1
|
Mo N, Zhou P, Liu F, Su H, Han L, Lu C. Integrating network pharmacology, molecular docking, and experimental validation to reveal the mechanism of Radix Rehmanniae in psoriasis. Medicine (Baltimore) 2024; 103:e40211. [PMID: 39470475 PMCID: PMC11520997 DOI: 10.1097/md.0000000000040211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Radix Rehmanniae (RR) plays an important role in treating psoriasis. However, the active compounds of RR and potential mechanisms are unclear. The current study was designed to investigate the potential active ingredients, targets, and mechanisms of RR in treating psoriasis through network pharmacology, molecular docking, and vitro experiments. METHODS Initially, the TCMSP database and literature retrieval were used to access the active ingredients of RR. The psoriasis target proteins were obtained from Therapeutic Target Database, OMIM, GeneCards, and DrugBank databases. The target proteins were then converted into target genes using Uniprot. Secondly, overlapping genes were obtained through Venn online tool. Then, protein-protein interactions network diagram is finished by STRING database. Next, Cytoscape software was used to acquire the top 10 hub proteins; gene ontology and Kyoto encyclopedia of genes and genomes enrichment analysis were then used to predict possible mechanisms. Afterwards, molecular docking validation of the active ingredients with the main targets was performed by AutoDock software. Finally, lipopolysaccharides induced RAW264.7, to assess the effects and molecular mechanisms by MTT, RT-qPCR, and Western blot assays. RESULTS Overall, there are 20 effective compounds and 33 targets involved in biological processes including apoptosis, intracellular signaling, vasodilation, and mitogen-activated protein kinase (MAPK) signaling cascade. The docking results showed strong binding capacity between the active ingredients and targets. We verified aucubin as the key active ingredient, tumor necrosis factor α, and IL6 as the core targets, and focused on the p38MAPK protein pathway. Cellular experiments showed that aucubin down-regulated the phosphorylated p38MAP protein and reduced the expression of tumor necrosis factor α mRNA, IL6 mRNA, and IL1βmRNA. CONCLUSION In summary, RR is featured with multicomponent, multi-target, and multi-pathway in treating psoriasis; the preliminary mechanism may be associated with the down-regulation of p38MAPK phosphorylation and curbing the expression of inflammatory factor by aucubin. This paper provides the scientific basis for Traditional Chinese medicine treating psoriasis.
Collapse
Affiliation(s)
- Nian Mo
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Panyu Zhou
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fanlu Liu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haojie Su
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling Han
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of TCM Moisture Syndrome at the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of TCM and Immune Disease Research, Guangzhou, China
- Guangdong Province Hospital of Chinese Medicine, Guangzhou, China
| | - Chuanjian Lu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of TCM Moisture Syndrome at the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of TCM and Immune Disease Research, Guangzhou, China
- Guangdong Province Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
Chen W, Ge L, Zhang C. The molecular mechanism of berberine affecting psoriasis skin inflammation by regulating keratinocyte pyroptosis via the p38 MAPK/NF-κB pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03461-5. [PMID: 39365309 DOI: 10.1007/s00210-024-03461-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/13/2024] [Indexed: 10/05/2024]
Abstract
Berberine (BBR), a Rhizoma Coptis-sourced isoquinoline alkaloid, is an effective drug for psoriasis treatment with its therapeutic mechanism remaining unclear. We delved into the mechanism of BBR affecting psoriatic skin inflammation by regulating keratinocyte pyroptosis. A psoriasis-like skin inflammation mouse model was induced by imiquimod (IMQ) and treated with BBR and a p38 activator anisomycin. Human epidermal keratinocytes (HEKs) were stimulated with five chemokines (M5) [interleukin (IL)-17A, IL-22A, oncostatin M, tumor necrosis factor-α, IL-1α] to simulate psoriasis immune microenvironment, then treated with BBR and anisomycin. Psoriasis skin lesions, skin tissue damage, cell viability and death, and gasdermin D-N (GSDMD-N) and NOD-like receptor protein 3 (NLRP3) positive cell numbers were assessed. The p38 mitogen-activated protein kinase (MAPK)/nuclear factor-kappa B (NF-κB) pathway and levels of the NLRP3/GSDMD pathway-related proteins and inflammatory factors were determined. BBR alleviated M5-induced HEK pyroptosis by inactivating NLRP3 inflammasomes. BBR inhibited the p38 MAPK/NF-κB pathway, and its effects on HEKs were partly averted by activating the p38 MAPK/NF-κB pathway. BBR repressed NLRP3 inflammasome activation and pyroptosis by inhibiting the p38 MAPK/NF-κB pathway. Collectively, BBR suppressed keratinocyte NLRP3/GSDMD pathway pyroptosis by suppressing the p38 MAPK/NF-κB pathway, thereby affecting psoriasis skin inflammation.
Collapse
Affiliation(s)
- Wenfang Chen
- Department of Dermatovenereology, The Second Affiliated Hospital of Shandong First Medical University, No.366 Taishan Street, Taian, 271000, China
| | - Lingzhi Ge
- Department of Dermatovenereology, The Second Affiliated Hospital of Shandong First Medical University, No.366 Taishan Street, Taian, 271000, China
| | - Chao Zhang
- Department of Dermatovenereology, The Second Affiliated Hospital of Shandong First Medical University, No.366 Taishan Street, Taian, 271000, China.
| |
Collapse
|
3
|
Gujarathi PP, Korat RH, Gujarathi PS. Preclinical techniques for drug discovery in psoriasis. Int Immunopharmacol 2024; 137:112378. [PMID: 38852518 DOI: 10.1016/j.intimp.2024.112378] [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: 03/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
Psoriasis is a chronic, inflammatory, papulosquamous, noncontagious disease characterized by scaly, demarcated erythematous plaque, affecting skin, nails, and scalp. The IL-23/Th17 axis is the main operator in the development of psoriasis. Psoriasis is affecting worldwide, and new treatment options are urgently needed. Various local and systemic treatments are available for psoriasis but they only provide symptomatic relief because of numerous unknown mechanisms. Clinical trials demand overwhelming resources; therefore, drug development predominantly depends on the in-vivo, in-vitro, and ex-vivo techniques. Immediate attention is required to develop experimental techniques that completely imitate human psoriasis to assist drug development. This review portrays the various in-vivo, in-vitro, and ex-vivo techniques used in psoriasis research. It describes these techniques' characteristics, pathological presentations, and mechanisms. The experimental techniques of psoriasis provide significant information on disease progression mechanisms and possible therapeutic targets. However, until now, it has been challenging to invent a timely, affordable model that precisely imitates a human disease. Only the xenotransplantation model is reckoned as the closer, that mimics the complete genetic, and immunopathogenic event. Imiquimod-induced psoriasis and HaCat cell lines are popular among researchers because of their convenience, ease of use, and cost-effectiveness. There need to further improve the experimental techniques to best serve the disease imitation and meet the research goal.
Collapse
Affiliation(s)
- Pranjal P Gujarathi
- Department of Pharmacology, Vidhyadeep Institute of Pharmacy, Vidhyadeep University, Anita, Surat, Gujarat, India; Bhagwan Mahavir Centre for Advance Research, Bhagwan Mahavir College of Pharmacy, Bhagwan Mahavir University, Vesu, Surat, Gujarat, India.
| | - Rashmi H Korat
- Department of Pharmacognosy, Bhagwan Mahavir College of Pharmacy, Bhagwan Mahavir Univeristy, Vesu, Surat, Gujarat, India
| | - Piyush S Gujarathi
- Department of Community Medicine, Vidhyadeep Homeopathic Medical College and Research Centre, Vidhyadeep University, Anita, Surat, Gujarat, India
| |
Collapse
|
4
|
Zheng T, Deng J, Wen J, Xiao S, Huang H, Shang J, Zhang L, Chen H, Li J, Wang Y, Ouyang S, Yang M, Otsu K, Liu X, Huang G. p38α deficiency ameliorates psoriasis development by downregulating STAT3-mediated keratinocyte proliferation and cytokine production. Commun Biol 2024; 7:999. [PMID: 39147860 PMCID: PMC11327308 DOI: 10.1038/s42003-024-06700-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
Psoriasis is characterized by keratinocyte (KC) hyperproliferation and inflammatory cell infiltration, but the mechanisms remain unclear. In an imiquimod-induced mouse psoriasiform model, p38 activity is significantly elevated in KCs and p38α specific deletion in KCs ameliorates skin inflammation. p38α signaling promotes KC proliferation and psoriasis-related proinflammatory gene expression during psoriasis development. Mechanistically, p38α enhances KC proliferation and production of inflammatory cytokines and chemokines by activating STAT3. While p38α signaling in KCs does not affect the expression of IL-23 and IL-17, it substantially amplifies the IL-23/IL-17 pathogenic axis in psoriasis. The therapeutic effect of IL-17 neutralization is associated with decreased p38 and STAT3 activities in KCs and targeting the p38α-STAT3 axis in KCs ameliorates the severity of psoriasis. As IL-17 also highly activates p38 and STAT3 in KCs, our findings reveal a sustained signaling circuit important for psoriasis development, highlighting p38α-STAT3 axis as an important target for psoriasis treatment.
