1
|
Yoshida T, Takashima K, Mtali YS, Miyashita Y, Iwamoto A, Fukushima Y, Nakamura K, Oshiumi H. Regulation of IL-17A-mediated hypersensitivity by extracellular vesicles and lipid nanoparticles carrying miR-451a. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:651-665. [PMID: 40073105 DOI: 10.1093/jimmun/vkae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 12/04/2024] [Indexed: 03/14/2025]
Abstract
Extracellular vesicles (EVs), including exosomes, mediate intercellular communication by transporting functional molecules between donor cells and recipient cells, thereby regulating biological processes, such as immune responses. miR-451a, an immune regulatory microRNA, is highly abundant in circulating EVs; however, its precise physiological significance remains to be fully elucidated. Here, we demonstrate that miR-451a deficiency exacerbates delayed-type hypersensitivity (DTH) in mice. Notably, miR-451a knockout resulted in a significant increase in the number of interleukin (IL)-17A-expressing T helper 17 and γδ T cells infiltrating DTH-induced ear lesions. miR-451a deficiency also increased the number of γδ T cells in the secondary lymphoid tissues. Comprehensive analyses revealed that miR-451 deficiency promoted the expression of Rorc and γδ T cell-related genes following sensitization with allergens. Moreover, intravenous administration of wild-type EVs to miR-451a knockout mice increased cellular miR-451a levels in tissues and significantly attenuated the severity of DTH. Furthermore, synthetic lipid nanoparticles encapsulating miR-451a effectively mitigated DTH. Our findings indicate the importance of circulating miR-451a in the proliferation of γδ T cells and highlight the therapeutic potential of lipid nanoparticle-based microRNA delivery platforms for interventions in immune-related diseases.
Collapse
Affiliation(s)
- Takanobu Yoshida
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ken Takashima
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yohana S Mtali
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yusuke Miyashita
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Asuka Iwamoto
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshimi Fukushima
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
2
|
Zhao D, Ge A, Yan C, Liu X, Yang K, Yan Y, Hao M, Chen J, Daga P, Dai CC, Li C, Cao H. T helper cell 17/regulatory T cell balance regulates ulcerative colitis and the therapeutic role of natural plant components: a review. Front Med (Lausanne) 2025; 11:1502849. [PMID: 40196424 PMCID: PMC11973383 DOI: 10.3389/fmed.2024.1502849] [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: 09/27/2024] [Accepted: 12/23/2024] [Indexed: 04/09/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic relapsing inflammatory disease characterized by progressive mucosal damage. The incidence rate of UC is rising rapidly, which makes the burden of medical resources aggravated. In UC, due to various pathogenic factors such as mucosal immune system disorders, gene mutations and environmental factors disrupting the mucosal barrier function, the midgut pathogenic bacteria and exogenous antigens translocate into the lamina propria, thereby aggravating the inflammatory response and further damages the mucosal barrier. During the progression of UC, Th17 populations that cause inflammation generally increase, while Tregs that suppress Th17 activity decrease. Among them, Th17 mediates immune response, Treg mediates immunosuppression, and the coordinated balance of the two plays a key role in the inflammation and immune process of UC. Natural plant components can regulate biological processes such as immune inflammation from multiple levels of proinflammatory cytokines and signaling pathways. These characteristics have unique advantages and broad prospects in the treatment of UC. In immunomodulation, there is substantial clinical and experimental evidence for the modulatory role of natural plant products in restoring balance between Th17/Treg disturbances in UC. This review summarizes the previous studies on the regulation of Th17/Treg balance in UC by natural plant active ingredients, extracts, and traditional Chinese medicine prescriptions, and provides new evidence for the development and design of lead compounds and natural new drugs for the regulation of Th17/Treg balance in the future, and then provides ideas and evidence for future clinical intervention in the treatment of UC immune disorders and clinical trials.
Collapse
Affiliation(s)
- Da Zhao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Cong Yan
- Department of Urology, The Affiliated Children’s Hospital of Xiangya School of Medicine, Central South University (Hunan Children’s Hospital), Changsha, China
| | - Xingci Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Kailin Yang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Psychology, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China
| | - Yexing Yan
- Department of Psychology, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Department of Psychology, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Junpeng Chen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Center for Cardiometabolic Science, Division of Environmental Medicine, Christina Lee Brown Envirome Insttitute, University of Louisville, Louisville, KY, United States
| | - Pawan Daga
- Department of Internal Medicine, University of Louisville, Louisville, KY, United States
| | - Charles C. Dai
- Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Maryland Baltimore, Baltimore, MD, United States
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, James Clark Hall, College Park, MD, United States
| | - Changping Li
- School of Mechanical Engineering and Automation, Fuyao University of Science and Technology, Fuzhou, China
| | - Hui Cao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
3
|
Wei Y, Chen S, Ling Y, Wang W, Huang Y. Multi-omics revealed that the postbiotic of hawthorn-probiotic alleviated constipation caused by loperamide in elderly mice. Front Nutr 2025; 12:1498004. [PMID: 40070478 PMCID: PMC11895004 DOI: 10.3389/fnut.2025.1498004] [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: 09/18/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
BackgroundConstipation is a prevalent and recurrent gastrointestinal disorder causing significant discomfort. However, current treatments often prove ineffective. Previous research indicates that the postbiotic derived from a combination of hawthorn and probiotics can alleviate constipation. This study aimed to investigate its mechanisms using loperamide-induced constipation in aged KM mice.MethodsConstipated mice were divided into groups receiving 10% lactulose (Y), hawthorn extract (S), probiotics (F), and the postbiotic of hawthorn-probiotic (FS). UPLC–MS metabolomics identified constituents of F, S, and FS. Network pharmacological analysis identified targets affected by FS. RT-qPCR assessed target expression in mouse colons, along with IL-6 and IL-17A levels. Molecular docking with AutoDock Tools1.5.6 evaluated interactions between FS components and targets. ex vivo colonic organ culture and RT-qPCR assessed target changes. Molecular dynamics analysis further scrutinized interactions. Targeted metabolomics measured short-chain fatty acid levels in mouse stool.ResultsUPLC–MS metabolomics revealed distinct profiles for F, S, and FS, with FS showing decreased toxic substances and increased beneficial ones compared to S. Network pharmacology identified 20 cross-targets of FS in constipation. RT-qPCR showed decreased NR1I2 and SULT1A1 and increased GLP-2r in FS-treated mice. Inflammatory cytokines IL-6 and IL-17A were also reduced. ex vivo colonic organ culture and molecular docking identified effective combinations such as TNF-Baicalin and AQP3-Quinacridone. RMSD, RMSF, and RG analyses indicated favorable interactions between small molecules and targets. Targeted metabolomics revealed differing short-chain fatty acid contents in feces among groups.ConclusionThe postbiotic of hawthorn-probiotic alleviates constipation by regulating intestinal water and sodium metabolism, maintaining the intestinal barrier and gut flora, promoting epithelial cell proliferation, reducing inflammatory responses, and improving short-chain fatty acid metabolism.
Collapse
Affiliation(s)
- Yu Wei
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Basic Medical Science College, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuai Chen
- Yunnan University of Chinese Medicine, Kunming, China
| | - Ying Ling
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yali Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Basic Medical Science College, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
4
|
Singh Gautam A, Panda ES, Balki S, Pandey SK, Tiwari A, Singh RK. Therapeutic potential of chrysin in regulation of interleukin-17 signaling in a repeated intranasal amyloid-beta-induced Alzheimer's disease model. Food Funct 2025; 16:731-749. [PMID: 39748776 DOI: 10.1039/d4fo05278a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Objective: The aim of the current study was to study the therapeutic potential of chrysin against repeated intranasal amyloid-beta (Aβ)-induced interleukin-17 (IL-17) signaling in a mouse model of AD. Methods: Male BALB/c mice were daily exposed to intranasal Aβ1-42 (10 μg/10 μL) for seven consecutive days. Chrysin was orally administered at doses of 25, 50 and 100 mg kg-1 in 0.5% sodium carboxy methyl cellulose suspension from day 5 of Aβ1-42 administration for seven days. Following the treatment, the memory of the animals was appraised using Morris water maze, novel object recognition and passive avoidance tests. Further, the effects of chrysin on Aβ1-42-induced IL-17 signaling and redox levels were evaluated in the cortex and hippocampus regions of the mouse brain through western blot and immunohistochemistry. Results: The exposure to Aβ1-42 through the intranasal route induced a significant decline in the spatial, learning and cognitive memory of the animals, and most interestingly, exposure to Aβ1-42 triggered IL-17-mediated signaling, which resulted in a significant increase in the expression of IL-17RA, Act1 and TRAF6. Furthermore, Aβ1-42 impaired the tissue redox level and inflammatory cytokines in the mouse brain. Alternatively, treatment with chrysin at 25, 50 and 100 mg kg-1 oral doses alleviated Aβ1-42-mediated memory decline, impaired redox levels and inflammation. Specifically, chrysin downregulated the expression of IL-17 and mediated signaling in the brain regions of the mice. Conclusion: Chrysin was evidenced to be a potent antioxidant and anti-inflammatory agent, clearly showing a protective role against Aβ1-42-induced IL-17-mediated inflammation in the brain of the mice.
