1
|
Majchrzycka M, Wegner J, Adamski Z, Jenerowicz D. Interleukins 30 and 27 in psoriasis and inflammation. Postepy Dermatol Alergol 2025; 42:1-4. [PMID: 40114761 PMCID: PMC11921921 DOI: 10.5114/ada.2025.147548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/06/2024] [Indexed: 03/22/2025] Open
Abstract
This review explores the roles of interleukin-30 (IL-30) and interleukin-27 (IL-27) in inflammation and autoimmune diseases, with a focus on psoriasis. The two coexisting cytokines should be analysed in conjunction as their actions are antagonistic in vivo. While IL-27 exhibits diverse anti-inflammatory mechanisms, the understanding of IL-30's functions remains limited. Studies suggest that IL-27 may play a role in regulating psoriasis, but findings are inconsistent. IL-30 shows promise in mitigating psoriatic lesions and suppressing inflammatory responses. However, research on IL-30's involvement in autoimmune diseases presents conflicting results. This article provides a literature review on the complex correlations between cytokines, their role in the pathogenesis of psoriasis, inflammation, carcinogenesis, and autoimmune diseases, and provides a detailed picture of the interplay between IL-27 and IL-30 to uncover novel therapeutic targets for psoriasis and other autoimmune conditions.
Collapse
Affiliation(s)
| | - Joanna Wegner
- Department of Dermatology, Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Mainz, Germany
| | - Zygmunt Adamski
- Department of Dermatology, University Clinical Hospital, Poznan, Poland
| | - Dorota Jenerowicz
- Department of Dermatology, University Clinical Hospital, Poznan, Poland
| |
Collapse
|
2
|
Kang Z, Du Y, Cui R, Wang Q, Chen M, Wang YH, Wei JCC, Dai SM. Psoriasis increases the risk of Sjögren's syndrome: evidence from a propensity score-matched cohort study and transcriptomic analysis. BMC Med 2025; 23:26. [PMID: 39838439 PMCID: PMC11752802 DOI: 10.1186/s12916-025-03856-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Despite the well-documented immune dysregulation in both psoriasis and Sjögren's syndrome (SS), the specific link between these two autoimmune diseases has not been extensively explored. The present study aims to investigate the impact of psoriasis on the risk of SS. METHODS A retrospective cohort study using TriNetX data compared SS development in patients with psoriasis and controls using propensity score matching, Kaplan-Meier curves, and Cox models. Transcriptome data were analyzed to identify shared differentially expressed genes and pathways between the two diseases. RESULTS A total of 293,905 patients with psoriasis and an equal number of individuals without psoriasis were included. After propensity score matching, the baseline characteristics of both groups were balanced. During the follow-up period, 3339 patients with psoriasis and 1937 individuals without psoriasis developed SS. The Kaplan-Meier curves indicated a significantly higher risk of developing SS in the psoriasis group compared to the non-psoriasis group. Upon adjustment for multiple confounding factors, the risk of developing SS in the psoriasis group was 50% higher in the psoriasis group than the non-psoriasis group (hazard ratio [HR] 1.50, 95% confidence interval [CI] 1.42-1.58). Subgroup analyses confirmed the elevated risk of SS associated with psoriasis. Patients with psoriatic arthritis (PsA) and those treated with biological agents had an even higher risk of developing SS. Transcriptomic analysis revealed potential shared pathogenesis of psoriasis and SS involving cellular proliferation, immune cell recruitment, cytokine secretion, and the interferon response to viral infections. CONCLUSIONS Psoriasis might increase the risk of developing SS, which is augmented by PsA. The overlapping immunological mechanisms may underlie the co-occurrence of psoriasis and SS.
Collapse
Affiliation(s)
- Zijian Kang
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Du
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ran Cui
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Wang
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Chen
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Hsun Wang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - James Cheng-Chung Wei
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China.
- Department of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan.
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.
