- Department of Dermatology, CHU de Nice, Hôpital Archet 2, 151 Route de Saint Antoine de Ginestière, 06202 Nice CEDEX 2, France.,CREBHN, CHU de Nice, Nice, France
| | - T Passeron
- Department of Dermatology, CHU de Nice, Hôpital Archet 2, 151 Route de Saint Antoine de Ginestière, 06202 Nice CEDEX 2, France.,INSERM U1065, Team 12, C3M, Nice, France
| | - C Chiaverini
- Department of Dermatology, CHU de Nice, Hôpital Archet 2, 151 Route de Saint Antoine de Ginestière, 06202 Nice CEDEX 2, France.,CREBHN, CHU de Nice, Nice, France
| |
Collapse
|
24
|
Sequeira I, Neves JF, Carrero D, Peng Q, Palasz N, Liakath-Ali K, Lord GM, Morgan PR, Lombardi G, Watt FM. Immunomodulatory role of Keratin 76 in oral and gastric cancer. Nat Commun 2018; 9:3437. [PMID: 30143634 PMCID: PMC6109110 DOI: 10.1038/s41467-018-05872-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/26/2018] [Indexed: 11/09/2022] Open
Abstract
Keratin 76 (Krt76) is expressed in the differentiated epithelial layers of skin, oral cavity and squamous stomach. Krt76 downregulation in human oral squamous cell carcinomas (OSCC) correlates with poor prognosis. We show that genetic ablation of Krt76 in mice leads to spleen and lymph node enlargement, an increase in regulatory T cells (Tregs) and high levels of pro-inflammatory cytokines. Krt76-/- Tregs have increased suppressive ability correlated with increased CD39 and CD73 expression, while their effector T cells are less proliferative than controls. Loss of Krt76 increases carcinogen-induced tumours in tongue and squamous stomach. Carcinogenesis is further increased when Treg levels are elevated experimentally. The carcinogenesis response includes upregulation of pro-inflammatory cytokines and enhanced accumulation of Tregs in the tumour microenvironment. Tregs also accumulate in human OSCC exhibiting Krt76 loss. Our study highlights the role of epithelial cells in modulating carcinogenesis via communication with cells of the immune system.
Collapse
Affiliation(s)
- Inês Sequeira
- Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Joana F Neves
- Department of Experimental Immunobiology, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Dido Carrero
- Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Qi Peng
- Immunoregulation Laboratory, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Natalia Palasz
- Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Kifayathullah Liakath-Ali
- Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.,Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, 265 Campus Drive, CA, 94305-5453, USA
| | - Graham M Lord
- Department of Experimental Immunobiology, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Peter R Morgan
- Department of Mucosal and Salivary Biology, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Giovanna Lombardi
- Immunoregulation Laboratory, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Fiona M Watt
- Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.
| |
Collapse
|
25
|
Keratin gene mutations influence the keratinocyte response to DNA damage and cytokine induced apoptosis. Arch Dermatol Res 2017. [DOI: 10.1007/s00403-017-1757-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
26
|
Varga N, Mózes J, Keegan H, White C, Kelly L, Pilkington L, Benczik M, Zsuzsanna S, Sobel G, Koiss R, Babarczi E, Nyíri M, Kovács L, Attila S, Kaltenecker B, Géresi A, Kocsis A, O'Leary J, Martin CM, Jeney C. The Value of a Novel Panel of Cervical Cancer Biomarkers for Triage of HPV Positive Patients and for Detecting Disease Progression. Pathol Oncol Res 2016; 23:295-305. [PMID: 27497597 DOI: 10.1007/s12253-016-0094-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/14/2016] [Indexed: 01/12/2023]
Abstract
In the era of primary vaccination against HPV and at the beginning of the low prevalence of cervical lesions, introduction of screening methods that can distinguish between low- and high-grade lesions is necessary in order to maintain the positive predictive value of screening. This case-control study included 562 women who attended cervical screening or were referred for colposcopy and 140 disease free controls, confirmed by histology and/or cytology. The cases were stratified by age. Using routine exfoliated liquid based cytological samples RT-PCR measurements of biomarker genes, high-risk HPV testing and liquid based cytology were performed and used to evaluate different testing protocols including sets of genes/tests with different test cut-offs for the diagnostic panels. Three new panels of cellular biomarkers for improved triage of hrHPV positive women (diagnostic panel) and for prognostic assessment of CIN lesions were proposed. The diagnostic panel (PIK3AP1, TP63 and DSG3) has the potential to distinguish cytologically normal hrHPV+ women from hrHPV+ women with CIN2+. The prognostic gene panels (KRT78, MUC5AC, BPIFB1 and CXCL13, TP63, DSG3) have the ability to differentiate hrHPV+ CIN1 and carcinoma cases. The diagnostic triage panel showed good likelihood ratios for all age groups. The panel showed age-unrelated performance and even better diagnostic value under age 30, a unique feature among the established cervical triage tests. The prognostic gene-panels demonstrated good discriminatory power and oncogenic, anti-oncogenic grouping of genes. The study highlights the potential for the gene expression panels to be used for diagnostic triage and lesion prognostics in cervical cancer screening.