Collapse
Affiliation(s)
- Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli. Transit campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| | - Ekta Swarnamayee Panda
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli. Transit campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| | - Sneha Balki
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli. Transit campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| | - Shivam Kumar Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli. Transit campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| | - Aman Tiwari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli. Transit campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli. Transit campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| |
Collapse
|
5
|
Yan L, Wang W, Dong M, Wang R, Li C. Skin Metabolic Signatures of Psoriasis and Psoriasis Concurrent with Metabolic Syndrome. J Inflamm Res 2025; 18:505-517. [PMID: 39810975 PMCID: PMC11730757 DOI: 10.2147/jir.s493338] [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: 08/29/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025] Open
Abstract
Purpose Psoriasis is a complex inflammatory skin disorder that is closely associated with metabolic syndrome (MetS). Limited information is available on skin metabolic changes in psoriasis; the effect of concurrent MetS on psoriatic skin metabolite levels is unknown. We aimed to expand this information through skin metabolomic analysis. Patients and Methods Untargeted metabolomics was conducted using skin samples from 38 patients with psoriasis vulgaris with MetS (PVMS), 23 patients with psoriasis vulgaris without MetS (PVNMS), and 10 healthy controls (HC). Data analyses, including multivariate statistical analysis, KEGG pathway enrichment analysis, correlation analysis, and receiver operating characteristic curve analysis, were performed. Results Significant discrepancies were found between skin metabolites in the HC and PVNMS groups, particularly those involved in nucleotide and glycerophospholipid metabolism. Fifteen of these metabolites were positively correlated with psoriasis severity. Furthermore, MetS was found to affect the metabolic profiles of patients with psoriasis. There were some metabolites with consistent alterations in both the PVNMS/HC and PVMS/PVNMS comparisons. Conclusion This study may provide new insights into the link between skin metabolism and psoriatic inflammation and the mechanism underlying the interaction between psoriasis and MetS.
Collapse
Affiliation(s)
- Liang Yan
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
- Department of Dermatology, General Hospital of Central Theater Command of PLA, Wuhan, 430070, People’s Republic of China
| | - Wenqiu Wang
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Meihan Dong
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Rui Wang
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Chengxin Li
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, 100853, People’s Republic of China
| |
Collapse
|
6
|
De Santis M, Tonutti A, Isailovic N, Motta F, Rivara RM, Ragusa R, Guidelli GM, Caprioli M, Ceribelli A, Renna D, Luciano N, Selmi C. Serum IL-23 levels reflect a myeloid inflammatory signature and predict the response to apremilast in patients with psoriatic arthritis. Front Immunol 2024; 15:1455134. [PMID: 39697337 PMCID: PMC11652657 DOI: 10.3389/fimmu.2024.1455134] [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: 06/26/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Background The phosphodiesterase 4 (PDE4) inhibitor apremilast downregulates the production of IL-23 and other pro-inflammatory cytokines involved in the pathogenesis of psoriatic arthritis (PsA). Aim To investigate the effects of apremilast on the production of cytokines by peripheral blood monocyte-derived macrophages, innate-like lymphocyte cells (ILCs), mucosal-associated invariant T (MAIT) cells, γδ T cells, natural killer (NK) cells, and NKT-like cells from patients with PsA manifesting different clinical responses to the treatment. Methods Peripheral blood samples were obtained from patients with PsA at baseline and after 1 and 4 months of apremilast therapy (n = 23) and 20 controls with osteoarthritis. Cytokine expression in peripheral blood monocyte-derived macrophages and ILCs/MAIT/γδT/NK/NKT-like cells was tested by RT-PCR and FACS analyses, respectively; cytokine levels in culture supernatants and sera were analyzed by ELISA. Results PsA monocyte-derived macrophages exhibited higher expressions of IL-23, IL-1β, and TNF-α, compared with OA controls, more profoundly in patients responding to apremilast. There were 17/23 (74%) PsA patients who were classified as responders to apremilast at 4 months, and a baseline serum IL-23 >1.4 pg/mL was associated with the responder status (AUCROC 0.79; sensitivity 100%, specificity 68%). Of note, apremilast led to a significantly reduced expression of IL-23 in peripheral blood monocyte-derived macrophages; IL-17 in ILC1 and in T cells of responder patients; IFN-γ in γδ T lymphocytes. Conclusion An enhanced myeloid inflammatory signature characterizes PsA monocyte-derived macrophages, and serum IL-23 levels represent candidate biomarkers for PsA response to apremilast.
Collapse
Affiliation(s)
- Maria De Santis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Antonio Tonutti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Natasa Isailovic
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Francesca Motta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Radu Marian Rivara
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Rita Ragusa
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Giacomo M. Guidelli
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Marta Caprioli
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Angela Ceribelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Daniela Renna
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Nicoletta Luciano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
7
|
Wang C, Yang P, Wang J, Liu L, Chen J, Cai X, Zhang M, Lin N, Wang S, Yu Y, Li L, Li X. Evidence and potential mechanism of action of indigo naturalis and its active components in the treatment of psoriasis. Ann Med 2024; 56:2329261. [PMID: 39316667 PMCID: PMC11423532 DOI: 10.1080/07853890.2024.2329261] [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: 12/12/2022] [Revised: 12/11/2023] [Accepted: 03/05/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Indigo naturalis is effective against psoriasis. Indigo, indirubin and tryptanthrin, the main active components of indigo naturalis, have anti-inflammatory properties. OBJECTIVE To evaluate the efficacy and safety of indigo naturalis and its active components in the treatment of psoriasis. METHODS Seven databases were searched for studies of indigo naturalis and its active components for the treatment of psoriasis. RESULTS The findings demonstrated a higher response rate in the Chinese herbal medicine (CHM) formula groups than in the control group for Psoriasis Area and Severity Index 60 (PASI60) (Rate difference [RD] = 0.22, p < .0001). Among all adverse events, only the incidence of gastrointestinal adverse reactions was higher in the CHM formula group than in the control group (RD = 0.09, p < .0001). In preclinical in vivo studies, indirubin showed better performance in improving the phenotype of psoriasis-like mice compared to that in controls, including the PASI score (mean difference [MD] = -3.58, p < .0001), epidermal thickness (MD = -29.13, p < .0001), interleukin-(IL) 17 A mRNA expression (MD = -2.27, p = .0066) and IL-23 mRNA (MD = -5.36, p = .01). CONCLUSION Indigo naturalis combined with conventional treatments is useful for treating psoriasis. Indigo naturalis display anti-proliferative, anti-inflammatory and anti-angiogenic effects by regulating the TAK1, JAK3/STAT3, Wnt/β-catenin, Akt/PKB, FAK and AP-1/c-Jun pathway.
Collapse
Affiliation(s)
- Chunxiao Wang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Panpan Yang
- Department of Dermatology, Shijiazhuang TCM Hospital, Shijiazhuang, China
| | - Jiao Wang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Liu Liu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Jiale Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoce Cai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Miao Zhang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Naixuan Lin
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sihan Wang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanting Yu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linge Li
- Department of Dermatology, Shijiazhuang TCM Hospital, Shijiazhuang, China
| | - Xin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
8
|
Xu Y, Zhang X, Liu S, Qu N, Gao Y, Lu C, Zhai J, Zhu J. The role of Interleukin-38 in modulating T cells in chronic Colitis: A mouse model study. Cytokine 2024; 184:156769. [PMID: 39342821 DOI: 10.1016/j.cyto.2024.156769] [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: 06/09/2024] [Revised: 09/11/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Interleukin (IL)-38 belongs to the IL-36 subfamily within the IL-1 family. Patients with inflammatory bowel diseases (IBD) exhibit higher levels of IL-38 in their intestinal tissue compared to healthy controls, suggesting that IL-38 may play a role in the pathogenesis of IBD. However, IL-38's impact on T cell-mediated immune responses in gastrointestinal inflammation has not been investigated. Therefore, the objective of this work was to elucidate the role of IL-38 in modulating T cells in a mouse model of dextran sulfate sodium (DSS)-induced chronic colitis. METHODS Recombinant IL-38 (rIL-38) was administered intraperitoneally (i.p.) to mice with chronic colitis induced by DSS. Clinical symptoms, length of colon, and histologic alterations were assessed. Cytokine production was quantified using ELISA, and helper T (Th) cell subsets were evaluated via flow cytometry. RESULTS Administration of recombinant IL-38 (rIL-38) alleviated DSS-induced chronic colitis. In addition, animals with chronic colitis treated with rIL-38 exhibited a significant decrease in the spontaneous production of inflammatory cytokines by neutrophils in the lamina propria. Furthermore, rIL-38 treatment increased the absolute numbers and percentages of regulatory T cells (Tregs) but decreased the absolute numbers and percentages of Th1 and Th17 cells. Moreover, rIL-38 treatment also decreased IL-17A-producing γδT cells substantially. CONCLUSION This study's results show that IL-38 reduces the effects of chronic colitis caused by DSS by boosting Treg reactions and reducing Th1/Th17 reactions and IL-17A-producing γδT cells in LPL. Therefore, we propose that IL-38 has the potential to be utilized as a biological therapy agent for IBD.