- Institute of Medicine/Department of Nursing, Chung Shan Medical University, Taichung, Taiwan.
| | - Sheng-Ming Dai
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Wang X, Wang S, Luo X. Clinical Study on the Induction of Psoriasis Flare-Ups by PM2.5 Air Pollutants via Immune Barrier Dysfunction. J Cutan Med Surg 2025; 29:18-23. [PMID: 39506319 DOI: 10.1177/12034754241287555] [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: 11/08/2024]
Abstract
OBJECTIVE This study explores the clinical correlation between the air pollutant PM2.5 and the induction of psoriasis flare-ups through the disruption of the immune barrier. METHODS Air quality data, the average mass concentration of the primary atmospheric pollutant PM2.5, and meteorological data spanning from November 2023 to March 2024 were gathered from 9 air quality monitoring stations situated within our city, courtesy of the local Meteorological Information Center. Psoriasis cases were sourced from our hospital's dermatology department, encompassing patients diagnosed and treated from November 2023 to March 2024 and residing within the city. RESULTS From January 2023 to May 2024, aberrant expression of Th1 cells and Th2 cells was observed in psoriasis flare-up patients. Spearman correlation coefficient analysis revealed a positive correlation between PM2.5, PM10, and Th1 cells, and a negative correlation with Th2 cells, exhibiting significant differences (P < .05). Supplemental Figure 1 illustrates an increase in psoriasis flare-ups on the fifth day following a unit increase in PM2.5 concentration, with an excess risk (ER) value of 0.046 [95% confidence interval (CI): 0.137-0.893]. Conversely, after an increase of 1 unit of PM10, there was a decrease on the third and fourth days, followed by increases on the fifth, sixth, and seventh days, with ER values of 0.038 (95% CI: 0.013-0.067), 0.045 (95% CI: 0.019-0.073), 0.051 (95% CI: 0.034-0.078), 0.057 (95% CI: 0.045-0.083), and 0.061 (95% CI: 0.051-0.087), respectively. CONCLUSION Air pollutant PM2.5 could potentially exacerbate psoriasis flare-ups by compromising the immune barrier, suggesting a plausible mechanism linked to the onset of this condition.
Collapse
Affiliation(s)
- Xueliang Wang
- Department of Dermatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuixia Wang
- Department of Dermatology, Affiliated Hospital of Army Military Medical University Officer School, Shijiazhuang, China
| | - Xuejiao Luo
- Department of Dermatology, Affiliated Hospital of Army Military Medical University Officer School, Shijiazhuang, China
| |
Collapse
|
4
|
Chen L, Wang X, Liu C, Chen X, Li P, Qiu W, Guo K. Integrative analysis of gene and microRNA expression profiles reveals candidate biomarkers and regulatory networks in psoriasis. Medicine (Baltimore) 2024; 103:e39002. [PMID: 39028999 PMCID: PMC11398825 DOI: 10.1097/md.0000000000039002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 06/28/2024] [Indexed: 07/21/2024] Open
Abstract
Psoriasis (PS) is a chronic inflammatory skin disease with a long course and tendency to recur, the pathogenesis of which is not fully understood. This article aims to identify the key differentially expressed genes (DEGs) and microRNA (miRNAs) of PS, construct the core miRNA-mRNA regulatory network, and investigate the underlying molecular mechanism through integrated bioinformatics approaches. Two gene expression profile datasets and 2 miRNA expression profile datasets were downloaded from the gene expression omnibus (GEO) database and analyzed by GEO2R. Intersection DEGs and intersection differentially expressed miRNAs (DEMs) were each screened. The Metascape database and R software were used to perform enrichment analysis of intersecting DEGs and study their functions. Target genes of DEMs were predicted from the online database miRNet. The protein-protein interaction files of the overlapping target genes were obtained from string and the miRNA-mRNA network was constructed by Cytoscape software. In addition, the online web tool CIBERSORT was used to analyze the immune infiltration of dataset GSE166388, and the relative abundance of 22 immune cells in the diseased and normal control tissues was calculated and assessed. Finally, quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to verify the relative expression of the screened miRNAs and mRNAs to assess the applicability of DEMs and DEGs as biomarkers in PS. A total of 205 mating DEGs and 6 mating DEMs were screened. 103 dysregulated crossover genes from 205 crossover DEGs and 7878 miRNA target genes were identified. The miRNA-mRNA regulatory network was constructed and the top 10 elements were obtained from CytoHubba, including hsa-miR-146a-5p, hsa-miR-17-5p, hsa-miR-106a-5p, hsa-miR-18a-5p, CDK1, CCNA2, CCNB1, MAD2L1, RRM2, and CCNB2. QRT-PCR revealed significant differences in miRNA and gene expression between inflammatory and normal states. In this study, the miRNA-mRNA core regulator pairs hsa-miR-146a-5p, hsa-miR-17-5p, hsa-miR-106a-5p, hsa-miR-18a-5p, CDK1, CCNA2, CCNB1, MAD2L1, RRM2, and CCNB2 may be involved in the course of PS. This study provides new insights to discover new potential targets and biomarkers to further investigate the molecular mechanism of PS.