Collapse
Affiliation(s)
- Tingting Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
| | - Jiaqi Deng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jiahong Wen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Shuxiu Xiao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyong Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jiawen Shang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Luwen Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Huan Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jingyu Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yanyan Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Meng Yang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Kinya Otsu
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Cardiovascular Division, King's College London, London, UK
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Dongguan, China
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Richardson KC, Aubert A, Turner CT, Nabai L, Hiroyasu S, Pawluk MA, Cederberg RA, Zhao H, Jung K, Burleigh A, Crawford RI, Granville DJ. Granzyme K mediates IL-23-dependent inflammation and keratinocyte proliferation in psoriasis. Front Immunol 2024; 15:1398120. [PMID: 38903528 PMCID: PMC11188347 DOI: 10.3389/fimmu.2024.1398120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/17/2024] [Indexed: 06/22/2024] Open
Abstract
Psoriasis is an inflammatory disease with systemic manifestations that most commonly presents as itchy, erythematous, scaly plaques on extensor surfaces. Activation of the IL-23/IL-17 pro-inflammatory signaling pathway is a hallmark of psoriasis and its inhibition is key to clinical management. Granzyme K (GzmK) is an immune cell-secreted serine protease elevated in inflammatory and proliferative skin conditions. In the present study, human psoriasis lesions exhibited elevated GzmK levels compared to non-lesional psoriasis and healthy control skin. In an established murine model of imiquimod (IMQ)-induced psoriasis, genetic loss of GzmK significantly reduced disease severity, as determined by delayed plaque formation, decreased erythema and desquamation, reduced epidermal thickness, and inflammatory infiltrate. Molecular characterization in vitro revealed that GzmK contributed to macrophage secretion of IL-23 as well as PAR-1-dependent keratinocyte proliferation. These findings demonstrate that GzmK enhances IL-23-driven inflammation as well as keratinocyte proliferation to exacerbate psoriasis severity.
Collapse
Affiliation(s)
- Katlyn C. Richardson
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Alexandre Aubert
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Christopher T. Turner
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Layla Nabai
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Sho Hiroyasu
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Megan A. Pawluk
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Rachel A. Cederberg
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Integrative Oncology Department, British Columbia (BC) Cancer Research Centre, Vancouver, BC, Canada
| | - Hongyan Zhao
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Karen Jung
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Angela Burleigh
- Department of Dermatology and Skin Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Richard I. Crawford
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Dermatology and Skin Sciences, University of British Columbia, Vancouver, BC, Canada
| | - David J. Granville
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Lee JH, Lee JE, Son SE, Son SH, Kim NJ, Im DS. NJK14047 inhibition of p38 MAPK ameliorates inflammatory immune diseases by suppressing T cell differentiation. Int Immunopharmacol 2024; 130:111800. [PMID: 38447416 DOI: 10.1016/j.intimp.2024.111800] [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: 01/03/2024] [Revised: 02/19/2024] [Accepted: 03/02/2024] [Indexed: 03/08/2024]
Abstract
p38 MAPK has been implicated in the pathogenesis of rheumatoid arthritis and psoriasis. To assess the therapeutic efficacy of the p38 MAPK inhibitor NJK14047 in the treatment of rheumatoid arthritis and psoriasis, we developed mouse models of collagen-induced rheumatoid arthritis (CIA) and imiquimod-induced psoriasis (IIP). NJK14047 was found to suppress arthritis development and psoriasis symptoms and also suppressed histopathological changes induced by CIA and IIP. Furthermore, we established that CIA and IIP evoked increases in the mRNA expression levels of Th1/Th17 inflammatory cytokines in the joints and skin, which was again suppressed by NJK14047. NJK14047 reversed the enlargement of spleens induced by CIA and IIP as well as increases in the levels of inflammatory cytokine in spleens following induction by CIA and IIP. In human SW982 synovial cells, NJK14047 was found to suppress lipopolysaccharide-induced increases in the mRNA expression of proinflammatory cytokines. NJK14047 inhibition of p38 MAPK suppressed the differentiation of naïve T cells to Th17 and Th1 cells. Our findings in this study provide convincing evidence indicating the therapeutic efficacy of the p38 MAPK inhibitor NJK14047 against CIA and IIP, which we speculate could be associated with the suppression on T-cell differentiation.
Collapse
Affiliation(s)
- Ju-Hyun Lee
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02446, Republic of Korea
| | - Jung-Eun Lee
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02446, Republic of Korea
| | - So-Eun Son
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02446, Republic of Korea
| | - Seung-Hwan Son
- Department of Basic Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02446, Republic of Korea
| | - Nam-Jung Kim
- Department of Basic Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02446, Republic of Korea
| | - Dong-Soon Im
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02446, Republic of Korea; Department of Basic Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02446, Republic of Korea.
| |
Collapse
|
7
|
刘 学, 贾 皇, 王 丽, 王 子, 许 孟, 李 云, 王 茸. [Screening for Characteristic Genes of Different Traditional Chinese Medicine Syndromes of Psoriasis Vulgaris: A Study Based on Bioinformatics and Machine Learning]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:337-345. [PMID: 38645867 PMCID: PMC11026890 DOI: 10.12182/20240360402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Indexed: 04/23/2024]
Abstract
Objective To screen for the key characteristic genes of the psoriasis vulgaris (PV) patients with different Traditional Chinese Medicine (TCM) syndromes, including blood-heat syndrome (BHS), blood stasis syndrome (BSS), and blood-dryness syndrome (BDS), through bioinformatics and machine learning and to provide a scientific basis for the clinical diagnosis and treatment of PV of different TCM syndrome types. Methods The GSE192867 dataset was downloaded from Gene Expression Omnibus (GEO). The limma package was used to screen for the differentially expressed genes (DEGs) of PV, BHS, BSS, and BDS in PV patients and healthy populations. In addition, KEGG (Kyoto Encyclopedia of Genes and Genes) pathway enrichment analysis was performed. The DEGs associated with PV, BHS, BSS, and BDS were identified in the screening and were intersected separately to obtain differentially characterized genes. Out of two algorithms, the support vector machine (SVM) and random forest (RF), the one that produced the optimal performance was used to analyze the characteristic genes and the top 5 genes were identified as the key characteristic genes. The receiver operating characteristic (ROC) curves of the key characteristic genes were plotted by using the pROC package, the area under curve (AUC) was calculated, and the diagnostic performance was evaluated, accordingly. Results The numbers of DEGs associated with PV, BHS, BSS, and BDS were 7699, 7291, 7654, and 6578, respectively. KEGG enrichment analysis was focused on Janus kinase (JAK)/signal transducer and activator of transcription (STAT), cyclic adenosine monophosphate (cAMP), mitogen-activated protein kinase (MAPK), apoptosis, and other pathways. A total of 13 key characteristic genes were identified in the screening by machine learning. Among the 13 key characteristic genes, malectin (MLEC), TUB like protein 3 (TULP3), SET domain containing 9 (SETD9), nuclear envelope integral membrane protein 2 (NEMP2), and BTG anti-proliferation factor 3 (BTG3) were the key characteristic genes of BHS; phosphatase 15 (DUSP15), C1q and tumor necrosis factor related protein 7 (C1QTNF7), solute carrier family 12 member 5 (SLC12A5), tripartite motif containing 63 (TRIM63), and ubiquitin associated protein 1 like (UBAP1L) were the key characteristic genes of BSS; recombinant mouse protein (RRNAD1), GTPase-activating protein ASAP3 Protein (ASAP3), and human myomesin 2 (MYOM2) were the key characteristic genes of BDS. Moreover, all of them showed high diagnostic efficacy. Conclusion There are significant differences in the characteristic genes of different PV syndromes and they may be potential biomarkers for diagnosing TCM syndromes of PV.
Collapse
Affiliation(s)
- 学伟 刘
- 河南中医药大学 (郑州 450046)Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - 皇超 贾
- 河南中医药大学 (郑州 450046)Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - 丽云 王
- 河南中医药大学 (郑州 450046)Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - 子雯 王
- 河南中医药大学 (郑州 450046)Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - 孟月 许
- 河南中医药大学 (郑州 450046)Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - 云飞 李
- 河南中医药大学 (郑州 450046)Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - 茸慧 王
- 河南中医药大学 (郑州 450046)Henan University of Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|
8
|
Zhao Q, Wu Y, Wu X, Liu M, Nan L. Single-cell transcriptome analysis reveals keratinocyte subpopulations contributing to psoriasis in corneum and granular layer. Skin Res Technol 2024; 30:e13572. [PMID: 38279596 PMCID: PMC10818132 DOI: 10.1111/srt.13572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Psoriasis is a chronic, inflammatory skin disease that is common and relapses easily. While the importance of keratinocyte proliferation in psoriasis development is well-documented, the specific functional subpopulations of epidermal keratinocytes associated with this disease remain enigmatic. MATERIALS AND METHODS Therefore, in our analysis of single-cell transcriptome data from both normal and psoriatic skin tissues, we observed significant increases in certain keratinocytes in the stratum corneum (KC) and stratum granulosum (KG) within psoriatic skin. Furthermore, we identified upregulated expression of specific secreted factors known to promote inflammatory responses. Additionally, we conducted a KEGG pathway enrichment analysis on these identified subsets. RESULTS In the stratum corneum, the expression of FTL was upregulated in HIST1H1C+ KC. S100P+ KC displayed a significant increase in the expression of both S100P and S100A10, whereas PRR9+ KC showed upregulated expression of DEFB4B, S100A8, and S100A12. SLURP1+ KC was characterized by elevated expression levels of IL-36G, SLURP1, and S100A12. Meanwhile, in the stratum granulosum, KRT1+ KG highly expressed SLURP1, S100A7, S100A8, and S100A9, while DEFB4B expression was upregulated in PI3+ KG. Our findings indicated that subsets within the stratum corneum primarily participate in pathways related to MAPK, NOD-like receptors, HIF-1, cell senescence, and other crucial processes. In contrast, subsets in the stratum granulosum were predominantly associated with pathways involving MAPK, NOD-like receptors, HIF-1, Hippo, mTOR, and IL-17. CONCLUSION These findings not only uncover the keratinocyte subsets linked to psoriasis but also unveil the molecular mechanisms and related signaling pathways that drive psoriasis development. This knowledge opens new horizons for the development of innovative clinical treatment strategies for psoriasis.