Collapse
Affiliation(s)
- Norbert Varga
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Johanna Mózes
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Helen Keegan
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Christine White
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Lynne Kelly
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Loretto Pilkington
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Márta Benczik
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Schaff Zsuzsanna
- 2nd Department of Pathology, Semmelweis University, Üllöi út 93, Budapest, 1091, Hungary
| | - Gábor Sobel
- 2nd Department of Obstetrics and Gynecology, Semmelweis University, Üllői út 78/a, Budapest, 1082, Hungary
| | - Róbert Koiss
- Department of Gynecology-Oncology, United Hospital of St. Stephan and Laszlo, Nagyvárad tér 1, Budapest, 1087, Hungary
| | - Edit Babarczi
- Department of Gynecology-Oncology, United Hospital of St. Stephan and Laszlo, Nagyvárad tér 1, Budapest, 1087, Hungary
| | - Miklos Nyíri
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Laura Kovács
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Sebe Attila
- Institute of Pathophysiology, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | | | - Adrienn Géresi
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Adrienn Kocsis
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - John O'Leary
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Cara M Martin
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Csaba Jeney
- Department of Medical Microbiology, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary.
| |
Collapse
|
27
|
Wei X, Fricker K, Enk AH, Hadaschik EN. Altered expression of keratin 14 in lesional epidermis of autoimmune skin diseases. Int J Dermatol 2015; 55:620-8. [PMID: 26547800 DOI: 10.1111/ijd.13011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 12/10/2014] [Accepted: 12/28/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND Keratin 14 (K14) is an intermediate filament protein that is mainly expressed in the basal layer of healthy stratified epithelia. K14 has been identified as an autoantigen in the autoimmune-mediated skin disease of Scurfy mice and patients with the "immune dysregulation polyendocrinopathy, enteropathy, and X-linked" syndrome. OBJECTIVES To examine whether K14 is a target protein in autoimmune skin diseases (ASD), we analyzed the expression pattern of K14 in lesional skin of patients with lichen ruber, cutaneous lupus erythematosus, dermatomyositis, graft-versus-host disease, psoriasis, and pemphigus vulgaris, and evaluated the reactivity of patient sera with recombinantly expressed and epidermis-derived K14. METHODS K14 expression was analyzed by immunohistochemistry on paraffin-embedded tissue sections of 17 healthy individuals and 58 patients with ASD. Sera from 10 healthy individuals and 41 patients with ASD were analyzed by Western blot for the presence of anti-K14 autoantibodies. RESULTS In skin of patients with ASD, K14 expression is retained in suprabasal layers. In ASD with interface dermatitis, we observed focal loss of K14 within the basal layer and in hair follicles as well. A scattered dot-like K14 staining is seen in papillary dermis, most prominently in cutaneous lupus erythematosus and lichen ruber. Using Western blot, we show that sera of different patients with ASD recognize recombinantly expressed K14. CONCLUSION We show focal loss of K14 in the basal epidermis correlating with interface dermatitis and hair follicle involvement. Moreover, enhanced reactivity of sera of patients with atopic dermatitis with K14 suggests K14 may function as an autoantigen in ASD.