Collapse
Affiliation(s)
- Ying Xu
- Office of Drug Clinical Trials, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Xuan Zhang
- Department of Clinical Laboratory, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Shanshan Liu
- Department of Clinical Laboratory, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Nanfang Qu
- Department of Gastroenterology, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Yi Gao
- General Surgery I Department, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Changlong Lu
- Institute of Immunology, China Medical University, Shenyang 110122, China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu Universiry, Tongliao 028000, China
| | - Junfeng Zhu
- Department of Clinical Laboratory, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China.
| |
Collapse
|
9
|
Su QY, Gao HY, Duan YR, Luo J, Wang WZ, Qiao XC, Zhang SX. The immunologic role of IL-23 in psoriatic arthritis: a potential therapeutic target. Expert Opin Biol Ther 2024; 24:1119-1132. [PMID: 39230202 DOI: 10.1080/14712598.2024.2401148] [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: 03/10/2024] [Revised: 07/25/2024] [Accepted: 09/02/2024] [Indexed: 09/05/2024]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a debilitating chronic condition characterized by inflammation of the joints, bones, enthesis, and skin. The pivotal role of interleukin-23 (IL-23) in the pathogenesis of PsA has become increasingly evident. This proinflammatory cytokine is markedly elevated in patients with PsA, suggesting its potential as a therapeutic target. Consequently, IL-23 inhibitors have emerged as promising first-line biologic treatments for PsA. AREAS COVERED This review delves into the immunopathogenic mechanisms of IL-23 at the cellular and molecular levels in PsA. Furthermore, it provides the recent efficacy and safety profiles of IL-23 inhibitors. We conducted a literature search in PubMed for the following terms: 'IL-23 and psoriatic arthritis,' 'Ustekinumab,' 'Guselkumab,' 'Risankizumab,' and 'Tildrakizumab.' In addition, we retrieved clinical trials involving IL-23 inhibitors registered in ClinicalTrials.gov, EudraCT, and ICTRP. EXPERT OPINION Despite the promising outcomes observed with IL-23 inhibitors, several challenges persist. The long-term effects of these agents require further investigation through prospective studies, and their limited accessibility worldwide necessitates urgent attention. Additionally, ongoing research is warranted to explore other potential drug targets within the IL-23/IL-23 R axis. The development of reliable biomarkers could greatly enhance early detection, tailored management strategies, and personalized treatment approaches for patients with PsA.
Collapse
Affiliation(s)
- Qin-Yi Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Heng-Yan Gao
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Yue-Ru Duan
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Jing Luo
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Wei-Ze Wang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Xi-Chao Qiao
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi medical university, Taiyuan, China
| |
Collapse
|
10
|
Meyer A. Illuminating the impact of γδ T cells in man and mice in spondylarthritides. Eur J Immunol 2024; 54:e2451071. [PMID: 39077953 DOI: 10.1002/eji.202451071] [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: 03/28/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024]
Abstract
Spondylarthritides (SpA) are a group of autoinflammatory diseases affecting the spine, peripheral joints, and entheses, including axial spondyloarthritis (axSpA) and psoriatic arthritis. AxSpA has a multifactorial etiology that involves genetic predispositions, such as HLA-B27 and IL-23R. Although HLA-B27 is strongly associated with axSpA, its role remains unclear. GWAS studies have demonstrated that genetic polymorphisms related to the IL-23 pathway occur throughout the spectrum of SpA, including but not limited to axSpA and PsA. IL-23 promotes the production of IL-17, which drives inflammation and tissue damage. This pathway contributes not only to peripheral enthesitis but also to spinal inflammation. γδ T cells in axSpA express IL-23R and RORγt, crucial for their activation, although specific pathogenic cells and factors remain elusive. Despite drug efficacy in PsA, IL-23R inhibition is ineffective in axSpA. Murine models provide valuable insights into the intricate cellular and molecular interactions that contribute to the development and progression of SpA. Those models are useful tools to elucidate the dynamics of γδ T cell involvement, offering insights into disease mechanisms and potential therapeutic targets. This review aims to illuminate the complex interplay between IL-23 and γδ T cells in SpA pathogenesis, emphasizing their roles in chronic inflammation, tissue damage, and disease heterogeneity.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Disease Models, Animal
- Interleukin-23/immunology
- Interleukin-23/metabolism
- Interleukin-23/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- HLA-B27 Antigen/genetics
- HLA-B27 Antigen/immunology
- Genetic Predisposition to Disease
- Spondylarthritis/immunology
- Receptors, Interleukin/genetics
- Receptors, Interleukin/metabolism
- Receptors, Interleukin/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
Collapse
Affiliation(s)
- Anja Meyer
- Center for Molecular Neurobiology Hamburg, Institute for Systems Immunology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
11
|
Chen Y, Liang CL, Liu H, Chen H, He Y, Lin J, He Z, Qiu F, Yang B, Lu C, Dai Z. Percutaneous Delivery of Hederacoside C-Loaded Nanoliposome Gel Alleviates Psoriasiform Skin Inflammation through the CCL17/Treg Axis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:48969-48981. [PMID: 39233638 DOI: 10.1021/acsami.4c06720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Psoriasis is a chronic, recurrent, and inflammatory skin disease. Topical agents, which can avoid the adverse effects of systemic treatment, are the first-choice therapy for patients with mild-to-moderate psoriasis. Hederacoside C (HSC) with anti-inflammatory properties has been used to treat some inflammatory diseases. We speculated that HSC might also be effective for psoriasis treatment. However, topical application of HSC for psoriasis treatment is challenging because of its low water solubility and poor skin permeability. Therefore, it is important to effectively deliver HSC percutaneously using certain biomaterials. Here we constructed a hydroxypropyl-β-cyclodextrin-coated liposome gel formulation for the loading and percutaneously delivering of HSC, referred to as HSC-Lipo@gel. The characterization, stability, release properties, and mechanical or transdermal features of the HSC-Lipo@gel were evaluated. Its therapeutic potential was also demonstrated using mouse models of IMQ-induced psoriasis. We found that HSC-Lipo@gel effectively improved the skin permeability of HSC with the property of good stability and sustained release. Importantly, HSC-Lipo@gel showed higher efficacy than HSC@gel without liposomes in alleviating psoriatic skin lesions. It attenuated epidermal hyperplasia and suppressed expression of IL-17A, TNF-α, IL-6, and IL-23 in lesional skin. Interestingly, HSC-Lipo@gel reduced the expression of CC chemokine ligand 17 (CCL17), but not CCL22, in the skin. Especially, HSC-Lipo@gel inhibited CCL17 expression by skin dendritic cells while increasing regulatory T cells (Tregs) in both skin and draining lymph nodes of psoriatic mice. Administration of CCL17 resulted in severe skin lesions and reduced CD4+FoxP3+ Tregs in psoriatic mice previously treated with HSC-Lipo@gel. Finally, HSC or HSC-Lipo also suppressed the CCL17 production by dendritic cells in vitro. Therefore, HSC-Lipo@gel alleviated psoriasiform skin inflammation by increasing cutaneous Tregs via downregulation of the expression of CCL17, but not CCL22. Thus, HSC-Lipo@gel may be a stable, highly permeable, and effective system for topical treatment of psoriasis.
Collapse
Affiliation(s)
- Yuchao Chen
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, P. R. China
| | - Chun-Ling Liang
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, P. R. China
| | - Huazhen Liu
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, P. R. China
| | - Haiming Chen
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, P. R. China
| | - Yuming He
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
| | - Jingru Lin
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
| | - Zenghua He
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
| | - Feifei Qiu
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, P. R. China
| | - Bin Yang
- Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, Leicester LE1 9HN, U.K
| | - Chuanjian Lu
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, P. R. China
| | - Zhenhua Dai
- Joint Immunology Program, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P. R. China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, P. R. China
| |
Collapse
|
12
|
Dimitriou F, Cheng PF, Saltari A, Schaper-Gerhardt K, Staeger R, Haunerdinger V, Sella F, Tastanova A, Urban C, Dettwiler S, Mihic-Probst D, Matter CM, Michielin O, Gutzmer R, Long GV, Becher B, Levesque MP, Dummer R. A targetable type III immune response with increase of IL-17A expressing CD4 + T cells is associated with immunotherapy-induced toxicity in melanoma. NATURE CANCER 2024; 5:1390-1408. [PMID: 39210005 PMCID: PMC11424476 DOI: 10.1038/s43018-024-00810-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
Immune checkpoint inhibitors are standard-of-care for the treatment of advanced melanoma, but their use is limited by immune-related adverse events. Proteomic analyses and multiplex cytokine and chemokine assays from serum at baseline and at the adverse event onset indicated aberrant T cell activity with differential expression of type I and III immune signatures. This was in line with the finding of an increase in the proportion of CD4+ T cells with IL-17A expression at the adverse event onset in the peripheral blood using flow cytometry. Multiplex immunohistochemistry and spatial transcriptomics on immunotherapy-induced skin rash and colitis showed an increase in the proportion of CD4+ T cells with IL-17A expression. Anti-IL-17A was administered in two patients with mild myocarditis, colitis and skin rash with resolution of the adverse events. This study highlights the potential role of type III CD4+ T cells in adverse event development and provides proof-of-principle evidence for a clinical trial using anti-IL-17A for treating adverse events.
Collapse
Affiliation(s)
- Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.
- Faculty of Medicine, University of Zurich, Zurich, Switzerland.
| | - Phil F Cheng
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Annalisa Saltari
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Katrin Schaper-Gerhardt
- Department of Dermatology, Johannes Wesling Medical Center, Ruhr University Bochum Campus Minden, Minden, Germany
- Department of Dermatology, Medical School Hannover, Hannover, Germany
| | - Ramon Staeger
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Veronika Haunerdinger
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Federica Sella
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Aizhan Tastanova
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Christian Urban
- Functional Genomics Center Zurich, University of Zurich/ETH Zurich, Zurich, Switzerland
| | - Susanne Dettwiler
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Daniela Mihic-Probst
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Christian M Matter
- Department of Cardiology, University Heart Center and Center for Experimental Cardiology (CTEC), University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Olivier Michielin
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Ralf Gutzmer
- Department of Dermatology, Johannes Wesling Medical Center, Ruhr University Bochum Campus Minden, Minden, Germany
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich (UZH), Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.