Collapse
Affiliation(s)
- Lu Chen
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Xiaochen Wang
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Chang Liu
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Xiaoqing Chen
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Peng Li
- Department of Dermatology, Wuhan Central Hospital, Wuhan, Hubei, PR China
| | - Wenhong Qiu
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Kaiwen Guo
- Department of Pathogenic Biology, Wuhan University of Science and Technology, Medical College, Wuhan, Hubei, PR China
| |
Collapse
|
5
|
Zhang M, Li D, Zhu J, Xia X, Zhang H, Wu J, Wang S, Deng A, Wen Q, Tan J, Hao J, Jiang J, Bao X, Sun G, Lu J, Yang Q, Yang H, Cao G, Yin Z, Wang Q. IL-27 disturbs lipid metabolism and restrains mitochondrial activity to inhibit γδ T17 cell-mediated skin inflammation. Cell Death Dis 2024; 15:491. [PMID: 38982043 PMCID: PMC11233514 DOI: 10.1038/s41419-024-06887-0] [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: 02/22/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
IL-17+ γδ T cells (γδ T17) are kick-starters of inflammation due to their strict immunosurveillance of xenobiotics or cellular damages and rapid response to pro-inflammatory stimulators. IL-27 is a well-recognized pleiotropic immune regulator with potent inhibitory effects on type 17 immune responses. However, its actions on γδ T17 mediated inflammation and the underlying mechanisms are less well understood. Here we find that IL-27 inhibits the production of IL-17 from γδ T cells. Mechanistically, IL-27 promotes lipolysis while inhibits lipogenesis, thus reduces the accumulation of lipids and subsequent membrane phospholipids, which leads to mitochondrial deactivation and ensuing reduction of IL-17. More importantly, Il27ra deficient γδ T cells are more pathogenic in an imiquimod-induced murine psoriasis model, while intracutaneous injection of rmIL-27 ameliorates psoriatic inflammation. In summary, this work uncovered the metabolic basis for the immune regulatory activity of IL-27 in restraining γδ T17 mediated inflammation, which provides novel insights into IL-27/IL-27Ra signaling, γδ T17 biology and the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Mingyue Zhang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Dehai Li
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jing Zhu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
| | - Xue Xia
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Hua Zhang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Jie Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
| | - Shengli Wang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Anyi Deng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Qiong Wen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Jingyi Tan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Jun Jiang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
| | - Xiucong Bao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Guodong Sun
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China
| | - Jiajing Lu
- Department of Dermatology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
- Institute of Psoriasis, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Quanli Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
| | - Hengwen Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
| | - Guangchao Cao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
| | - Qian Wang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
| |
Collapse
|
6
|
Chen ZY, Yu XQ, Xiang YY, Liu LH, Yin XP. Moyamoya syndrome may result from psoriasis: Four case reports. World J Clin Cases 2024; 12:1830-1836. [PMID: 38660070 PMCID: PMC11036477 DOI: 10.12998/wjcc.v12.i10.1830] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/28/2024] [Accepted: 03/11/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Moyamoya syndrome (MMS) is a group of diseases that involves more than one underlying disease and is accompanied by moyamoya vascular phenomena. Psoriasis is a chronic immune skin disease closely linked to high blood pressure and heart disease. However, psoriasis-related MMS has not been reported. CASE SUMMARY We collected data on patients with stroke due to MMS between January 2017 and December 2019 and identified four cases of psoriasis. Case histories, imaging, and hematological data were collected. The average age of the initial stroke onset was 58.25 ± 11.52 years; three cases of hemorrhagic and one case of ischemic stroke were included. The average duration from psoriasis confirmation to the initial MMS-mediated stroke onset was 17 ± 3.56 years. All MMS-related stenoses involved the bilateral cerebral arteries: Suzuki grade III in one case, grade IV in two cases, and grade V in one case. Abnormally elevated plasma interleukin-6 levels were observed in four patients. Two patients had abnormally elevated immunoglobulin E levels, and two had thrombocytosis. All four patients received medication instead of surgery. With an average follow-up time of 2 years, two causing transient ischemic attacks occurred in two patients, and no hemorrhagic events occurred. CONCLUSION Psoriasis may be a potential risk factor for MMS. Patients with psoriasis should be screened for MMS when they present with neurological symptoms.