Collapse
Affiliation(s)
- Xiaoying Wei
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany.,Department of Pathology, Affiliated Zhong-Da Hospital, Southeast University, Nanjing, China
| | - Katharina Fricker
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Alexander H Enk
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Eva N Hadaschik
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
28
|
Abstract
The autoimmune regulator (Aire) was initially identified as the gene causing multiorgan system autoimmunity in humans, and deletion of this gene in mice also resulted in organ-specific autoimmunity. Aire regulates the expression of tissue-specific antigens (TSAs) in medullary thymic epithelial cells (mTECs), which play a critical role in the negative selection of autoreactive T cells and the generation of regulatory T cells. More recently, the role of Aire in the development of mTECs has helped elucidate its ability to present the spectrum of TSAs needed to prevent autoimmunity. Molecular characterization of the functional domains of Aire has revealed multiple binding partners that assist Aire's function in altering gene transcription and chromatin remodeling. These recent advances have further highlighted the importance of Aire in central tolerance.
Collapse
Affiliation(s)
- Alice Chan
- Diabetes Center, University of California, San Francisco, San Francisco, California
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Mark S. Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, California
| |
Collapse
|
29
|
Okano J, Kojima H, Katagi M, Nakae Y, Terashima T, Nakagawa T, Kurakane T, Okamoto N, Morohashi K, Maegawa H, Udagawa J. Epidermis-dermis junction as a novel location for bone marrow-derived cells to reside in response to ionizing radiation. Biochem Biophys Res Commun 2015; 461:695-701. [PMID: 25922286 DOI: 10.1016/j.bbrc.2015.04.094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 04/19/2015] [Indexed: 11/17/2022]
Abstract
Bone marrow-derived cells (BMDCs) can migrate into the various organs in the mice irradiated by ionizing radiation (IR). However, it may not be the case in the skin. While IR is used for bone marrow (BM) transplantation, studying with the epidermal sheets demonstrated that the BMDC recruitment is extraordinarily rare in epidermis in the mouse. Herein, using the chimera mice with BM from green fluorescent protein (GFP) transgenic mice, we simply examined if BMDCs migrate into any layers in the total skin, as opposed to the epidermal sheets, in response to IR. Interestingly, we identified the presence of GFP-positive (GFP(+)) cells in the epidermis-dermis junction in the total skin sections although the epidermal cell sheets failed to have any GFP cells. To examine a possibility that the cells in the junction could be mechanically dissociated during separating epidermal sheets, we then salvaged such dissociated cells and examined its characteristics. Surprisingly, some GFP(+) cells were found in the salvaged cells, indicating that these cells could be derived from BM. In addition, such BMDCs were also associated with inflammation in the junction. In conclusion, BMDCs can migrate to and reside in the epidermis-dermis junction after IR.
Collapse
Affiliation(s)
- Junko Okano
- Division of Anatomy and Cell Biology, Shiga University of Medical Science, Shiga, Japan.
| | - Hideto Kojima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Miwako Katagi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Yuki Nakae
- Department of Internal Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Tomoya Terashima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Takahiko Nakagawa
- TMK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Kurakane
- Division of Anatomy and Cell Biology, Shiga University of Medical Science, Shiga, Japan
| | - Naoki Okamoto
- Division of Anatomy and Cell Biology, Shiga University of Medical Science, Shiga, Japan
| | - Keita Morohashi
- Division of Anatomy and Cell Biology, Shiga University of Medical Science, Shiga, Japan
| | - Hiroshi Maegawa
- Department of Internal Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Jun Udagawa
- Division of Anatomy and Cell Biology, Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
30
|
The danger model approach to the pathogenesis of the rheumatic diseases. J Immunol Res 2015; 2015:506089. [PMID: 25973436 PMCID: PMC4417989 DOI: 10.1155/2015/506089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 12/12/2014] [Indexed: 12/19/2022] Open
Abstract
The danger model was proposed by Polly Matzinger as complement to the traditional self-non-self- (SNS-) model to explain the immunoreactivity. The danger model proposes a central role of the tissular cells' discomfort as an element to prime the immune response processes in opposition to the traditional SNS-model where foreignness is a prerequisite. However recent insights in the proteomics of diverse tissular cells have revealed that under stressful conditions they have a significant potential to initiate, coordinate, and perpetuate autoimmune processes, in many cases, ruling over the adaptive immune response cells; this ruling potential can also be confirmed by observations in several genetically manipulated animal models. Here, we review the pathogenesis of rheumatic diseases such as systemic lupus erythematous, rheumatoid arthritis, spondyloarthritis including ankylosing spondylitis, psoriasis, and Crohn's disease and provide realistic approaches based on the logic of the danger model. We assume that tissular dysfunction is a prerequisite for chronic autoimmunity and propose two genetically conferred hypothetical roles for the tissular cells causing the disease: (A) the Impaired cell and (B) the paranoid cell. Both roles are not mutually exclusive. Some examples in human disease and in animal models are provided based on current evidence.