- Faculty of Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Zhang Y, Liu K, Guo M, Yang Y, Zhang H. Negative regulator IL-1 receptor 2 (IL-1R2) and its roles in immune regulation of autoimmune diseases. Int Immunopharmacol 2024; 136:112400. [PMID: 38850793 DOI: 10.1016/j.intimp.2024.112400] [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: 04/09/2024] [Revised: 05/22/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
The decoy receptor interleukin 1 receptor 2 (IL-1R2), also known as CD121b, has different forms: membrane-bound (mIL-1R2), soluble secreted (ssIL-1R2), shedded (shIL-1R2), intracellular domain (IL-1R2ICD). The different forms of IL-1R2 exert not exactly similar functions. IL-1R2 can not only participate in the regulation of inflammatory response by competing with IL-1R1 to bind IL-1 and IL-1RAP, but also regulate IL-1 maturation and cell activation, promote cell survival, participate in IL-1-dependent internalization, and even have biological activity as a transcriptional cofactor. In this review, we provide a detailed description of the biological characteristics of IL-1R2 and discuss the expression and unique role of IL-1R2 in different immune cells. Importantly, we summarize the role of IL-1R2 in immune regulation from different autoimmune diseases, hoping to provide a new direction for in-depth studies of pathogenesis and therapeutic targets in autoimmune diseases.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Muyao Guo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha City, Hunan Province, China
| | - Yiying Yang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China; Postdoctoral Research Station of Biology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China.
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| |
Collapse
|
14
|
Chojnacka-Purpurowicz J, Owczarczyk-Saczonek A, Nedoszytko B. The Role of Gamma Delta T Lymphocytes in Physiological and Pathological Condition-Focus on Psoriasis, Atopic Dermatitis, Autoimmune Disorders, Cancer and Lymphomas. Int J Mol Sci 2024; 25:7960. [PMID: 39063202 PMCID: PMC11277122 DOI: 10.3390/ijms25147960] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Gamma delta (γδ) T cells are a heterogeneous population of cells that play roles in inflammation, host tissue repair, clearance of viral and bacterial pathogens, regulation of immune processes, and tumor surveillance. Recent research suggests that these are the main skin cells that produce interleukin-17 (I-17). Furthermore, γδ T cells exhibit memory-cell-like characteristics that mediate repeated episodes of psoriatic inflammation. γδ T cells are found in epithelial tissues, where many cancers develop. There, they participate in antitumor immunity as cytotoxic cells or as immune coordinators. γδ T cells also participate in host defense, immune surveillance, and immune homeostasis. The aim of this review is to present the importance of γδ T cells in physiological and pathological diseases, such as psoriasis, atopic dermatitis, autoimmune diseases, cancer, and lymphoma.
Collapse
Affiliation(s)
- Joanna Chojnacka-Purpurowicz
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, The University of Warmia and Mazury, 10-719 Olsztyn, Poland;
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, The University of Warmia and Mazury, 10-719 Olsztyn, Poland;
| | - Bogusław Nedoszytko
- Department of Medical Laboratory Diagnostics–Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 3A M. Skłodowskiej-Curie Street, 80-210 Gdansk, Poland;
- Molecular Laboratory, Invicta Fertility and Reproductive Center, 81-740 Sopot, Poland
| |
Collapse
|
15
|
He F, Yu J, Ma S, Zhao W, Zhang M, Wang J, Zhang C, Wu J, Zhu L. γδT Cells Induce the Inflammatory Response of Human Fibroblast-Like Synoviocytes Directly or by Stimulating B Cells to Activate IL-17/STAT3 Signaling Pathway. Int Arch Allergy Immunol 2024; 185:1154-1165. [PMID: 38991517 DOI: 10.1159/000539703] [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: 01/16/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) combined with hashimoto thyroiditis (HT) is an important cause of various fatal comorbidities of RA. There is no precise conclusion about the cause of this disease. METHODS Peripheral blood and synovial tissue were collected from healthy participants, patients with RA, and patients with both RA and HT. Immunofluorescence staining and Pearson correlation analysis were used to detect the levels of γδTCR and the correlation between IL-17 and p-STAT3, respectively. ELISA, chemiluminescence assays, qRT-PCR and Western blot were performed to detect the levels of IgG, IgM, IFN-γ, IL-1β, TNF-α, Tg-Ab, Tpo-Ab, IL-17, IL-2, p-SATA3, and STAT3, respectively. RESULTS There was increased proportion of γδT cells, IL-17, and p-STAT3 levels in RA and HT patients. IL-17 was positively correlated with p-STAT3. γδT cells significantly promoted the expression of IgG, Tg-Ab, Tpo-Ab, and IL-17. When γδT and human fibroblast-like synoviocytes (FLSs) were co-cultured, the levels of IL-2, IFN-γ, IL-1β, TNF-α, and IL-17 were increased, and the IL-17/STAT3 signaling pathway was activated. When IL-17-silenced γδT cells and STAT3-silenced FLSs were co-cultured, the levels of IL-1β and TNF-α in FLSs were significantly decreased. Furthermore, when STAT3-silenced FLSs were added to the co-culture medium of B cells and γδT cells, the levels of IL-1β and TNF-α were also decreased significantly. CONCLUSION γδT cells induced RA directly or by stimulating B cells to activate STAT3 through IL-17.
Collapse
Affiliation(s)
- Fang He
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Juan Yu
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Sha Ma
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Weiqing Zhao
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Mingxing Zhang
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Juan Wang
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Chunpan Zhang
- Department of Pain, The First People's Hospital of Yunnan Province, Kunming, China
| | - Jiangping Wu
- Department of Pain, The First People's Hospital of Yunnan Province, Kunming, China
| | - Lixuan Zhu
- Department of Pain, The First People's Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
16
|
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
|
17
|
Zhang Y, Lu Q. Immune cells in skin inflammation, wound healing, and skin cancer. J Leukoc Biol 2024; 115:852-865. [PMID: 37718697 DOI: 10.1093/jleuko/qiad107] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/22/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
Given the self-evident importance of cutaneous immunity in the maintenance of body-surface homeostasis, disturbance of the steady-state skin is inextricably intertwined with dysfunction in cutaneous immunity. It is often overlooked by people that skin, well-known as a solid physical barrier, is also a strong immunological barrier, considering the abundant presence of immune cells including lymphocytes, granulocytes, dendritic cells, and macrophages. What's more, humoral immune components including cytokines, immunoglobulins, and antimicrobial peptides are also rich in the skin. This review centers on skin inflammation (acute and chronic, infection and aseptic inflammation), wound healing, and skin cancer to elucidate the elaborate network of immune cells in skin diseases.
Collapse
Affiliation(s)
- Yuhan Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| |
Collapse
|
18
|
Pinto-Tasende JA, Fernandez-Moreno M, Rego Perez I, Fernandez-Lopez JC, Oreiro-Villar N, De Toro Santos FJ, Blanco-García FJ. Higher Synovial Immunohistochemistry Reactivity of IL-17A, Dkk1, and TGF-β1 in Patients with Early Psoriatic Arthritis and Rheumatoid Arthritis Could Predict the Use of Biologics. Biomedicines 2024; 12:815. [PMID: 38672170 PMCID: PMC11048598 DOI: 10.3390/biomedicines12040815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Delay in diagnosis and therapy in patients with arthritis commonly leads to progressive articular damage. The study aimed to investigate the immunohistochemical reactivity of synovial cytokines associated with inflammation and the bone erosives/neoformatives processes among individuals diagnosed with psoriatic arthritis (PsA), rheumatoid arthritis (RA), osteoarthritis (OA), and radiographic axial spondyloarthritis (r-axSpA), with the intention of identifying potential biomarkers. METHODS Specimens were collected from the inflamed knee joints of patients referred for arthroscopic procedures, and the synovial tissue (ST) was prepared for quantifying protein expression through immunohistochemical analysis (% expressed in Ratio_Area-Intensity) for TGF-β1, IL-17A, Dkk1, BMP2, BMP4, and Wnt5b. The collected data underwent thorough analysis and examination of their predictive capabilities utilising receiver operating characteristic (ROC) curves. RESULTS Valid synovial tissue samples were acquired from 40 patients for IHC quantification analysis. Initially, these patients had not undergone treatment with biologics. However, after 5 years, 4 out of 13 patients diagnosed with PsA and two out of nine patients diagnosed with RA had commenced biologic treatments. Individuals with early PsA who received subsequent biologic treatment exhibited significantly elevated IHC reactivity in ST for TGF-β1 (p = 0.015). Additionally, patients with both PsA and RA who underwent biologic therapy displayed increased IHC reactivity for IL-17A (p = 0.016), TGF-β1 (p = 0.009), and Dkk1 (p = 0.042). ROC curve analysis of IHC reactivity for TGF-β1, Dkk1, and IL-17A in the synovial seems to predict future treatment with biologics in the next 5 years with the area under the curve (AUC) of a combined sum of the three values: AUC: 0.828 (95% CI: 0.689-0.968; p 0.005) S 75% E 84.4%. CONCLUSIONS Higher synovial immunohistochemistry reactivity of IL-17A, Dkk1, and TGF-β1 in patients with early psoriatic arthritis and rheumatoid arthritis may serve as potential indicators for predicting the necessity of utilising biologic treatments.