Collapse
Affiliation(s)
- Zhi-Ying Chen
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang 332000, Jiangxi Province, China
| | - Xiao-Qin Yu
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang 332000, Jiangxi Province, China
| | - Yuan-Yuan Xiang
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang 332000, Jiangxi Province, China
| | - Ling-Hua Liu
- Department of Dermatology, The Affiliated Hospital of Jiujiang University, Jiujiang 332000, Jiangxi Province, China
| | - Xiao-Ping Yin
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang 332000, Jiangxi Province, China
| |
Collapse
|
7
|
Chen Y, Zhu M, Hu J, He S, Li S, Liu B, Yang J. IL-27 Alleviates Airway Inflammation and Airway Hyperresponsiveness in Asthmatic Mice by Targeting the CD39/ATP Axis of Dendritic Cells. Inflammation 2024; 47:807-821. [PMID: 38117410 DOI: 10.1007/s10753-023-01945-9] [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: 10/17/2023] [Revised: 11/18/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Interleukin-27 receptor (IL-27R) is expressed in a variety of immune cells and structural cells, including dendritic cells. The mechanism of IL-27 in asthma has not been fully elucidated. This study aimed to examine whether IL-27 regulated the CD39/ATP axis of dendritic cells in asthma. Our results showed that in ovalbumin (OVA)-induced asthma mouse model, IL-27Rα-/- asthmatic mice showed increased airway resistance, increased infiltration of inflammatory cells in lung tissue, proliferation of goblet cells, enhanced expression of Muc5 AC around airway epithelium, increased total number of cells and eosinophils, increased levels of total IgE, OVA-IgE, IL-4, IL-5, IL-13 and IL-17 A, and increased expression of transcription factors GATA-3 and RORγt in lung tissue. The expression of CD39 mRNA and protein in the lung tissue of IL-27Rα-/- asthmatic mice decreased, and the expression of NLRP3, ASC and Caspase-1 in NLRP3 inflammasome components increased. The concentration of ATP was significantly increased compared with WT asthmatic mice. In vitro experiments showed that the expression of CD39 in lung dendritic cells of IL-27Rα-/- asthmatic mice decreased, while the expression of NLRP3 inflammasome components NLRP3, ASC and Caspase-1 increased. These findings indicate that IL-27 directly and indirectly regulates immunoinflammatory responses in asthma by acting on dendritic cells CD39/ATP Axis.