Collapse
Affiliation(s)
- Qianya Zhao
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
- Department of DermatologyGansu Provincial HospitalLanzhouGansuChina
| | - Yan Wu
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
| | - Xianwei Wu
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
- Department of DermatologyGansu Provincial HospitalLanzhouGansuChina
| | - Meng Liu
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
- Department of DermatologyGansu Provincial HospitalLanzhouGansuChina
| | - Lisheng Nan
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
- Department of DermatologyGansu Provincial HospitalLanzhouGansuChina
| |
Collapse
|
9
|
Dainichi T, Matsumoto R, Sakurai K, Kabashima K. Necessary and sufficient factors of keratinocytes in psoriatic dermatitis. Front Immunol 2024; 15:1326219. [PMID: 38312837 PMCID: PMC10834637 DOI: 10.3389/fimmu.2024.1326219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/05/2024] [Indexed: 02/06/2024] Open
Affiliation(s)
- Teruki Dainichi
- Department of Dermatology, Kagawa University Faculty of Medicine, Miki-cho, Japan
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Reiko Matsumoto
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Sakurai
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Agency for Science, Technology and Research (A*STAR) Skin Research Laboratories (ASRL), A*STAR, Singapore, Singapore
| |
Collapse
|
10
|
Qian Y, Zhu L, Chen J, Zhou Y, Huang Z, Liang L, Ding B. Di-(2-ethylhexyl) phthalate aggravates psoriasis-like skin lesions: In vitro and in vivo evaluation. Toxicol Appl Pharmacol 2023; 479:116707. [PMID: 37783235 DOI: 10.1016/j.taap.2023.116707] [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: 06/11/2023] [Revised: 09/17/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), which is a widely used phthalate (PAE), has recently received public attention owing to it causing health problems. The aim of this study was to elucidate the aggravating effects of DEHP on psoriasis and skin toxicity. Human keratinocyte (HaCaT) cells were treated with gradient concentrations of DEHP, and mice with imiquimod (IMQ)-induced psoriasiform dermatitis were hypodermically injected with 40 μg/kg/day of DEHP for seven consecutive days. The skin condition was assessed based on the psoriasis area and severity index score, which indicated the deterioration of IMQ-induced psoriasis-like skin lesions after DEHP exposure. To further analyze the effect of DEHP on psoriasis, the proliferation, inflammation, and tight junction (TJ) damage were examined, which correlated with the development and severity of psoriasis. The results showed that DEHP promoted proliferation both in vivo and in vitro, which manifested as epidermal thickening; an increase in cell viability; upregulation of Ki67, CDK2, cyclinD1, and proliferating cell nuclear antigen; and downregulation of p21. An excessive inflammatory response is an important factor that exacerbates psoriasis, and our results showed that DEHP can trigger the release of inflammatory cytokines as well as the infiltration of T cells. TJ disorders were found in mice and cells after DEHP treatment. Additionally, p38 mitogen-activated protein kinase (MAPK) was strongly activated during this process, which may have contributed to skin toxicity caused by DEHP. In conclusion, DEHP treatment promotes proliferation, inflammation, TJ disruption, and p38 MAPK activation in HaCaT cells and psoriasis-like skin lesions.
Collapse
Affiliation(s)
- Yuxin Qian
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Lijian Zhu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Jingya Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Yilin Zhou
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Zhiguang Huang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Linjie Liang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Bin Ding
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310000, China.
| |
Collapse
|
11
|
Dainichi T, Iwata M. Inflammatory loops in the epithelial-immune microenvironment of the skin and skin appendages in chronic inflammatory diseases. Front Immunol 2023; 14:1274270. [PMID: 37841246 PMCID: PMC10568311 DOI: 10.3389/fimmu.2023.1274270] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/05/2023] [Indexed: 10/17/2023] Open
Abstract
The epithelial-immune microenvironment (EIME) of epithelial tissues has five common elements: (1) microbial flora, (2) barrier, (3) epithelial cells, (4) immune cells, and (5) peripheral nerve endings. EIME provides both constant defense and situation-specific protective responses through three-layered mechanisms comprising barriers, innate immunity, and acquired immunity. The skin is one of the largest organs in the host defense system. The interactions between the five EIME elements of the skin protect against external dangers from the environment. This dysregulation can result in the generation of inflammatory loops in chronic inflammatory skin diseases. Here, we propose an understanding of EIME in chronic skin diseases, such as atopic dermatitis, psoriasis, systemic lupus erythematosus, alopecia areata, and acne vulgaris. We discuss the current treatment strategies targeting their inflammatory loops and propose possible therapeutic targets in the future.
Collapse
Affiliation(s)
- Teruki Dainichi
- Department of Dermatology, Kagawa University Faculty of Medicine, Kagawa, Japan
| | | |
Collapse
|
12
|
Lang M, Krump C, Meshcheryakova A, Tam-Amersdorfer C, Schwarzenberger E, Passegger C, Connolly S, Mechtcheriakova D, Strobl H. Microenvironmental and cell intrinsic factors governing human cDC2 differentiation and monocyte reprogramming. Front Immunol 2023; 14:1216352. [PMID: 37539048 PMCID: PMC10395083 DOI: 10.3389/fimmu.2023.1216352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/14/2023] [Indexed: 08/05/2023] Open
Abstract
cDC2s occur abundantly in peripheral tissues and arise from circulating blood cDC2s. However, the factors governing cDC2 differentiation in tissues, especially under inflammatory conditions, remained poorly defined. We here found that psoriatic cDC2s express the efferocytosis receptor Axl and exhibit a bone morphogenetic protein (BMP) and p38MAPK signaling signature. BMP7, strongly expressed within the lesional psoriatic epidermis, cooperates with canonical TGF-β1 signaling for inducing Axl+cDC2s from blood cDC2s in vitro. Moreover, downstream induced p38MAPK promotes Axl+cDC2s at the expense of Axl+CD207+ Langerhans cell differentiation from blood cDC2s. BMP7 supplementation allowed to model cDC2 generation and their further differentiation into LCs from CD34+ hematopoietic progenitor cells in defined serum-free medium. Additionally, p38MAPK promoted the generation of another cDC2 subset lacking Axl but expressing the non-classical NFkB transcription factor RelB in vitro. Such RelB+cDC2s occurred predominantly at dermal sites in the inflamed skin. Finally, we found that cDC2s can be induced to acquire high levels of the monocyte lineage identity factor kruppel-like-factor-4 (KLF4) along with monocyte-derived DC and macrophage phenotypic characteristics in vitro. In conclusion, inflammatory and psoriatic epidermal signals instruct blood cDC2s to acquire phenotypic characteristics of several tissue-resident cell subsets.
Collapse
Affiliation(s)
- Magdalena Lang
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Corinna Krump
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Anastasia Meshcheryakova
- Insitute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Carmen Tam-Amersdorfer
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Elke Schwarzenberger
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Christina Passegger
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Sally Connolly
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Diana Mechtcheriakova
- Insitute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
13
|
Zhu C, Chen Y, Tai Z, Pan H, Shen M, Chen Z, Zhu Q. Effect and mechanism of longkui yinxiao soup in treating psoriasis in mice. Front Pharmacol 2023; 14:1136604. [PMID: 36992832 PMCID: PMC10040526 DOI: 10.3389/fphar.2023.1136604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/22/2023] [Indexed: 03/14/2023] Open
Abstract
Objective: Longkui Yinxiao Soup is a traditional Chinese medicine formula used to treat psoriasis for decades. Although Longkui Yinxiao Soup showed promising efficacy in clinical practice, the regulatory mechanisms of Longkui Yinxiao Soup remain elusive. This study aimed to explore the underlying mechanisms of Longkui Yinxiao Soup in a psoriasis-like mouse model.Methods: Longkui Yinxiao Soup was quality controlled by determining the contents of imperatorin and rhoifolin using high-performance liquid chromatography. The imiquimod-induced psoriasis-like mouse model was used to study the therapeutic effect and mechanism of Longkui Yinxiao Soup. The histopathological skin changes were observed by hematoxylin and eosin staining; the infiltration of proliferating proteins, proliferating cell nuclear antigen and Ki67, in skin tissues were observed by immunohistochemical analysis; and the inflammatory factors such as interleukin (IL)-6, tumor necrosis factor (TNF)-α, IL-23, and IL-17 in serum were detected using enzyme-linked immunosorbent assay. RNA sequencing and bioinformatic analysis were used to predict the mechanism of LYS against psoriasis. mRNA expressions of p38, extracellular regulated protein kinases (ERK), mitogen-activated protein kinase 3 (MEK3), mitogen-activated protein kinase 6 (MEK6), RAP1 GTPase activating protein (Rap1gap), and Rap1 were determined using real-time quantitative polymerase chain reaction. The expression levels of proteins related to Rap1–mitogen-activated protein kinase signaling pathways were measured by Western blotting.Results: A quality-control method for Longkui Yinxiao Soup was successfully established using imperatorin and rhoifolin as content determination indexes. Longkui Yinxiao Soup significantly ameliorated the psoriatic symptoms in mice. The serum levels of inflammatory cytokines such as IL-6, TNF-α, IL-23, and IL-17 were decreased, and the expression levels of antigen identified by monoclonal antibody Ki67 (Ki67) and PCNA in skin tissues were downregulated. Moreover, the inhibition of Rap1–MAPK signaling pathways by Longkui Yinxiao Soup was detected.Conclusion: This study confirmed the antipsoriatic activity of Longkui Yinxiao Soup in psoriasis-like mice. This might be due to the inhibition of inflammatory factor secretion, keratinocyte proliferation, and the Rap1–MAPK signal pathway.