Collapse
Affiliation(s)
- Jose A. Pinto-Tasende
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| | - Mercedes Fernandez-Moreno
- Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, 15006 A Coruña, Spain; (M.F.-M.); (I.R.P.)
| | - Ignacio Rego Perez
- Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, 15006 A Coruña, Spain; (M.F.-M.); (I.R.P.)
| | - J. Carlos Fernandez-Lopez
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| | - Natividad Oreiro-Villar
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| | - F. Javier De Toro Santos
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| | - Francisco J. Blanco-García
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| |
Collapse
|
19
|
Sage SE, Leeb T, Jagannathan V, Gerber V. Single-cell profiling of bronchoalveolar cells reveals a Th17 signature in neutrophilic severe equine asthma. Immunology 2024; 171:549-565. [PMID: 38153159 DOI: 10.1111/imm.13745] [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: 08/03/2023] [Accepted: 12/10/2023] [Indexed: 12/29/2023] Open
Abstract
Severe equine asthma (SEA) is a complex respiratory condition characterized by chronic airway inflammation. It shares many clinical and pathological features with human neutrophilic asthma, making it a valuable model for studying this condition. However, the immune mechanisms driving SEA have remained elusive. Although SEA has been primarily associated with a Th2 response, there have also been reports of Th1, Th17, or mixed-mediated responses. To uncover the elusive immune mechanisms driving SEA, we performed single-cell mRNA sequencing (scRNA-seq) on cryopreserved bronchoalveolar cells from 11 Warmblood horses, 5 controls and 6 with SEA. We identified six major cell types, including B cells, T cells, monocytes-macrophages, dendritic cells, neutrophils, and mast cells. All cell types exhibited significant heterogeneity, with previously identified and novel cell subtypes. Notably, we observed monocyte-lymphocyte complexes and detected a robust Th17 signature in SEA, with CXCL13 upregulation in intermediate monocytes. Asthmatic horses exhibited expansion of the B-cell population, Th17 polarization of the T-cell populations, and dysregulation of genes associated with T-cell function. Neutrophils demonstrated enhanced migratory capacity and heightened aptitude for neutrophil extracellular trap formation. These findings provide compelling evidence for a predominant Th17 immune response in neutrophilic SEA, driven by dysregulation of monocyte and T-cell genes. The dysregulated genes identified through scRNA-seq have potential as biomarkers and therapeutic targets for SEA and provide insights into human neutrophilic asthma.
Collapse
Affiliation(s)
- Sophie E Sage
- Department of Clinical Veterinary Medicine, Vetsuisse Faculty, Swiss Institute of Equine Medicine, University of Bern, Bern, Switzerland
| | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, Institute of Genetics, University of Bern, Bern, Switzerland
| | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, Institute of Genetics, University of Bern, Bern, Switzerland
| | - Vinzenz Gerber
- Department of Clinical Veterinary Medicine, Vetsuisse Faculty, Swiss Institute of Equine Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
Gao X, Lin X, Wang Q, Chen J. Artemisinins: Promising drug candidates for the treatment of autoimmune diseases. Med Res Rev 2024; 44:867-891. [PMID: 38054758 DOI: 10.1002/med.22001] [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: 09/21/2023] [Revised: 11/02/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Autoimmune diseases are characterized by the immune system's attack on one's own tissues which are highly diverse and diseases differ in severity, causing damage in virtually all human systems including connective tissue (e.g., rheumatoid arthritis), neurological system (e.g., multiple sclerosis) and digestive system (e.g., inflammatory bowel disease). Historically, treatments normally include pain-killing medication, anti-inflammatory drugs, corticosteroids, and immunosuppressant drugs. However, given the above characteristics, treatment of autoimmune diseases has always been a challenge. Artemisinin is a natural sesquiterpene lactone initially extracted and separated from Chinese medicine Artemisia annua L., which has a long history of curing malaria. Artemisinin's derivatives such as artesunate, dihydroartemisinin, artemether, artemisitene, and so forth, are a family of artemisinins with antimalarial activity. Over the past decades, accumulating evidence have indicated the promising therapeutic potential of artemisinins in autoimmune diseases. Herein, we systematically summarized the research regarding the immunoregulatory properties of artemisinins including artemisinin and its derivatives, discussing their potential therapeutic viability toward major autoimmune diseases and the underlying mechanisms. This review will provide new directions for basic research and clinical translational medicine of artemisinins.
Collapse
Affiliation(s)
- Xu Gao
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Xian Lin
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Jian Chen
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| |
Collapse
|
21
|
Wang S, Kozai M, Hiraishi M, Rubel MZU, Ichii O, Inaba M, Matsuo K, Takada K. Roles of tumor necrosis factor-like ligand 1A in γδT-cell activation and psoriasis pathogenesis. Front Immunol 2024; 15:1340467. [PMID: 38348035 PMCID: PMC10859483 DOI: 10.3389/fimmu.2024.1340467] [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: 11/18/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024] Open
Abstract
Background Interleukin (IL)-17-producing γδT (γδT17) cells mediate inflammatory responses in barrier tissues. Dysregulated γδT17 cell activation can lead to the overproduction of IL-17 and IL-22 and the development of inflammatory diseases, including psoriasis. IL-23 and IL-1β are known to synergistically activate γδT17 cells, but the regulatory mechanisms of γδT17 cells have not been fully elucidated. This study aimed to reveal the contribution of the inflammatory cytokine tumor necrosis factor-like ligand 1A (TL1A) to γδT17 cell activation and psoriasis development. Methods Anti-TL1A antibody was injected into an imiquimod (IMQ)-induced murine psoriasis model. TL1A receptor expression was analyzed in splenic and dermal γδT cells. γδT cells were tested for cytokine production in vitro and in vivo under stimulation with IL-23, IL-1β, and TL1A. TL1A was applied to a psoriasis model induced by intradermal IL-23 injection. Mice deficient in γδT cells were intradermally injected with IL-23 plus TL1A to verify the contribution of TL1A-dependent γδT-cell activation to psoriasis development. Results Neutralization of TL1A attenuated γδT17 cell activation in IMQ-treated skin. TL1A induced cytokine production by splenic γδT17 cells in synergy with IL-23. Dermal γδT17 cells constitutively expressed a TL1A receptor at high levels and vigorously produced IL-22 upon intradermal IL-23 and TL1A injection but not IL-23 alone. TL1A exacerbated the dermal symptoms induced by IL-23 injection in wild-type but not in γδT cell-deficient mice. Conclusion These findings suggest a novel regulatory mechanism of γδT cells through TL1A and its involvement in psoriasis pathogenesis as a possible therapeutic target.
Collapse
Affiliation(s)
- Shangyi Wang
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mina Kozai
- Division of Vaccinology for Clinical Development, Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Masaya Hiraishi
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Md. Zahir Uddin Rubel
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Laboratory of Agrobiomedical Science, Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Mutsumi Inaba
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuhiro Matsuo
- Division of Vaccinology for Clinical Development, Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Kensuke Takada
- Division of Vaccinology for Clinical Development, Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| |
Collapse
|
22
|
Cai X, Li Y, Liu Q, Gao X, Li J. Exploration of the Shared Gene Signatures and Molecular Mechanisms between Chronic Bronchitis and Antineutrophil Cytoplasmic Antibody-associated Glomerulonephritis: Evidence from Transcriptome Data. Curr Pharm Des 2024; 30:1966-1984. [PMID: 38847168 DOI: 10.2174/0113816128297623240521070426] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/18/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Chronic Bronchitis (CB) is a recurrent and persistent pulmonary inflammation disease. Growing evidence suggests an association between CB and Anti-neutrophil Cytoplasmic Antibody-associated Glomerulonephritis (ANCA-GN). However, the precise mechanisms underlying their association remain unclear. AIMS The purpose of this study was to further explore the molecular mechanism of the occurrence of chronic bronchitis (CB) associated with anti-neutrophil cytoplasmic antibody-associated glomerulonephritis (ANCA- GN). OBJECTIVE Our study aimed to investigate the potential shared pathogenesis of CB-associated ANCA-GN. METHODS Datasets of ANCA (GSE108113 and GSE104948) and CB (GSE151052 and GSE162635) were obtained from the Gene Expression Omnibus (GEO) datasets. Firstly, GSE108113 and GSE151052 were analyzed to identify common differentially expressed genes (DEGs) by Limma package. Based on common DEGs, protein-protein interaction (PPI) network and functional enrichment analyses, including GO, KEGG, and GSEA, were performed. Then, hub genes were identified by degree algorithm and validated in GSE104948 and GSE162635. Further PPI network and functional enrichment analyses were performed on hub genes. Additionally, a competitive ceRNA network was constructed through miRanda and spongeScan. Transcription factors (TFs) were predicted and verified using the TRRUST database. Furthermore, the CIBERSORT algorithm was employed to explore immune cell infiltration. The Drug Gene Interaction Database (DGIDB) was utilized to predict small-molecular compounds of CB and ANCA-GN. RESULTS A total of 963 DEGs were identified in the integrated CB dataset, and 610 DEGs were identified in the integrated ANCA-GN dataset. Totally, we identified 22 common DEGs, of which 10 hub genes (LYZ, IRF1, PIK3CG, IL2RG, NT5E, ARG2, HBEGF, NFATC2, ALPL, and FKBP5) were primarily involved in inflammation and immune responses. Focusing on hub genes, we constructed a ceRNA network composed of 323 miRNAs and 348 lncRNAs. Additionally, five TFs (SP1, RELA, NFKB1, HIF1A, and SP3) were identified to regulate the hub genes. Furthermore, immune cell infiltration results revealed immunoregulation in CB and ANCA-GN. Finally, some small-molecular compounds (Daclizumab, Aldesleukin, and NT5E) were predicted to predominantly regulate inflammation and immunity, especially IL-2. CONCLUSION Our study explores the inflammatory-immune pathways underlying CB-associated ANCA-GN and emphasizes the importance of NETs and lymphocyte differentiation, providing novel insights into the shared pathogenesis and therapeutic targets.