Collapse
Affiliation(s)
- Yifei Chen
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Miaojuan Zhu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiahao Hu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shaojun He
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuhua Li
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bing Liu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiong Yang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
8
|
Lobão B, Lourenço D, Giga A, Mendes-Bastos P. From PsO to PsA: the role of T RM and Tregs in psoriatic disease, a systematic review of the literature. Front Med (Lausanne) 2024; 11:1346757. [PMID: 38405187 PMCID: PMC10884248 DOI: 10.3389/fmed.2024.1346757] [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: 11/29/2023] [Accepted: 01/25/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Psoriasis (PsO) is a chronic skin condition driven by immune mediators like TNFα, INFγ, IL-17, and IL-23. Psoriatic arthritis (PsA) can develop in PsO patients. Although psoriatic lesions may apparently resolve with therapy, subclinical cutaneous inflammation may persist. The role of tissue-resident memory T-cells (TRM), and regulatory T cells (Tregs) that also contribute to chronic inflammation are being explored in this context. This systematic review explores TRM and Tregs in psoriatic disease (PsD) and its progression. Methods A systematic review, following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, was performed using Pubmed® and Web of Science™ databases on June 3rd 2023, using patient/population, intervention, comparison, and outcomes (PICO) criteria limited to the English language. Results A total of 62 reports were identified and included. In PsO, chronic inflammation is driven by cytokines including IL-17 and IL-23, and cellular mediators such as CD8+ and CD4+ T cells. TRM contributes to local inflammation, while Tregs may be dysfunctional in psoriatic skin lesions. Secukinumab and guselkumab, which target IL-17A and the IL-23p19 subunit, respectively, have different effects on CD8+ TRM and Tregs during PsO treatment. Inhibition of IL-23 may provide better long-term results due to its impact on the Treg to CD8+ TRM ratio. IL-23 may contribute to inflammation persisting even after treatment. In PsA, subclinical enthesitis is perceived as an early occurence, and Th17 cells are involved in this pathogenic process. Recent EULAR guidelines highlight the importance of early diagnosis and treatment to intercept PsA. In PsA, CD8+ TRM cells are present in synovial fluid and Tregs are reduced in peripheral blood. The progression from PsO to PsA is marked by a shift in immune profiles, with specific T-cells subsets playing key roles in perpetuating inflammation. Early intervention targeting TRM cells may hold promising, but clinical studies are limited. Ongoing studies such as IVEPSA and PAMPA aim to improve our knowledge regarding PsA interception in high-risk PsO patients, emphasizing the need for further research in this area. Conclusion Early intervention is crucial for PsO patients at high risk of PsA; T cells, particularly type 17 helper T cells, and CD8+ cells are key in the progression from PsO-to-PsA. Early targeting of TRM in PsD shows promise but more research is needed.
Collapse
Affiliation(s)
- Bárbara Lobão
- Instituto Português de Reumatologia, Lisboa, Portugal
- Centro Hospitalar de Setúbal, Setúbal, Portugal
| | | | - Ana Giga
- Janssen Portugal, Oeiras, Portugal
| | | |
Collapse
|
9
|
Yan L, Yu C, Zhao Z, Zhang Y, Wang R, Li C. Higher IL-9 Level is Associated with Psoriasis Vulgaris Complicated by Metabolic Syndrome. Clin Cosmet Investig Dermatol 2023; 16:2297-2307. [PMID: 37641663 PMCID: PMC10460594 DOI: 10.2147/ccid.s422355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Purpose The underlying pathophysiology linking psoriasis vulgaris (PV) and metabolic syndrome (MetS) is not fully understood. The present study aimed to investigate the serum level of interleukin (IL)-9 and tissue levels of IL-9 and its receptor in PV patients with MetS and analyze the correlation of IL-9 levels with psoriasis disease severity and MetS. Methods This study enrolled 75 PV patients with MetS, 57 PV patients without MetS, 20 healthy blood donors, and 7 healthy skin donors. Clinical, socio-demographic, and anthropometric data were obtained from all individuals. Fasting blood glucose, insulin, lipid profile levels, and serum levels of IL-9 and IL-17A were measured. The expression of IL-9 and its receptor in skin specimens in PV patients and healthy controls was determined using immunohistochemistry. Normal human epidermal keratinocytes were stimulated with five pro-inflammatory cytokines (tumor necrosis factor-α, oncostatin M, IL-22, IL-17A, and IL-1α) to establish a psoriatic keratinocyte model and subsequently treated with IL-9. Their mRNA levels of antimicrobial peptides and chemokines were measured using quantitative real-time polymerase chain reaction. Results Serum level of IL-9 and tissue levels of IL-9 and its receptor were upregulated in PV patients with MetS. IL-9 level was positively correlated to IL-17A level; however, no significant correlation of IL-9 level with psoriasis area severity index was observed. IL-9 level had a positive correlation with the presence of MetS and its components. Correspondingly, IL-9 level positively correlated with waist circumference, body mass index, homeostasis model assessment-insulin resistance, blood pressure, and triglyceride level and negatively correlated with high-density lipoprotein cholesterol level. Additionally, IL-9 stimulated the expression of antimicrobial peptides and chemokines in a psoriatic keratinocyte model. Conclusion Our findings confirmed that higher IL-9 level is associated with PV complicated by MetS, suggesting that IL-9 may be a link between PV and MetS.