Collapse
Affiliation(s)
- Congcong Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Ya Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Huijun Pan
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Min Shen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
- *Correspondence: Quangang Zhu, ; Zhongjian Chen,
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
- *Correspondence: Quangang Zhu, ; Zhongjian Chen,
| |
Collapse
|
14
|
Saika A, Tiwari P, Nagatake T, Node E, Hosomi K, Honda T, Kabashima K, Kunisawa J. Mead acid inhibits retinol-induced irritant contact dermatitis via peroxisome proliferator-activated receptor alpha. Front Mol Biosci 2023; 10:1097955. [PMID: 36825199 PMCID: PMC9941550 DOI: 10.3389/fmolb.2023.1097955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Retinol is widely used in topical skincare products to ameliorate skin aging and treat acne and wrinkles; however, retinol and its derivatives occasionally have adverse side effects, including the induction of irritant contact dermatitis. Previously, we reported that mead acid (5,8,11-eicosatrienoic acid), an oleic acid metabolite, ameliorated skin inflammation in dinitrofluorobenzene-induced allergic contact hypersensitivity by inhibiting neutrophil infiltration and leukotriene B4 production by neutrophils. Here, we showed that mead acid also suppresses retinol-induced irritant contact dermatitis. In a murine model, we revealed that mead acid inhibited keratinocyte abnormalities such as keratinocyte hyperproliferation. Consistently, mead acid inhibited p38 MAPK (mitogen-activated protein kinase) phosphorylation, which is an essential signaling pathway in the keratinocyte hyperplasia induced by retinol. These inhibitory effects of mead acid were associated with the prevention of both keratinocyte hyperproliferation and the gene expression of neutrophil chemoattractants, including Cxcl1 and Cxcl2, and they were mediated by a PPAR (peroxisome proliferator-activated receptor)-α pathway. Our findings identified the anti-inflammatory effects of mead acid, the use of which can be expected to minimize the risk of adverse side effects associated with topical retinoid application.
Collapse
Affiliation(s)
- Azusa Saika
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Prabha Tiwari
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan,Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan,Laboratory of Functional Anatomy, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Eri Node
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Tetsuya Honda
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan,International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Minato, Tokyo, Japan,Graduate School of Medicine, Graduate School of Dentistry, Graduate School of Pharmaceutical Sciences, Graduate School of Science, Osaka University, Suita, Osaka, Japan,Department of Microbiology and Immunology, Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan,Research Organization for Nano and Life Innovation, Waseda University, Shinjuku, Tokyo, Japan,Graduate School of Biomedical and Health Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan,*Correspondence: Jun Kunisawa,
| |
Collapse
|
15
|
Luo L, Pasquali L, Srivastava A, Freisenhausen JC, Pivarcsi A, Sonkoly E. The Long Noncoding RNA LINC00958 Is Induced in Psoriasis Epidermis and Modulates Epidermal Proliferation. J Invest Dermatol 2023; 143:999-1010. [PMID: 36641130 DOI: 10.1016/j.jid.2022.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023]
Abstract
Psoriasis is a common, immune-mediated skin disease characterized by epidermal hyperproliferation and chronic skin inflammation. Long noncoding RNAs are >200 nucleotide-long transcripts that possess important regulatory functions. To date, little is known about the contribution of long noncoding RNAs to psoriasis. In this study, we identify LINC00958 as a long noncoding RNA overexpressed in keratinocytes (KCs) from psoriasis skin lesions, in a transcriptomic screen performed on KCs sorted from psoriasis and healthy skin. Increased levels of LINC00958 in psoriasis KCs were confirmed by RT-qPCR and single-molecule in situ hybridization. Confocal microscopy and analysis of subcellular fractions showed that LINC00958 is mainly localized in the cytoplasm of KCs. IL-17A, a key psoriasis cytokine, induced LINC00958 in KCs through C/EBP-β and the p38 pathway. The inhibition of LINC00958 led to decreased proliferation as measured by Ki-67 expression, IncuCyte imaging, and 5-ethynyl-2-deoxyuridine assays. Transcriptomic analysis of LINC00958-depleted KCs revealed enrichment of proliferation- and cell cycle‒related genes among differentially expressed transcripts. Moreover, LINC00958 depletion led to decreased basal and IL-17A‒induced phosphorylation of p38. Furthermore, IL-17A‒induced KC proliferation was counteracted by the inhibition of LINC00958. In summary, our data support a role for the IL-17A‒induced long noncoding RNA, LINC00958, in the pathological circuits of psoriasis by reinforcing IL-17A‒induced epidermal hyperproliferation.
Collapse
Affiliation(s)
- Longlong Luo
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Lorenzo Pasquali
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Ankit Srivastava
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Science for Life Laboratory, Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Solna, Sweden; Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Jan C Freisenhausen
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Andor Pivarcsi
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Enikö Sonkoly
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
16
|
Sun L, Yu G, Jiang H, Shi K, Huang D, Bao H, Huang Y, Che L, Chen M. Experimental study on the anti-inflammatory effect of Mongolian medicine Sendeng Decoction on mice with psoriasis. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
17
|
Bai D, Cheng X, Li Q, Zhang B, Zhang Y, Lu F, Sun T, Hao J. Eupatilin inhibits keratinocyte proliferation and ameliorates imiquimod-induced psoriasis-like skin lesions in mice via the p38 MAPK/NF-κB signaling pathway. Immunopharmacol Immunotoxicol 2022; 45:133-139. [PMID: 36305632 DOI: 10.1080/08923973.2022.2121928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Psoriasis is a chronic inflammatory skin disease that is currently incurable and causes long-term distress to patients. Therefore, there is an urgent need to develop safe and effective psoriatic drugs. Eupatilin is a natural flavone, that has a variety of pharmacological effects. However, the anti-psoriatic effect of eupatilin and its underlying mechanism remain unclear. METHODS HaCaT cells were treated with 20 μg/mL LPS for 24 h to establish the proliferation model of HaCaT cells. Cell viability was measured by MTT assay. Western blotting was used to detect the expression of p-p38 MAPK, p38 MAPK, p-NF-κB p65 and NF-κB p65 in HaCaT cells. Imiquimod (IMQ) was used to induce psoriasis-like mouse model. Psoriasis Area Severity Index (PASI) score was used to evaluate the degree of skin injury, H&E staining was used to observe the pathological damage of skin tissues, and the expression levels of TNF-α, IL-6, IL-23 and IL-17 in the serum were detected by enzyme-linked immunosorbent assay (ELISA). RESULTS Eupatilin could inhibit the hyperproliferation of LPS-stimulated HaCaT cells through p38 MAPK/NF-κB signaling pathway in vitro. In psoriatic mice, eupatilin could significantly reduce skin erythema, scales and thickening scores, ameliorate skin histopathological lesions, and decrease the levels of TNF-α, IL-6, IL-23 and IL-17 in the serum. CONCLUSION Eupatilin had a good anti-proliferative effect in LPS-stimulated HaCaT cells, and significantly alleviated IMQ-induced psoriasis-like lesions in mice. Eupatilin was a promising drug for the treatment of psoriasis.
Collapse
Affiliation(s)
- Donghui Bai
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, PR China
| | | | - Qiong Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Bo Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, PR China
| | - Yan Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, PR China
| | - Fang Lu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, PR China
| | - Tianxiao Sun
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, PR China
| | - Jiejie Hao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, PR China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, PR China
| |
Collapse
|
18
|
Exploration of the Protective Mechanism of Bax Removal against Ischemia Reperfusion Injury of Skin Flap through the p38 Mitogen-Activated Protein Kinase Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1175078. [PMID: 36299606 PMCID: PMC9592197 DOI: 10.1155/2022/1175078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/21/2022]
Abstract
This research is aimed at exploring the influences of the Bax gene in the p38 mitogen-activated protein kinase (MAPK) pathway and its protective mechanism against ischemia-reperfusion injury (IRI) of skin flap. Forty male Sprague-Dawley (SD) rats were equally divided into the experimental group (Bax gene knockout rats) and control group. The dorsal flap model was prepared, and the survival rate of flap was observed after surgery. The rat flap tissue was cut and stained with hematoxylin-eosin (HE) and in situ terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL). The distribution characteristics of p38MAPK and Bax were detected to evaluate the protective mechanism of Bax gene knockout on IRI of skin flap. After surgery, the survival rate of flaps in the experimental group (82.32%, 70.28%) was significantly higher than that in the control group (57.64%, 46.14%) (P < 0.05). The results of HE staining showed that on the 1st day after surgery, compared with those in the control group, the skin flaps of the rats in the experimental group were arranged more neatly. The results of TUNEL staining showed that compared with that of the control group, the tissue structure of the skin flap of the experimental group was normal and only a few apoptotic cells appeared. In addition, compared with that in the control group (7.14, 4.25, 3.48, 2.18/6.46, 7.12, 4.86, and 2.44), the expression of Bax and p38 MAPK in the experimental group (0.96, 0.81, 0.76, 0.55/1.63, 1.33, 1.01, and 0.56) significantly decreased (P < 0.05). In short, after the Bax gene was knocked out, injury of the flap after ischemia-reperfusion was considerably improved, which may play a protective role on the IRI of the flap by affecting the p38MAPK pathway.