Collapse
Affiliation(s)
- Xiaojing Cai
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yueqiang Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingquan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Gao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junhua Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Venerito V, Del Vescovo S, Lopalco G, Proft F. Beyond the horizon: Innovations and future directions in axial-spondyloarthritis. Arch Rheumatol 2023; 38:491-511. [PMID: 38125058 PMCID: PMC10728740 DOI: 10.46497/archrheumatol.2023.10580] [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: 11/18/2023] [Accepted: 11/18/2023] [Indexed: 12/23/2023] Open
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory disease of the spine and sacroiliac joints. This review discusses recent advances across multiple scientific fields that promise to transform axSpA management. Traditionally, axSpA was considered an immune-mediated disease driven by human leukocyte antigen B27 (HLA-B27), interleukin (IL)-23/IL-17 signaling, biomechanics, and dysbiosis. Diagnosis relies on clinical features, laboratory tests, and imaging, particularly magnetic resonance imaging (MRI) nowadays. Management includes exercise, lifestyle changes, non-steroidal anti-inflammatory drugs and if this is not sufficient to achieve disease control also biological and targeted-synthetic disease modifying anti-rheumatic drugs. Beyond long-recognized genetic risks like HLA-B27, high-throughput sequencing has revealed intricate gene-environment interactions influencing dysbiosis, immune dysfunction, and aberrant bone remodeling. Elucidating these mechanisms promises screening approaches to enable early intervention. Advanced imaging is revolutionizing the assessment of axSpA's hallmark: sacroiliac bone-marrow edema indicating inflammation. Novel magnetic resonance imaging (MRI) techniques sensitively quantify disease activity, while machine learning automates complex analysis to improve diagnostic accuracy and monitoring. Hybrid imaging like synthetic MRI/computed tomography (CT) visualizes structural damage with new clarity. Meanwhile, microbiome analysis has uncovered gut ecosystem alterations that may initiate joint inflammation through HLA-B27 misfolding or immune subversion. Correcting dysbiosis represents an enticing treatment target. Moving forward, emerging techniques must augment patient care. Incorporating patient perspectives will be key to ensure innovations like genetics, microbiome, and imaging biomarkers translate into improved mobility, reduced pain, and increased quality of life. By integrating cutting-edge, multidisciplinary science with patients' lived experience, researchers can unlock the full potential of new technologies to deliver transformative outcomes. The future is bright for precision diagnosis, tightly controlled treatment, and even prevention of axSpA.
Collapse
Affiliation(s)
- Vincenzo Venerito
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Polyclinic Hospital, University of Bari, Bari, Italy
| | - Sergio Del Vescovo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Polyclinic Hospital, University of Bari, Bari, Italy
| | - Giuseppe Lopalco
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Polyclinic Hospital, University of Bari, Bari, Italy
| | - Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
24
|
Shah AM, Aral AM, Zamora R, Gharpure N, El-Dehaibi F, Zor F, Kulahci Y, Karagoz H, Barclay DA, Yin J, Breidenbach W, Tuder D, Gorantla VS, Vodovotz Y. Peripheral nerve repair is associated with augmented cross-tissue inflammation following vascularized composite allotransplantation. Front Immunol 2023; 14:1151824. [PMID: 37251389 PMCID: PMC10213935 DOI: 10.3389/fimmu.2023.1151824] [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: 01/26/2023] [Accepted: 04/20/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Vascularized composite allotransplantation (VCA), with nerve repair/coaptation (NR) and tacrolimus (TAC) immunosuppressive therapy, is used to repair devastating traumatic injuries but is often complicated by inflammation spanning multiple tissues. We identified the parallel upregulation of transcriptional pathways involving chemokine signaling, T-cell receptor signaling, Th17, Th1, and Th2 pathways in skin and nerve tissue in complete VCA rejection compared to baseline in 7 human hand transplants and defined increasing complexity of protein-level dynamic networks involving chemokine, Th1, and Th17 pathways as a function of rejection severity in 5 of these patients. We next hypothesized that neural mechanisms may regulate the complex spatiotemporal evolution of rejection-associated inflammation post-VCA. Methods For mechanistic and ethical reasons, protein-level inflammatory mediators in tissues from Lewis rats (8 per group) receiving either syngeneic (Lewis) or allogeneic (Brown-Norway) orthotopic hind limb transplants in combination with TAC, with and without sciatic NR, were compared to human hand transplant samples using computational methods. Results In cross-correlation analyses of these mediators, VCA tissues from human hand transplants (which included NR) were most similar to those from rats undergoing VCA + NR. Based on dynamic hypergraph analyses, NR following either syngeneic or allogeneic transplantation in rats was associated with greater trans-compartmental localization of early inflammatory mediators vs. no-NR, and impaired downregulation of mediators including IL-17A at later times. Discussion Thus, NR, while considered necessary for restoring graft function, may also result in dysregulated and mis-compartmentalized inflammation post-VCA and therefore necessitate mitigation strategies. Our novel computational pipeline may also yield translational, spatiotemporal insights in other contexts.
Collapse
Affiliation(s)
- Ashti M. Shah
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ali Mubin Aral
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nitin Gharpure
- Department of Surgery, Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston Salem, NC, United States
| | - Fayten El-Dehaibi
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fatih Zor
- Department of Surgery, Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston Salem, NC, United States
| | - Yalcin Kulahci
- Department of Surgery, Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston Salem, NC, United States
| | - Huseyin Karagoz
- Department of Surgery, Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston Salem, NC, United States
| | - Derek A. Barclay
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jinling Yin
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Dmitry Tuder
- Plastic Surgery, San Antonio Military Medical Center, Fort Sam Houston, San Antonio, TX, United States
| | - Vijay S. Gorantla
- Department of Surgery, Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston Salem, NC, United States
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
25
|
Wang T, Su X, Peng J, Tan X, Yang G, Zhang T, Chen F, Wang C, Ma K. Deciphering the pharmacological mechanisms of Fraxini Cortex for ulcerative colitis treatment based on network pharmacology and in vivo studies. BMC Complement Med Ther 2023; 23:152. [PMID: 37161415 PMCID: PMC10170718 DOI: 10.1186/s12906-023-03983-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 04/28/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a common type of inflammatory bowel disease. Due to the elusive pathogenesis, safe and effective treatment strategies are still lacking. Fraxini Cortex (FC) has been widely used as a medicinal herb to treat some diseases. However, the pharmacological mechanisms of FC for UC treatment are still unclear. METHODS An integrated platform combining network pharmacology and experimental studies was introduced to decipher the mechanism of FC against UC. The active compounds, therapeutic targets, and the molecular mechanism of action were acquired by network pharmacology, and the interaction between the compounds and target proteins were verified by molecular docking. Dextran sulfate sodium (DSS)-induced colitis model was employed to assess the therapeutic effect of FC on UC, and validate the molecular mechanisms of action predicted by network pharmacology. RESULTS A total of 20 bioactive compounds were retrieved, and 115 targets were predicted by using the online databases. Ursolic acid, fraxetin, beta-sitosterol, and esculetin were identified as the main active compounds of FC against UC. PPI network analysis identified 28 FC-UC hub genes that were mainly enriched in the IL-17 signaling pathway, the TNF signaling pathway, and pathways in cancer. Molecular docking confirmed that the active compounds had high binding affinities to the predicted target proteins. GEO dataset analysis showed that these target genes were highly expressed in the UC clinical samples compared with that in the healthy controls. Experimental studies showed that FC alleviated DSS-induced colitis symptoms, reduced inflammatory cytokines release, and suppressed the expression levels of IL1β, COX2, MMP3, IL-17 and RORγt in colon tissues. CONCLUSION FC exhibits anti-UC properties through regulating multi-targets and multi-pathways with multi-components. In vivo results demonstrated that FC alleviated DSS-induced colitis.
Collapse
Affiliation(s)
- Tianming Wang
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Xuyang Su
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China
| | - Jing Peng
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China
| | - Xiaofen Tan
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China
| | - Guangshan Yang
- The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, 230001, People's Republic of China
| | - Tengyue Zhang
- The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, 230001, People's Republic of China
| | - Feng Chen
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China
| | - Changzhong Wang
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China
| | - Kelong Ma
- College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China.