Collapse
|
31
|
Dirks RP, Burgerhout E, Brittijn SA, de Wijze DL, Ozupek H, Tuinhof-Koelma N, Minegishi Y, Jong-Raadsen SA, Spaink HP, van den Thillart GEEJM. Identification of molecular markers in pectoral fin to predict artificial maturation of female European eels (Anguilla anguilla). Gen Comp Endocrinol 2014; 204:267-76. [PMID: 24992558 DOI: 10.1016/j.ygcen.2014.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 06/17/2014] [Accepted: 06/21/2014] [Indexed: 11/29/2022]
Abstract
The European eel is a critically endangered species that cannot be reproduced in captivity yet. Artificial maturation of female European eels can be achieved via a laborious and expensive procedure, including weekly injections with pituitary extracts for up to 6 months. The success rate is highly variable and a minimally invasive method for early selection of responsive eels would prevent the unnecessary and lengthy treatment of non-responding individuals. Since sexual maturation of European eels is accompanied by morphological changes of the pectoral fin, we examined whether fin could be used to monitor the response to the hormone treatment. Farmed eels were subjected to weekly injections with pituitary extracts and representative groups were sampled at 0 and 14-18 weeks of hormone treatment. Responders and non-responders were identified based on the gonado-somatic index. Transcriptomes of pectoral fin samples obtained at the start and end of the trial were mapped using Illumina RNAseq. Responders showed 384 and non-responders only 54 differentially expressed genes. Highly stringent selection based on minimum expression levels and fold-changes and a manual re-annotation round yielded 23 up-regulated and 21 down-regulated maturation marker genes. The up-regulated markers belong to five categories: proteases, skin/mucus structural proteins, steroid hormone signaling, tyrosine/dopamine metabolism and lipid metabolism. The down-regulated markers are either blood markers or lectin-related genes. In conclusion, pectoral fin transcriptomes are a rich source of indicator markers for monitoring hormone induced sexual maturation of female European eels. In addition, these markers provide important new insight into several fundamental processes in eel biology.
Collapse
Affiliation(s)
- Ron P Dirks
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA Leiden, The Netherlands; ZF-screens B.V., J.H. Oortweg 19, 2333 CH Leiden, The Netherlands.
| | - Erik Burgerhout
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA Leiden, The Netherlands.
| | | | - Danielle L de Wijze
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA Leiden, The Netherlands.
| | - Hulya Ozupek
- ZF-screens B.V., J.H. Oortweg 19, 2333 CH Leiden, The Netherlands.
| | | | - Yuki Minegishi
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA Leiden, The Netherlands.
| | | | - Herman P Spaink
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA Leiden, The Netherlands.
| | - Guido E E J M van den Thillart
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA Leiden, The Netherlands; ZF-screens B.V., J.H. Oortweg 19, 2333 CH Leiden, The Netherlands.
| |
Collapse
|
32
|
ZHANG ZHONGKUI, YANG YONG, BAI SHURONG, ZHANG GUIZHEN, LIU TAIHUA, ZHOU ZHOU, WANG CHUNMEI, TANG LIJUN, WANG JUN, HE SIXIAN. Screening for key genes associated with atopic dermatitis with DNA microarrays. Mol Med Rep 2014; 9:1049-55. [DOI: 10.3892/mmr.2014.1908] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 12/16/2013] [Indexed: 11/05/2022] Open
|