Collapse
|
19
|
Hu X, Qi C, Feng F, Wang Y, Di T, Meng Y, Wang Y, Zhao N, Zhang X, Li P, Zhao J. Combining network pharmacology, RNA-seq, and metabolomics strategies to reveal the mechanism of Cimicifugae Rhizoma - Smilax glabra Roxb herb pair for the treatment of psoriasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154384. [PMID: 35963195 DOI: 10.1016/j.phymed.2022.154384] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/14/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Psoriasis is a prevalent chronic inflammatory skin condition marked by immune cell infiltration and keratinocyte abnormal proliferation. Cimicifugae Rhizoma - Smilax glabra Roxb (CS) herb pair, the main component of Shengma Detoxification Decoction, has been proven effective for the treatment of psoriasis. However, the mechanism is yet to be deciphered. PURPOSE To explore the mechanism of CS for the treatment of psoriasis. METHODS The imiquimod-induced psoriasis-like lesion mouse model was used to identify the targets and the molecular mechanisms of CS. Network pharmacology combined with RNA-seq strategy was employed to predict the targets and mechanisms of CS for psoriasis. Metabolomics approaches were used to demonstrate the complexity of CS for the treatment of psoriasis. Finally, a compound-response-enzyme-gene network was constructed based on the multi-omics results to elucidate potential connections. RESULTS The CS herb pair could significantly improve psoriatic lesions and reduce the inflammatory cell infiltration and proliferation of keratinocytes in skin lesions. Network pharmacology predicted that TNF, JNK, IL-6, and IL-1β could be potential targets. RNA-seq data revealed that CS could significantly regulate genes and signaling pathways associated with Th17 responses, such as IL-36, IL-1β, CCl2, CXCL16, keratin 14, keratin 5, and antimicrobial peptides S100A8 and S100A9 well as MAPK, mTOR, and other signaling pathways. Further experimental data validated that CS treatment remarkably reduced the expression of inflammatory cytokines and factors, such as CCL2, CCL7, IL1F6, IL-17, IL-23, IL-1β, TNF-α, and IL-6, and inhibited the phosphorylation of p38 and ERK1/2. This indicated that CS exerts its therapeutic effect by inhibiting the MAPK signaling pathways. In addition, metabolomic analyses demonstrated that CS treatment improved seven metabolic pathways, these included phenylalanine, tyrosine, pyruvate metabolism, carnitine metabolism, etc. Four key metabolites (L-Arginine, L-Phenylalanine, L-Carnitine, O-Acetylcarnitine) and nine differential genes (CMA1, PCBD2, TPSAB1, TPSB2, etc.) were identified that affected amino acid metabolism, carnitine metabolism, and other pathways contributing to the infiltration of Th17 cells in psoriatic lesions. CONCLUSION CS could alleviate IMQ-induced psoriasis-like dermatitis by reducing the expression of cytokines and chemokines mediated by the MAPK pathway, and improved amino acid and carnitine metabolism in vivo. Our study is the first to demonstrate the complex mechanism of CS for the treatment of psoriasis and provides a new paradigm to elucidate the pharmacological effects of Traditional Chinese Medicine (TCM) drugs for psoriasis from multiple perspectives.
Collapse
Affiliation(s)
- XueQing Hu
- Beijing University of Chinese Medicine, Beijing 100105, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Cong Qi
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Fang Feng
- Beijing University of Chinese Medicine, Beijing 100105, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Yan Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - TingTing Di
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - YuJiao Meng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Yazhuo Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Ning Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - XiaWei Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China.
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China.
| |
Collapse
|
20
|
Mandal AK, Sahoo A, Dwivedi K, Singh R, Kumar V. Potential therapeutic application of biophenols - plants secondary metabolites in rheumatoid arthritis. Crit Rev Food Sci Nutr 2022; 63:8900-8918. [PMID: 35593234 DOI: 10.1080/10408398.2022.2062700] [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] [Indexed: 11/03/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease showed that persistent inflammation in the joints, induces the cartilage destruction, bone erosion, and leukocyte infiltration in the synovium. RA mostly affects the joints of hands, feet, wrists, ankles, and knees. Each year, approximately 20-40 new cases are reported per lac population and the disease affects women more than men. The etiology of RA is still unknown, but many pathways have been identified as potential targets in its pathophysiology, including the PI3K/AKT signaling pathway, NF-κB signaling, Adenosine signaling, Wnt, SYK/BTK, and mTOR signaling pathways. Biophenol, plant secondary metabolite, is considered one of the most abundantly phytoconstituents to have potential anti-inflammatory effects associated with multiple pathways. These indicate that biophenols can be used for its protective effect on the development and symptoms of RA. The current review explores and discusses the role of different biophenols in the treatment of RA disease.
Collapse
Affiliation(s)
| | - Ankit Sahoo
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Science, Shalom Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, Uttar Pradesh, India
| | - Khusbu Dwivedi
- Department of Pharmaceutics, Shambhunath Institute of Pharmacy, Prayagraj, Uttar Pradesh, India
| | - Richa Singh
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Science, Shalom Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, Uttar Pradesh, India
| | - Vikas Kumar
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Science, Shalom Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, Uttar Pradesh, India
| |
Collapse
|
21
|
Selenium-Rich Yeast Peptide Fraction Ameliorates Imiquimod-Induced Psoriasis-like Dermatitis in Mice by Inhibiting Inflammation via MAPK and NF-κB Signaling Pathways. Int J Mol Sci 2022; 23:ijms23042112. [PMID: 35216231 PMCID: PMC8875820 DOI: 10.3390/ijms23042112] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 02/05/2023] Open
Abstract
Psoriasis, a chronic and immune-mediated inflammatory disease, adversely affects patients’ lives. We previously prepared selenium-rich yeast peptide fraction (SeP) from selenium-rich yeast protein hydrolysate and found that SeP could effectively alleviate ultraviolet radiation-induced skin damage in mice and inhibited H2O2-induced cytotoxicity in cultured human epidermal keratinocyte (HaCaT) cells. This study aimed to investigate whether SeP had a protective effect on imiquimod (IMQ)-induced psoriasis-like dermatitis in mice and the underlying mechanisms. Results showed that SeP significantly ameliorated the severity of skin lesion in IMQ-induced psoriasis-like mouse model. Moreover, SeP treatment significantly attenuated the expression of key inflammatory cytokines, including interleukin (IL)-23, IL-17A, and IL-17F, in the dorsal skin of mice. Mechanistically, SeP application not only inhibited the activation of JNK and p38 MAPK, but also the translocation of NF-κB into the nucleus in the dorsal skin. Furthermore, SeP treatment inhibited the levels of inflammatory cytokines and the activation of MAPK and NF-κB signaling induced by lipopolysaccharide in HaCaT cells and macrophage cell line RAW264.7. Overall, our findings showed that SeP alleviated psoriasis-like skin inflammation by inhibiting MAPK and NF-κB signaling pathways, which suggested that SeP would have a potential therapeutic effect against psoriasis.
Collapse
|
22
|
Inhibition of CtBP-Regulated Proinflammatory Gene Transcription Attenuates Psoriatic Skin Inflammation. J Invest Dermatol 2022; 142:390-401. [PMID: 34293351 PMCID: PMC8770725 DOI: 10.1016/j.jid.2021.06.029] [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: 02/25/2020] [Revised: 05/25/2021] [Accepted: 06/03/2021] [Indexed: 02/03/2023]
Abstract
Psoriasis is a chronic immune-mediated disease characterized by excessive proliferation of epidermal keratinocytes and increased immune cell infiltration to the skin. Although it is well-known that psoriasis pathogenesis is driven by aberrant production of proinflammatory cytokines, the mechanisms underlying the imbalance between proinflammatory and anti-inflammatory cytokine expression are incompletely understood. In this study, we report that the transcriptional coregulators CtBP1 and 2 can transactivate a common set of proinflammatory genes both in the skin of imiquimod-induced mouse psoriasis model and in human keratinocytes and macrophages stimulated by imiquimod. We find that mice overexpressing CtBP1 in epidermal keratinocytes display severe skin inflammation phenotypes with increased expression of T helper type 1 and T helper type 17 cytokines. We also find that the expression of CtBPs and CtBP-target genes is elevated both in human psoriatic lesions and in the mouse imiquimod psoriasis model. Moreover, we were able to show that topical treatment with a peptidic inhibitor of CtBP effectively suppresses the CtBP-regulated proinflammatory gene expression and thus attenuates psoriatic inflammation in the imiquimod mouse model. Together, our findings suggest to our knowledge previously unreported strategies for therapeutic modulation of the immune response in inflammatory skin diseases.
Collapse
|
23
|
Zhou X, Chen Y, Cui L, Shi Y, Guo C. Advances in the pathogenesis of psoriasis: from keratinocyte perspective. Cell Death Dis 2022; 13:81. [PMID: 35075118 PMCID: PMC8786887 DOI: 10.1038/s41419-022-04523-3] [Citation(s) in RCA: 196] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 12/14/2021] [Accepted: 01/11/2022] [Indexed: 02/08/2023]
Abstract
Psoriasis is a complex long-lasting inflammatory skin disease with high prevalence and associated comorbidity. It is characterized by epidermal hyperplasia and dermal infiltration of immune cells. Here, we review the role of keratinocytes in the pathogenesis of psoriasis, focusing on factors relevant to genetics, cytokines and receptors, metabolism, cell signaling, transcription factors, non-coding RNAs, antimicrobial peptides, and proteins with other different functions. The critical role of keratinocytes in initiating and maintaining the inflammatory state suggests the great significance of targeting keratinocytes for the treatment of psoriasis.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China
| | - Youdong Chen
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China
| | - Lian Cui
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China.