Collapse
Affiliation(s)
- Liang Yan
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
- Department of Dermatology, General Hospital of Central Theater Command of PLA, Wuhan, Hubei, 430070, People’s Republic of China
| | - Chongli Yu
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Zhenkai Zhao
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Yuan Zhang
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Rui Wang
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Chengxin Li
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| |
Collapse
|
10
|
Cruz CJG, Yang CC. Clinical application of serum biomarkers for detecting and monitoring of chronic plaque psoriasis. Front Mol Biosci 2023; 10:1196323. [PMID: 37546687 PMCID: PMC10403288 DOI: 10.3389/fmolb.2023.1196323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/29/2023] [Indexed: 08/08/2023] Open
Abstract
Psoriasis, a chronic, multisystemic inflammatory disease affecting millions of people globally, manifests as erythematous, thick, scaly plaques on the skin. Clinical evaluation remains to be the benchmark for diagnosis and monitoring of this debilitating disease. With current advancements in targeted molecular therapy for psoriasis such as biologics, molecular detection methods may also help guide clinical decisions and therapeutic strategies through quantification of circulating biomarkers, which could reflect the underlying pathogenic events happening at a certain point of the disease course. In this review, we will discuss how biomarkers are detected in serum samples using enzyme-linked immunosorbent assay (ELISA). This review will feature candidate biomarkers supported by clinical data for psoriasis including, but not limited to, cytokines, chemokines, adipokines, and antimicrobial peptides. A better understanding of the common method used for biomarker detection would enable physicians to interpret and correlate laboratory results with the disease pathogenesis and clinical outcomes, e.g., severity assessment and/or therapeutic response. With better health outcomes as the main goal, the utility of such information to evaluate and even predict treatment response would be a major step closer towards patient-tailored management.
Collapse
Affiliation(s)
- Criselda Jean G. Cruz
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Chao-Chun Yang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
11
|
Zhang H, Ren C, Liu Q, Wang Q, Wang D. TFAP2C exacerbates psoriasis-like inflammation by promoting Th17 and Th1 cells activation through regulating TEAD4 transcription. Allergol Immunopathol (Madr) 2023; 51:124-134. [PMID: 37169570 DOI: 10.15586/aei.v51i3.854] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/20/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Psoriasis is one of the chronic and autoimmune skin diseases. It is important to uncover the mechanisms underlying the psoriasis. Transcription factor activator protein (TFAP-2) gamma, also known as AP2-gamma, is a protein encoded by the TFAP2C gene. Immune-mediated pathophysiological processes could be linked to psoriasis, but the mechanism is still unclear. Therefore, to date the cause of psoriasis has not been understood completely. MATERIALS AND METHODS Psoriasis is a complex disease triggered by genetic, immunological, and environmental stimuli. Keratinocytes play an important role in both initiation and maintenance phases of psoriasis. A psoriatic keratinocyte model was established by stimulating high sensitivity of human epidermal keratinocytes (HaCaT) to topoisomerase inhibitor cell lines using the accumulation of M5 cytokines comprising interleukin (IL)-17A, IL-22, oncostatin M, IL-1α, and tumor necrosis factor-α (TNF-α). The TFAP2C and transcriptional enhanced associate domain 4 (TEAD4) genes expression was evaluated by reverse transcription-quantitative polymerase chain reaction. Western blot analysis was used to examine protein expression. Cell viability (quantitative) of keratinocytes, including cytotoxicity, proliferation, and cell activation, was evaluated by the MTT assay. The relative percentage values of interleukin (IL)-17a, interferon gamma, and IL-4+ cells were measured by flow cytometry. Accordingly, chromatin immunoprecipitation and luciferase reporter assays were applied to evaluate the binding affinity of TFAP2C and TEAD4 promoter. RESULTS Level of the TFAP2C gene was elevated in the lesional skin of psoriasis patients. On the other hand, silencing of the TFAP2C gene suppressed the proliferation and inflammatory response in M5-induced keratinocytes. In addition, inhibition of TFAP2C alleviated imiquimod (IMQ)-induced skin injury in mice model. We also observed that suppression of TFAP2C inhibited the activation of T-helper 17 (Th17) and Th1 cells in IMQ-induced mice model. Mechanically, TFAP2C promoted TEAD4 transcriptional activation. CONCLUSION TFAP2C exacerbated psoriasis-like inflammation by increasing the activation of Th17 and Th1 cells by regulating TEAD4 transcription. This finding clearly indicated that TFAP2C could be considered a valuable biomarker for the prevention and treatment for psoriasis.