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China.
| |
Collapse
|
26
|
Del Vescovo S, Venerito V, Iannone C, Lopalco G. Uncovering the Underworld of Axial Spondyloarthritis. Int J Mol Sci 2023; 24:6463. [PMID: 37047435 PMCID: PMC10095023 DOI: 10.3390/ijms24076463] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Axial spondyloarthritis (axial-SpA) is a multifactorial disease characterized by inflammation in sacroiliac joints and spine, bone reabsorption, and aberrant bone deposition, which may lead to ankylosis. Disease pathogenesis depends on genetic, immunological, mechanical, and bioenvironmental factors. HLA-B27 represents the most important genetic factor, although the disease may also develop in its absence. This MHC class I molecule has been deeply studied from a molecular point of view. Different theories, including the arthritogenic peptide, the unfolded protein response, and HLA-B27 homodimers formation, have been proposed to explain its role. From an immunological point of view, a complex interplay between the innate and adaptive immune system is involved in disease onset. Unlike other systemic autoimmune diseases, the innate immune system in axial-SpA has a crucial role marked by abnormal activity of innate immune cells, including γδ T cells, type 3 innate lymphoid cells, neutrophils, and mucosal-associated invariant T cells, at tissue-specific sites prone to the disease. On the other hand, a T cell adaptive response would seem involved in axial-SpA pathogenesis as emphasized by several studies focusing on TCR low clonal heterogeneity and clonal expansions as well as an interindividual sharing of CD4/8 T cell receptors. As a result of this immune dysregulation, several proinflammatory molecules are produced following the activation of tangled intracellular pathways involved in pathomechanisms of axial-SpA. This review aims to expand the current understanding of axial-SpA pathogenesis, pointing out novel molecular mechanisms leading to disease development and to further investigate potential therapeutic targets.
Collapse
Affiliation(s)
- Sergio Del Vescovo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Polyclinic Hospital, University of Bari, 70124 Bari, Italy
| | - Vincenzo Venerito
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Polyclinic Hospital, University of Bari, 70124 Bari, Italy
| | - Claudia Iannone
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, 20122 Milan, Italy
| | - Giuseppe Lopalco
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Polyclinic Hospital, University of Bari, 70124 Bari, Italy
| |
Collapse
|
27
|
Lavazais S, Jargosch M, Dupont S, Labéguère F, Menet C, Jagerschmidt C, Ohm F, Kupcsik L, Parent I, Cottereaux C, Marsais F, Oste L, Van de Water A, Christophe T, De Vos S, Fallon P, Lauffer F, Clément-Lacroix P, Eyerich K, Brys R. IRAK4 inhibition dampens pathogenic processes driving inflammatory skin diseases. Sci Transl Med 2023; 15:eabj3289. [PMID: 36791209 DOI: 10.1126/scitranslmed.abj3289] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Innate immunity not only shapes the way epithelial barriers interpret environmental cues but also drives adaptive responses. Therefore, modulators of innate immune responses are expected to have high therapeutic potential across immune-mediated inflammatory diseases. IRAK4 is a kinase that integrates signaling downstream of receptors acting at the interface between innate and adaptive immune responses, such as Toll-like receptors (TLRs), interleukin-1R (IL-1R), and IL-18R. Because effects of IRAK4 inhibition are stimulus, cell type, and species dependent, the evaluation of the therapeutic potential of IRAK4 inhibitors requires a highly translational approach. Here, we profiled a selective IRAK4 inhibitor, GLPG2534, in an extensive panel of models of inflammatory skin diseases, translationally expanding evidence from in vitro to in vivo and from mouse to human. In vitro, IRAK4 inhibition resulted in substantial inhibition of TLR and IL-1 responses in dendritic cells, keratinocytes, granulocytes, and T cells but only weakly affected dermal fibroblast responses. Furthermore, disease activity in murine models of skin inflammation (IL-23-, IL-33-, imiquimod-, and MC903-induced) was markedly dampened by IRAK4 inhibition. Last, inhibiting IRAK4 reversed pathogenic molecular signatures in human lesional psoriasis and atopic dermatitis biopsies. Over the variety of models used, IRAK4 inhibition consistently affected central mediators of psoriasis (IL-17A) and atopic dermatitis (IL-4 and IL-13). Overall, our data highlight IRAK4 as a central player in skin inflammatory processes and demonstrate the potential of IRAK4 inhibition as a therapeutic strategy in chronic inflammatory skin diseases.
Collapse
Affiliation(s)
| | - Manja Jargosch
- Department of Dermatology and Allergy, Technical University of Munich, 80802 Munich, Germany
| | | | | | | | | | - Frenz Ohm
- Department of Dermatology and Allergy, Technical University of Munich, 80802 Munich, Germany
| | | | | | | | | | - Line Oste
- Galapagos NV, 2800 Mechelen, Belgium
| | | | | | | | - Padraic Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Felix Lauffer
- Department of Dermatology and Allergy, Technical University of Munich, 80802 Munich, Germany
| | | | - Kilian Eyerich
- Department of Dermatology and Venerology, Medical Center-University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
28
|
Fan JL, Wu D, Zhu TT, Tian XL, Liu SJ, Zhang SL. The exploration of shared genes and molecular mechanisms of systemic lupus erythematosus and atherosclerosis. Lupus 2023; 32:239-251. [PMID: 36480924 DOI: 10.1177/09612033221144596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Despite widespread recognition, the mechanisms underlying the relationship between systemic lupus erythematosus (SLE) and atherosclerosis (AS) are still unclear. Our study aimed to explore the shared genetic signature and molecular mechanisms of SLE and AS using a bioinformatics approach. METHODS Gene expression profiles of GSE50772 (contains peripheral blood mononuclear cells from 61 SLE patients and 20 normal samples) and GSE100927 (contains 69 AS plaque tissue samples and 35 control samples) were downloaded from the Gene Expression Database (GEO) before the differentially expressed genes were obtained using the "limma" package in R. The differential genes were then subjected to gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis using the DAVID online platform to annotate their functions. The intersection targets of PPI and WGCNA were used as key shared genes for SLE and AS with their diagnostic value as shared genes being verified through ROC curves. Finally, Cytoscape 3.7.2 software was used to construct a miRNA-mRNA network map associated with the shared genes. RESULTS A total of 246 DEGs were identified, including 189 upregulated genes and 57 downregulated genes, which were mainly enriched in signaling pathways such as TNF signaling pathway, IL-17 signaling pathway, and NF-kB signaling pathway. The molecular basis for the relationship between SLE and AS may be the aforementioned signaling pathways. Following ROC curve validation, the intersection of PPI and WGCNA, as well as AQP9, CCR1, CD83, CXCL1, and FCGR2A, resulted in the identification of 15 shared genes. CONCLUSION The study provided a new perspective on the common molecular mechanisms between SLE and AS, and the key genes and pathways that were identified as being part of these pathways may offer fresh perspectives and suggestions for further experimental research.
Collapse
Affiliation(s)
- Ji-Lin Fan
- First Clinical School of Medicine, 74738Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Wu
- Department of Neurosurgery, 605788Binzhou Medical University Hospital, Binzhou, China
| | - Ting-Ting Zhu
- Department of Cardiology, 612366The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Ling Tian
- Department of Cardiology, 612366The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Si-Jia Liu
- Department of Neurosurgery, 605788Binzhou Medical University Hospital, Binzhou, China
| | - Shi-Liang Zhang
- Department of Neurosurgery, 605788Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
29
|
Time-resolved RNA signatures of CD4+ T cells in Parkinson's disease. Cell Death Dis 2023; 9:18. [PMID: 36681665 PMCID: PMC9867723 DOI: 10.1038/s41420-023-01333-0] [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: 11/07/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/22/2023]
Abstract
Parkinson's disease (PD) emerges as a complex, multifactorial disease. While there is increasing evidence that dysregulated T cells play a central role in PD pathogenesis, elucidation of the pathomechanical changes in related signaling is still in its beginnings. We employed time-resolved RNA expression upon the activation of peripheral CD4+ T cells to track and functionally relate changes on cellular signaling in representative cases of patients at different stages of PD. While only few miRNAs showed time-course related expression changes in PD, we identified groups of genes with significantly altered expression for each different time window. Towards a further understanding of the functional consequences, we highlighted pathways with decreased or increased activity in PD, including the most prominent altered IL-17 pathway. Flow cytometric analyses showed not only an increased prevalence of Th17 cells but also a specific subtype of IL-17 producing γδ-T cells, indicating a previously unknown role in PD pathogenesis.
Collapse
|
30
|
Hirano T, Kawano T, Kadowaki Y, Moriyama M, Umemoto S, Yoshinaga K, Matsunaga T, Suzuki M. Impact of IL-17-producing γδ T cells on chronic otitis media induced by nontypeable Haemophilus influenzae in a mouse model. Pathog Dis 2023; 81:ftad029. [PMID: 37833235 DOI: 10.1093/femspd/ftad029] [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: 04/24/2023] [Revised: 07/27/2023] [Accepted: 10/12/2023] [Indexed: 10/15/2023] Open
Abstract
Nontypeable Haemophilus influenzae (NTHi) is considered a major pathogen underlying middle ear infection. This study aimed to investigate the impact of IL-17 on chronic otitis media (COM) induced by NTHi in mice. NTHi was inoculated into the tympanic bulla with eustachian tubal obstruction. Middle ear effusions (MEEs) and tissues were collected on days 3, 14, and at 1, 2, and 6 months after injection. The expression of interleukin-17A (IL-17A) in MEEs was significantly elevated compared to that in the control group at the translational and transcriptional levels during the experiments. The quantities of IL-17-producing γδ T cells were significantly increased compared to that in the control group during COM, but that of Th17 cells did not. Depletion of γδ T cells by anti-γδ T-cell receptor (TCR) monoclonal antibody (mAb) administration significantly decreased the bacteria counts and the concentrations of IL-1β, IL-6, IL-17A, TNF-α, and IL-10 in MEEs. Our results suggest that IL-17 may play an important role in prolonging the inflammation in the middle ear in COM and that IL-17-producing γδ T cells may contribute to the exacerbated inflammatory response in the middle ear. In this study, anti-γδ TCR mAb administration was found to improve chronic middle ear inflammatory conditions.