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China.
| | - Chunyuan Guo
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China.
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China.
| |
Collapse
|
24
|
Loite U, Raam L, Reimann E, Reemann P, Prans E, Traks T, Vasar E, Silm H, Kingo K, Kõks S. The Expression Pattern of Genes Related to Melanogenesis and Endogenous Opioids in Psoriasis. Int J Mol Sci 2021; 22:ijms222313056. [PMID: 34884858 PMCID: PMC8657874 DOI: 10.3390/ijms222313056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
The melanocortin system is a major regulator of stress responses in the skin and is responsible for the induction of melanin synthesis through activation of melanogenesis enzymes. The expression of both melanocortin system genes and melanogenesis enzyme genes is altered in psoriasis, and the focus here was on twelve genes related to the signal transduction between them. Additionally, five endogenous opioid system genes that are involved in cutaneous inflammation were examined. Quantitative real-time-PCR was utilized to measure mRNA expression in punch biopsies from lesional and non-lesional skin of psoriasis patients and from the skin of healthy control subjects. Most of the genes related to melanogenesis were down-regulated in patients (CREB1, MITF, LEF1, USF1, MAPK14, ICAM1, PIK3CB, RPS6KB1, KIT, and ATRN). Conversely, an up-regulation occurred in the case of opioids (PENK, PDYN, and PNOC). The suppression of genes related to melanogenesis is in agreement with the reported reduction in pigmentation signaling in psoriatic skin and potentially results from the pro-inflammatory environment. The increase in endogenous opioids can be associated with their involvement in inflammatory dysregulation in psoriasis.
Collapse
Affiliation(s)
- Ulvi Loite
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
| | - Liisi Raam
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
- Dermatology Clinic, Tartu University Hospital, 31 Raja, 50417 Tartu, Estonia
| | - Ene Reimann
- Institute of Genomics, University of Tartu, 23b/2 Riia, 51010 Tartu, Estonia;
| | - Paula Reemann
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
| | - Ele Prans
- Department of Anaesthesiology and Intensive Care, Tartu University Hospital, 8 L. Puusepa, 51014 Tartu, Estonia;
| | - Tanel Traks
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
- Correspondence:
| | - Eero Vasar
- Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia;
| | - Helgi Silm
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
- Dermatology Clinic, Tartu University Hospital, 31 Raja, 50417 Tartu, Estonia
| | - Külli Kingo
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
- Dermatology Clinic, Tartu University Hospital, 31 Raja, 50417 Tartu, Estonia
| | - Sulev Kõks
- The Perron Institute for Neurological and Translational Science, 8 Verdun St., Nedlands, WA 6009, Australia;
- Centre for Comparative Genomics, Murdoch University, 90 South St., Murdoch, WA 6150, Australia
| |
Collapse
|
25
|
NF-κB1 Contributes to Imiquimod-Induced Psoriasis-Like Skin Inflammation by Inducing Vγ4 +Vδ4 +γδT17 Cells. J Invest Dermatol 2021; 142:1639-1649.e5. [PMID: 34774872 DOI: 10.1016/j.jid.2021.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/04/2021] [Accepted: 11/04/2021] [Indexed: 11/22/2022]
Abstract
Recent studies have identified NF-κB1 as a new disease susceptibility gene for psoriasis. Although accumulating evidence has shown the importance of NF-κB signaling in various cell types in the pathogenesis of psoriasis, it remains unclear how NF-κB1 contributes to the pathogenesis of psoriasis. In this study, we examined psoriasis-like skin diseases induced by topical administration of imiquimod in Nf-κb1‒deficient (Nf-κb1-/-) mice and littermate wild-type (WT) mice. Compared with WT mice, Nf-κb1-/- mice exhibited attenuated skin inflammation. The numbers of Vγ4+Vδ4+γδT17 cells, which cause skin inflammation in this model, were significantly reduced in the skin and draining lymph nodes in imiquimod-treated Nf-κb1-/- mice. Nf-κb1 is preferentially phosphorylated in Vγ4+Vδ4+γδT17 cells in WT mice. In vitro proliferation of Vγ4+Vδ4+γδT17 cells but not conventional CD4+ T cells was significantly impaired in Nf-κb1-/- mice compared with that in WT mice. RNA-sequencing analyses revealed that the expression of E2 factor target genes was decreased in Vγ4+Vδ4+γδT cells by the absence of NF-κB1. Consistently, the cell cycle progression of Vγ4+Vδ4+γδT cells was reduced in Nf-κb1-/- mice compared with that in WT mice. These results suggest that Nf-κb1 plays a crucial role in the pathogenesis of imiquimod-induced psoriasis-like skin inflammation by promoting the proliferation of Vγ4+Vδ4+γδT17 cells.
Collapse
|
26
|
Li D, Wu M. Pattern recognition receptors in health and diseases. Signal Transduct Target Ther 2021; 6:291. [PMID: 34344870 PMCID: PMC8333067 DOI: 10.1038/s41392-021-00687-0] [Citation(s) in RCA: 643] [Impact Index Per Article: 214.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/23/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
Pattern recognition receptors (PRRs) are a class of receptors that can directly recognize the specific molecular structures on the surface of pathogens, apoptotic host cells, and damaged senescent cells. PRRs bridge nonspecific immunity and specific immunity. Through the recognition and binding of ligands, PRRs can produce nonspecific anti-infection, antitumor, and other immunoprotective effects. Most PRRs in the innate immune system of vertebrates can be classified into the following five types based on protein domain homology: Toll-like receptors (TLRs), nucleotide oligomerization domain (NOD)-like receptors (NLRs), retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), C-type lectin receptors (CLRs), and absent in melanoma-2 (AIM2)-like receptors (ALRs). PRRs are basically composed of ligand recognition domains, intermediate domains, and effector domains. PRRs recognize and bind their respective ligands and recruit adaptor molecules with the same structure through their effector domains, initiating downstream signaling pathways to exert effects. In recent years, the increased researches on the recognition and binding of PRRs and their ligands have greatly promoted the understanding of different PRRs signaling pathways and provided ideas for the treatment of immune-related diseases and even tumors. This review describes in detail the history, the structural characteristics, ligand recognition mechanism, the signaling pathway, the related disease, new drugs in clinical trials and clinical therapy of different types of PRRs, and discusses the significance of the research on pattern recognition mechanism for the treatment of PRR-related diseases.
Collapse
Affiliation(s)
- Danyang Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Nițescu DAM, Mușetescu A, Nițescu M, Costescu M, Coman OA. Experimental research in topical psoriasis therapy (Review). Exp Ther Med 2021; 22:971. [PMID: 34335913 PMCID: PMC8290406 DOI: 10.3892/etm.2021.10403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Psoriasis, one of the most prevalent inflammatory diseases in dermatologic pathology, remains a challenge in regards to the therapeutic approach. Topical therapy for psoriasis is a current trending subject as it implies good compliance for the patient, few adverse systemic reactions and a targeted effect. Numerous substances are now being tested, from natural to synthetic compounds and already known substances in improved formulas such as vesicular systems. The aim of this article was to conduct a literature review regarding the topical therapy of psoriasis in animal models, between June, 27, 2019 and July 9, 2020. For this article, the authors conducted extensive research in PubMed with the following keywords: Psoriasis AND (topical OR local) and (therapy OR treatment) AND (mice OR rats). The main new studied substances included lycopene, sodium butyrate, salvianolic acid B, small interfering RNAs (siRNAs) in ionic liquids, albendazole, phosphodiesterase inhibitors, biomimetic reconstituted high-density lipoprotein nanocarrier gel containing microRNA (miRNA)-210 antisense, thymoquinone in ethosomal vesicle, Sea buckthorn oil (Hippophae rhamnoides), nitidine chloride, Melissa officinalis spp. Altissima extract and [1-(4-chloro-3-nitrobenzenesulfonyl)-1H-indol-3-yl]-methanol (CIM). New formulas of already known anti-psoriasis substances such as: Cyclosporine, methotrexate, calcipotriol, tazarotene, protein kinase p38 and integrin α5β1 as a target, are also reviewed. Recent research in topical psoriasis underlines the importance of animal experimental research in dermatology, providing a starting point for developing new therapeutic approaches in one of the most frequently diagnosed chronic dermatologic diseases. Vesicular systems are now providing the best vehicle for topical therapy, thus easing the action of the active substances at their target sites.