Collapse
Affiliation(s)
- Huanhuan Zhang
- Department of Dermatology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Cuimin Ren
- Department of Dermatology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiang Liu
- Department of Dermatology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qing Wang
- Department of Dermatology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dahu Wang
- Department of Dermatology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China;
| |
Collapse
|
12
|
Zhou G, Ren X, Tang Z, Li W, Chen W, He Y, Wei B, Zhang H, Ma F, Chen X, Zhang G, Shen M, Liu H. Exploring the association and causal effect between white blood cells and psoriasis using large-scale population data. Front Immunol 2023; 14:1043380. [PMID: 36865550 PMCID: PMC9971993 DOI: 10.3389/fimmu.2023.1043380] [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: 09/13/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction Psoriasis is a chronic inflammatory disease of the skin. A few studies have shown that psoriasis is an immune-mediated disease in which multiple immune cells play crucial roles. However, the association between circulating immune cells and psoriasis remains elusive. Methods To explore the role of circulating immune cells in psoriasis, 361,322 individuals from the UK Biobank (UKB) and 3,971 patients with psoriasis from China were included to investigate the association between white blood cells and psoriasis via an observational study. Genome-wide association studies (GWAS) and Mendelian randomization (MR) were used to evaluate the causal relationship between circulating leukocytes and psoriasis. Results The risk of psoriasis increased with high levels of monocytes, neutrophils, and eosinophils (relative risks and 95% confidence intervals, respectively: 1.430 (1.291-1.584) for monocytes, 1.527 (1.379-1.692) for neutrophils, and 1.417 (1.294-1.551) for eosinophils). Upon further MR analysis, eosinophils showed a definite causal relationship with psoriasis (odds ratio of inverse-variance weighted: 1.386, 95% confidence intervals: 1.092-1.759) and a positive correlation with the psoriasis area and severity index (PASI) score (P = 6.6 × 10-5). The roles of the neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), and lymphocyte-monocyte ratio (LMR) in psoriasis were also assessed. More than 20,000 genetic variations associated with NLR, PLR, and LMR were discovered in a GWAS analysis using the UKB data. Following adjustment for covariates in the observational study, NLR and PLR were shown to be risk factors for psoriasis, whereas LMR was a protective factor. MR results indicated that there was no causal relationship between these three indicators and psoriasis; however, NLR, PLR, and LMR correlated with the PASI score (NLR: rho = 0.244, P = 2.1 × 10-21; PLR: rho = 0.113, P = 1.4 × 10-5; LMR: rho = -0.242, P = 3.5×10-21). Discussion Our findings revealed an important association between circulating leukocytes and psoriasis, which is instructive for the clinical practice of psoriasis treatment.
Collapse
Affiliation(s)
- Guowei Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Xiangmei Ren
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Zhenwei Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Wang Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Wenqiong Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Yi He
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Benliang Wei
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hailun Zhang
- Department of Research and Development, Beijing GAP Biotechnology Co., Ltd, Beijing, China
| | - Fangyu Ma
- Department of Health Management Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guanxiong Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Minxue Shen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|