Collapse
Affiliation(s)
- Diana Ana-Maria Nițescu
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Alina Mușetescu
- Department of Dermatovenereology, 'Dr. Victor Babes' Clinical Hospital for Infectious and Tropical Diseases, 030303 Bucharest, Romania.,Department of Dermatovenereology, Faculty of Medicine, 'Titu Maiorescu' University, 040051 Bucharest, Romania
| | - Maria Nițescu
- Department of Hygiene and Medical Ecology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Monica Costescu
- Department of Dermatovenereology, 'Dr. Victor Babes' Clinical Hospital for Infectious and Tropical Diseases, 030303 Bucharest, Romania.,Department of Dermatovenereology, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Oana-Andreia Coman
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
28
|
Liu S, Ma H, Zhang H, Deng C, Xin P. Recent advances on signaling pathways and their inhibitors in rheumatoid arthritis. Clin Immunol 2021; 230:108793. [PMID: 34242749 DOI: 10.1016/j.clim.2021.108793] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 12/19/2022]
Abstract
Rheumatoid arthritis (RA) is characterized by systemic synovitis leading to joint destruction in which imbalances in pro-inflammatory and anti-inflammatory cytokines promote the induction of autoimmunity. Some pro-inflammatory cytokines can trigger the signaling pathways which responsible for immune-mediated inflammation in RA, and the activated signaling pathways produce pro-inflammatory cytokines, resulting in aggravation of RA. Hence, understanding of the signaling pathways and their inhibitors might be advantageous in the development of therapeutic targets and new drugs for RA. In the current review, we summarize the signaling pathways involved in the pathogenesis of RA as well as the potential role of specific inhibitors in its management. We hope this paper may serve a reference for future studies on signaling pathways implicated in the pathogenesis of RA and benefit the treatment of RA.
Collapse
Affiliation(s)
- Shuang Liu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Hongxing Ma
- Clinical Laboratory Department, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing 211200, China
| | - Huaxi Zhang
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Chengjie Deng
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Ping Xin
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China.
| |
Collapse
|
29
|
Lai B, Xie X, Li F, Cui Q, Dang E, Luo W, Wang N, Zheng Y, Wang G, Xiao L, Wang N. Xenobiotic Receptor CAR Is Highly Induced in Psoriasis and Promotes Keratinocyte Proliferation. J Invest Dermatol 2021; 141:2895-2907.e7. [PMID: 34097921 DOI: 10.1016/j.jid.2021.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 11/29/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease with abnormal epidermal proliferation. Xenobiotics contribute to the pathogenesis of psoriasis. The mechanism linking xenobiotic stimuli with epidermal proliferation remains largely unknown. In this study, we investigated the role of CAR, a nuclear receptor (NR1I3) responsible for xenobiotics detoxification. We showed that CAR and its target genes were induced in the lesions from patients with psoriasis and imiquimod-treated mice. Proinflammatory cytokines (IL-17A, IL-22, oncostatin M, IL-1α, and TNF-α) synergistically increased the expressions of CAR and its target genes in both human and mouse keratinocytes. Overexpression of CAR promoted the G1/S transition by regulating cyclin E and c-Myc expressions, whereas the silencing of CAR attenuated it. Importantly, a selective CAR agonist 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime or the proinflammatory cytokines induced cyclin E and c-Myc, which were largely blocked by clotrimazole, a selective CAR antagonist, or CAR small interfering RNA. In addition, we showed that topical application of 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene, a selective agonist for mouse CAR, exacerbated the IMQ-induced psoriasis lesions with increased expressions of proliferative and inflammatory markers. In contrast, Car-knockout mice developed significantly milder lesions. In conclusion, these results showed that CAR plays a pathogenic role and, potentially, may be a target for the treatment of psoriasis.
Collapse
Affiliation(s)
- Baochang Lai
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Xinya Xie
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Fan Li
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Qi Cui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Wenhuan Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Ning Wang
- Department of Dermatology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Zheng
- Department of Dermatology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Lei Xiao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China.
| | - Nanping Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
30
|
Sawada Y, Nakatsuji T, Dokoshi T, Kulkarni NN, Liggins MC, Sen G, Gallo RL. Cutaneous innate immune tolerance is mediated by epigenetic control of MAP2K3 by HDAC8/9. Sci Immunol 2021; 6:eabe1935. [PMID: 34021025 PMCID: PMC8363943 DOI: 10.1126/sciimmunol.abe1935] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 04/22/2021] [Indexed: 01/07/2023]
Abstract
The skin typically tolerates exposure to various microbes and chemicals in the environment. Here, we investigated how the epidermis maintains this innate immune tolerance to stimuli that are recognized by Toll-like receptors (TLRs). Loss of tolerance to TLR ligands occurred after silencing of the histone deacetylases (HDACs) HDAC8 and HDAC9 in keratinocytes. Transcriptional analysis identified MAP2K3 as suppressed by HDAC8/9 activity and a potential key intermediary for establishing this tolerance. HDAC8/9 influenced acetylation at H3K9 and H3K27 marks in the MAP2K3 promoter. Proteomic analysis further identified SSRP1 and SUPT16H as associated with HDAC8/9 and responsible for transcriptional elongation of MAP2K3. Silencing of MAP2K3 blocked the capacity of HDAC8/9 to influence cytokine responses. Relevance in vivo was supported by observations of increased MAP2K3 in human inflammatory skin conditions and the capacity of keratinocyte HDAC8/9 to influence dendritic cell maturation and T cell proliferation. Keratinocyte-specific deletion of HDAC8/9 also increased inflammation in mice after exposure to ultraviolet radiation, imiquimod, or Staphylococcus aureus These findings define a mechanism for the epidermis to regulate inflammation in the presence of ubiquitous TLR ligands.
Collapse
Affiliation(s)
- Yu Sawada
- Department of Dermatology, University of California, San Diego, San Diego, CA, USA
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California, San Diego, San Diego, CA, USA
| | - Tatsuya Dokoshi
- Department of Dermatology, University of California, San Diego, San Diego, CA, USA
| | | | - Marc C Liggins
- Department of Dermatology, University of California, San Diego, San Diego, CA, USA
| | - George Sen
- Department of Dermatology, University of California, San Diego, San Diego, CA, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
31
|
Koycheva IK, Vasileva LV, Amirova KM, Marchev AS, Balcheva-Sivenova ZP, Georgiev MI. Biotechnologically Produced Lavandula angustifolia Mill. Extract Rich in Rosmarinic Acid Resolves Psoriasis-Related Inflammation Through Janus Kinase/Signal Transducer and Activator of Transcription Signaling. Front Pharmacol 2021; 12:680168. [PMID: 33986690 PMCID: PMC8111009 DOI: 10.3389/fphar.2021.680168] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
Psoriasis is a common skin pathology, characterized by dysregulation of epidermal keratinocyte function attended by persistent inflammation, suggesting that molecules with anti-inflammatory potential may be effective for its management. Rosmarinic acid (RA) is a natural bioactive molecule known to have an anti-inflammatory potential. Here we examined the effect of biotechnologically produced cell suspension extract of Lavandula angustifolia Mill (LV) high in RA content as treatment for psoriasis-associated inflammation in human keratinocytes. Regulatory genes from the nuclear factor kappa B (NF-κB) and Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathways were upregulated upon stimulation with a combination of interferon gamma (IFN-γ), interleukin (IL)-17A and IL-22. We also observed that both LV extract and RA could inhibit JAK2, leading to reduced STAT1 phosphorylation. Further, we demonstrated that LV extract inhibited phosphoinositide 3-kinases (PI3K) and protein kinase B (AKT), which could be implicated in reduced hyperproliferation in keratinocytes. Collectively, these findings indicate that the biotechnologically produced LV extract resolved psoriasis-like inflammation in human keratinocytes by interfering the JAK2/STAT1 signaling pathway and its effectiveness is due to its high content of RA (10%). Hence, both LV extract and pure RA possess the potential to be incorporated in formulations for topical application as therapeutic approach against psoriasis.
Collapse
Affiliation(s)
- Ivanka K Koycheva
- Laboratory of Metabolomics, Department of Biotechnology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Plovdiv, Bulgaria.,Department Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, Plovdiv, Bulgaria
| | - Liliya V Vasileva
- Department Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, Plovdiv, Bulgaria
| | - Kristiana M Amirova
- Laboratory of Metabolomics, Department of Biotechnology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Plovdiv, Bulgaria.,Department Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, Plovdiv, Bulgaria
| | - Andrey S Marchev
- Laboratory of Metabolomics, Department of Biotechnology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Plovdiv, Bulgaria.,Department Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, Plovdiv, Bulgaria
| | - Zhivka P Balcheva-Sivenova
- Laboratory of Metabolomics, Department of Biotechnology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Plovdiv, Bulgaria.,Department Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, Plovdiv, Bulgaria
| | - Milen I Georgiev
- Laboratory of Metabolomics, Department of Biotechnology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Plovdiv, Bulgaria.,Department Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, Plovdiv, Bulgaria
| |
Collapse
|
32
|
Srivastava A, Luo L, Lohcharoenkal W, Meisgen F, Pasquali L, Pivarcsi A, Sonkoly E. Cross-talk between IFN-γ and TWEAK through miR-149 amplifies skin inflammation in psoriasis. J Allergy Clin Immunol 2021; 147:2225-2235. [PMID: 33705829 DOI: 10.1016/j.jaci.2020.12.657] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/12/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Psoriasis is a chronic inflammatory skin disease with disturbed interplay between immune cells and keratinocytes. A strong IFN-γ signature is characteristic for psoriasis skin, but the role of IFN-γ has been elusive. MicroRNAs are short RNAs regulating gene expression. OBJECTIVE Our aim was to investigate the role of miR-149 in psoriasis and in the inflammatory responses of keratinocytes. METHODS miR-149 expression was measured by quantitative RT-PCR in keratinocytes isolated from healthy skin and lesional and nonlesional psoriasis skin. Synthetic miR-149 was injected intradermally into the back skin of mice, and imiquimod was applied to induce psoriasis-like skin inflammation, which was then evaluated at the morphologic, histologic, and molecular levels. miR-149 was transiently overexpressed or inhibited in keratinocytes in combination with IFN-γ- and/or TNF-related weak inducer of apoptosis (TWEAK)-treatment. RESULTS Here we report a microRNA-mediated mechanism by which IFN-γ primes keratinocytes to inflammatory stimuli. Treatment with IFN-γ results in a rapid and long-lasting suppression of miR-149 in keratinocytes. Depletion of miR-149 in keratinocytes leads to widespread transcriptomic changes and induction of inflammatory mediators with enrichment of the TWEAK pathway. We show that IFN-γ-mediated suppression of miR-149 leads to amplified inflammatory responses to TWEAK. TWEAK receptor (TWEAKR/Fn14) is identified as a novel direct target of miR-149. The in vivo relevance of this pathway is supported by decreased miR-149 expression in psoriasis keratinocytes, as well as by the protective effect of synthetic miR-149 in the imiquimod-induced mouse model of psoriasis. CONCLUSION Our data define a new mechanism, in which IFN-γ primes keratinocytes for TWEAK-induced inflammatory responses through suppression of miR-149, promoting skin inflammation.
Collapse
Affiliation(s)
- Ankit Srivastava
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden
| | - Longlong Luo
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden
| | - Warangkana Lohcharoenkal
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden
| | - Florian Meisgen
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden
| | - Lorenzo Pasquali
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden
| | - Andor Pivarcsi
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden; Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Enikö Sonkoly
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden; Unit of Dermatology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
33
|
Dihydroartemisinin prevents dextran sodium sulphate-induced colitisthrough inhibition of the activation of NLRP3 inflammasome and p38 MAPK signaling. Int Immunopharmacol 2020; 88:106949. [PMID: 32892075 DOI: 10.1016/j.intimp.2020.106949] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/17/2020] [Accepted: 08/27/2020] [Indexed: 12/24/2022]
Abstract
Dihydroartemisinin (DHA), a sesquiterpene lactone derived from artemisinin, has been reported to possess anti-inflammation and anti-cancer activities. But its underlying protective mechanisms on dextran sodium sulphate (DSS)-induced colitis remain rarely reported. We applied a network pharmacology approach to predict the collective targets of DHA and acute colitis. GO and KEGG analyses were performed to investigate the enriched biological functions and signaling pathways of the collective targets. Furthermore, a DSS-induced colitis model was established to observe the protective effects of DHA. 83 common targets of DHA and acute colitis were identified and predominantly involved in several inflammation-related signaling pathways in colitis such as NOD-like receptor and MAPK signaling pathways. Additionally, DHA in vivo improved the clinical symptoms, reduced the production of pro-inflammatory factors IL-1β, IL-6 and TNF-α, and suppressed the formation of NLRP3 inflammasome. Moreover, DHA inhibited the phosphorylation of NF-κB p65 and p38 MAPK, but upregulated PPARγ and Ki-67 levels compared to the DSS group. Additionally, we found that DHA suppressed p38 activator-induced pro-inflammatory response, and p38 inhibitor attenuated the clinical symptoms and reduced the expression levels of pro-inflammatory mediators and NLRP3 while up-regulated the expression levels of PPARγ and Ki-67. Molecular docking analysis further verified the binding mode towards the DHA and p38 MAPK. In conclusion, DHA could protect DSS-induced colitis via suppressing the activation of NLRP3 inflammasome and p38 MAPK signaling.
Collapse
|
34
|
Jaganjac M, Milkovic L, Gegotek A, Cindric M, Zarkovic K, Skrzydlewska E, Zarkovic N. The relevance of pathophysiological alterations in redox signaling of 4-hydroxynonenal for pharmacological therapies of major stress-associated diseases. Free Radic Biol Med 2020; 157:128-153. [PMID: 31756524 DOI: 10.1016/j.freeradbiomed.2019.11.023] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023]
Abstract
Modern analytical methods combined with the modern concepts of redox signaling revealed 4-hydroxy-2-nonenal (4-HNE) as particular growth regulating factor involved in redox signaling under physiological and pathophysiological circumstances. In this review current knowledge of the relevance of 4-HNE as "the second messenger of reactive oxygen species" (ROS) in redox signaling of representative major stress-associated diseases is briefly summarized. The findings presented allow for 4-HNE to be considered not only as second messenger of ROS, but also as one of fundamental factors of the stress- and age-associated diseases. While standard, even modern concepts of molecular medicine and respective therapies in majority of these diseases target mostly the disease-specific symptoms. 4-HNE, especially its protein adducts, might appear to be the bioactive markers that would allow better monitoring of specific pathophysiological processes reflecting their complexity. Eventually that could help development of advanced integrative medicine approach for patients and the diseases they suffer from on the personalized basis implementing biomedical remedies that would optimize beneficial effects of ROS and 4-HNE to prevent the onset and progression of the illness, perhaps even providing the real cure.
Collapse
Affiliation(s)
- Morana Jaganjac
- Qatar Analytics & BioResearch Lab, Anti Doping Lab Qatar, Sport City Street, Doha, Qatar
| | - Lidija Milkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Div. of Molecular Medicine, Bijenicka 54, Zagreb, Croatia
| | - Agnieszka Gegotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Marina Cindric
- University of Zagreb, School of Medicine, Div. of Pathology, University Hospital Centre Zagreb, Kispaticeva 12, Zagreb, Croatia
| | - Kamelija Zarkovic
- University of Zagreb, School of Medicine, Div. of Pathology, University Hospital Centre Zagreb, Kispaticeva 12, Zagreb, Croatia
| | - Elzbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Neven Zarkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Div. of Molecular Medicine, Bijenicka 54, Zagreb, Croatia.
| |
Collapse
|
35
|
Tang KW, Lin ZC, Wang PW, Alalaiwe A, Tseng CH, Fang JY. Facile skin targeting of a thalidomide analog containing benzyl chloride moiety alleviates experimental psoriasis via the suppression of MAPK/NF-κB/AP-1 phosphorylation in keratinocytes. J Dermatol Sci 2020; 99:90-99. [PMID: 32622642 DOI: 10.1016/j.jdermsci.2020.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Thalidomide can be a TNF-α inhibitor for treating skin inflammation. This drug exhibits a strong toxicity that limits its application. OBJECTIVE We synthesized a thalidomide analog containing the benzyl chloride group (2-[1-(3-chlorobenzyl)-2,6-dioxopiperidin-3-yl]isoindoline-1,3-dione, CDI) to examine anti-inflammatory activity against psoriasis. METHODS The evaluation was conducted by the experimental platforms of in vitro TNF-α- or imiquimod (IMQ)-stimulated HaCaT cells and in vivo IMQ-induced psoriasiform plaque. RESULTS Using the in vitro keratinocyte model, we demonstrated a greater inhibition of IL-1β, IL-6, and IL-24 by CDI than by thalidomide. No significant cytotoxicity was observed at 100 μM. CDI delivered facilely into the skin with a cutaneous targeting ability 228-fold greater than thalidomide. CDI caused a negligible irritation on healthy mouse skin. We showed that topically applied CDI reduced IMQ-induced red scaly lesions, hyperplasia, microabscesses, and cytokine expression in the mouse model. The skin-barrier function measured by transepidermal water loss (TEWL) could be partially recovered from 50.6-36.3 g/m2/h by CDI. The mechanistic study showed that CDI suppressed cytokine production by inhibiting the phosphorylation of NF-κB and AP-1 via MAPK pathways. CONCLUSION CDI would be beneficial for the development of a therapeutic agent against psoriasis.
Collapse
Affiliation(s)
- Kai-Wei Tang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zih-Chan Lin
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Pei-Wen Wang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Ahmed Alalaiwe
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Chih-Hua Tseng
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Pharmacy, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan; Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan.
| |
Collapse
|
36
|
Ex vivo culture of lesional psoriasis skin for pharmacological testing. J Dermatol Sci 2019; 97:109-116. [PMID: 31948839 DOI: 10.1016/j.jdermsci.2019.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/18/2019] [Accepted: 12/24/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Psoriasis is a chronic, inflammatory skin disorder resulting from a complex interplay between immune and skin cells via release of soluble mediators. While a lot is known about the molecular mechanisms behind psoriasis pathogenesis, there is still a need for preclinical research models that accuratelyreplicate the disease. OBJECTIVE This study aimed to develop and characterize ex vivo culture of psoriasis skin as a model for pharmacological testing, where the immunological events of psoriasis can be followed. METHODS Full thickness punch biopsies of lesional psoriasis skin were cultured in submerged conditions up to 144 h followingin situ T cell stimulation with rhIL-23 and anti-CD3 and anti-CD28 antibodies. The T cell mediated skin inflammation was assessed by gene and protein l analysis for a panel of inflammatory mediators. Tissue integrity and morphology were evaluated by histological analysis. RESULTS T cell stimulation resulted in functional and psoriasis specificin situ activation of T cells. The expression levels of most of the proinflammatory mediators related to both immune and skin cells were comparable to these in freshly isolated tissue at 48 and 96 h of culture. Tissue integrity and morphology were sustained up to 96 h. Treatment with a corticosteroid reduced the expression of several pro-inflammatory cytokines and chemokines, whereas anti-IL-17A antibody treatment reduced the expression of the IL-17A downstream markers IL-8 and DEFB4. CONCLUSION By preserving keyimmunopathological mechanisms of psoriasis, ex vivo culture of psoriasis skin can be used for the investigation of inflammatory processes of psoriasis and for preclinical drug discovery research.
Collapse
|