Collapse
Affiliation(s)
- Takashi Hirano
- Department of Otolaryngology, Faculty of Medicine, Oita University, Hasama-machi, Oita 879-5593, Japan
| | - Toshiaki Kawano
- Department of Otolaryngology, Faculty of Medicine, Oita University, Hasama-machi, Oita 879-5593, Japan
| | - Yoshinori Kadowaki
- Department of Otolaryngology, Faculty of Medicine, Oita University, Hasama-machi, Oita 879-5593, Japan
| | - Munehito Moriyama
- Department of Otolaryngology, Faculty of Medicine, Oita University, Hasama-machi, Oita 879-5593, Japan
| | - Shingo Umemoto
- Department of Otolaryngology, Faculty of Medicine, Oita University, Hasama-machi, Oita 879-5593, Japan
| | - Kazuhiro Yoshinaga
- Department of Otolaryngology, Faculty of Medicine, Oita University, Hasama-machi, Oita 879-5593, Japan
| | - Takayuki Matsunaga
- Department of Otolaryngology, Faculty of Medicine, Oita University, Hasama-machi, Oita 879-5593, Japan
| | - Masashi Suzuki
- Department of Otolaryngology, Faculty of Medicine, Oita University, Hasama-machi, Oita 879-5593, Japan
| |
Collapse
|
31
|
Singh V, Lee G, Son H, Koh H, Kim ES, Unno T, Shin JH. Butyrate producers, "The Sentinel of Gut": Their intestinal significance with and beyond butyrate, and prospective use as microbial therapeutics. Front Microbiol 2023; 13:1103836. [PMID: 36713166 PMCID: PMC9877435 DOI: 10.3389/fmicb.2022.1103836] [Citation(s) in RCA: 201] [Impact Index Per Article: 100.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Gut-microbial butyrate is a short-chain fatty acid (SCFA) of significant physiological importance than the other major SCFAs (acetate and propionate). Most butyrate producers belong to the Clostridium cluster of the phylum Firmicutes, such as Faecalibacterium, Roseburia, Eubacterium, Anaerostipes, Coprococcus, Subdoligranulum, and Anaerobutyricum. They metabolize carbohydrates via the butyryl-CoA: acetate CoA-transferase pathway and butyrate kinase terminal enzymes to produce most of butyrate. Although, in minor fractions, amino acids can also be utilized to generate butyrate via glutamate and lysine pathways. Butyrogenic microbes play a vital role in various gut-associated metabolisms. Butyrate is used by colonocytes to generate energy, stabilizes hypoxia-inducible factor to maintain the anaerobic environment in the gut, maintains gut barrier integrity by regulating Claudin-1 and synaptopodin expression, limits pro-inflammatory cytokines (IL-6, IL-12), and inhibits oncogenic pathways (Akt/ERK, Wnt, and TGF-β signaling). Colonic butyrate producers shape the gut microbial community by secreting various anti-microbial substances, such as cathelicidins, reuterin, and β-defensin-1, and maintain gut homeostasis by releasing anti-inflammatory molecules, such as IgA, vitamin B, and microbial anti-inflammatory molecules. Additionally, butyrate producers, such as Roseburia, produce anti-carcinogenic metabolites, such as shikimic acid and a precursor of conjugated linoleic acid. In this review, we summarized the significance of butyrate, critically examined the role and relevance of butyrate producers, and contextualized their importance as microbial therapeutics.
Collapse
Affiliation(s)
- Vineet Singh
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - GyuDae Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - HyunWoo Son
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hong Koh
- Department of Pediatrics, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Soo Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Tatsuya Unno
- Faculty of Biotechnology, School of Life Sciences, SARI, Jeju National University, Jeju, Republic of Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
- Department of Integrative Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
32
|
Fears AC, Walker EM, Chirichella N, Slisarenko N, Merino KM, Golden N, Picou B, Spencer S, Russell-Lodrigue KE, Doyle-Meyers LA, Blair RV, Beddingfield BJ, Maness NJ, Roy CJ, Rout N. The dynamics of γδ T cell responses in nonhuman primates during SARS-CoV-2 infection. Commun Biol 2022; 5:1380. [PMID: 36526890 PMCID: PMC9756695 DOI: 10.1038/s42003-022-04310-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Although most SARS-CoV-2 infections are mild, some patients develop systemic inflammation and progress to acute respiratory distress syndrome (ARDS). However, the cellular mechanisms underlying this spectrum of disease remain unclear. γδT cells are T lymphocyte subsets that have key roles in systemic and mucosal immune responses during infection and inflammation. Here we show that peripheral γδT cells are rapidly activated following aerosol or intra-tracheal/intra-nasal (IT/IN) SARS-CoV-2 infection in nonhuman primates. Our results demonstrate a rapid expansion of Vδ1 γδT cells at day1 that correlate significantly with lung viral loads during the first week of infection. Furthermore, increase in levels of CCR6 and Granzyme B expression in Vδ1 T cells during viral clearance imply a role in innate-like epithelial barrier-protective and cytotoxic functions. Importantly, the early activation and mobilization of circulating HLA-DR+CXCR3+ γδT cells along with significant correlations of Vδ1 T cells with IL-1Ra and SCF levels in bronchoalveolar lavage suggest a novel role for Vδ1 T cells in regulating lung inflammation during aerosol SARS-CoV-2 infection. A deeper understanding of the immunoregulatory functions of MHC-unrestricted Vδ1 T cells in lungs during early SARS-CoV-2 infection is particularly important in the wake of emerging new variants with increased transmissibility and immune evasion potential.
Collapse
Affiliation(s)
- Alyssa C Fears
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Edith M Walker
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Nicole Chirichella
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Nadia Slisarenko
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Kristen M Merino
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Nadia Golden
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Breanna Picou
- High Containment Research Performance Core, Tulane National Primate Research Center, Covington, LA, USA
| | - Skye Spencer
- High Containment Research Performance Core, Tulane National Primate Research Center, Covington, LA, USA
| | - Kasi E Russell-Lodrigue
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, USA
| | - Lara A Doyle-Meyers
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, USA
| | - Robert V Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | | | - Nicholas J Maness
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane School of Medicine, New Orleans, LA, USA
| | - Chad J Roy
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane School of Medicine, New Orleans, LA, USA
| | - Namita Rout
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA.
- Department of Microbiology and Immunology, Tulane School of Medicine, New Orleans, LA, USA.
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
33
|
Nezhad Shamohammadi F, Yazdanifar M, Oraei M, Kazemi MH, Roohi A, Mahya Shariat Razavi S, Rezaei F, Parvizpour F, Karamlou Y, Namdari H. Controversial role of γδ T cells in pancreatic cancer. Int Immunopharmacol 2022; 108:108895. [PMID: 35729831 DOI: 10.1016/j.intimp.2022.108895] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/12/2022] [Accepted: 05/23/2022] [Indexed: 12/26/2022]
Abstract
γδ T cells are rare lymphocytes with cogent impact on immune responses. These cells are one of the earliest cells to be recruited in the sites of infection or tumors and play a critical role in coordinating innate and adaptive immune responses. The anti-tumor activity of γδ T cells have been numerously reported; nonetheless, there is controversy among published studies regarding their anti-tumor vs pro-tumor effect- especially in pancreatic cancer. A myriad of studies has confirmed that activated γδ T cells can potently lyse a broad variety of solid tumors and leukemia/lymphoma cells and produce an array of cytokines; however, early γδ T cell-based clinical trials did not lead to optimal efficacy, despite acceptable safety. Depending on the local micromilieu, γδ T cells can differentiate into tumor promoting or suppressing cells such as Th1-, Th2-, or Th17-like cells and produce prototypical cytokines such as interferon-γ (IFNγ) and interleukin (IL)-4/-10, IL-9, or IL-17. In an abstruse tumor such as pancreatic cancer- also known as immunologically cold tumor- γδ T cells are more likely to switch to their immunosuppressive phenotype. In this review we will adduce the accumulated knowledge on these two controversial aspects of γδ T cells in cancers- with a focus on solid tumors and pancreatic cancer. In addition, we propose strategies for enhancing the anti-tumor function of γδ T cells in cancers and discuss the potential future directions.
Collapse
Affiliation(s)
| | - Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Mona Oraei
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad H Kazemi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Azam Roohi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Parvizpour
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yalda Karamlou
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Haideh Namdari
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
High-dimensional profiling reveals Tc17 cell enrichment in active Crohn's disease and identifies a potentially targetable signature. Nat Commun 2022; 13:3688. [PMID: 35760777 PMCID: PMC9237103 DOI: 10.1038/s41467-022-31229-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 05/25/2022] [Indexed: 11/20/2022] Open
Abstract
The immune-pathology in Crohn’s disease is linked to dysregulated CD4+ T cell responses biased towards pathogenic TH17 cells. However, the role of CD8+ T cells able to produce IL-17 (Tc17 cells) remains unclear. Here we characterize the peripheral blood and intestinal tissue of Crohn’s disease patients (n = 61) with flow and mass cytometry and reveal a strong increase of Tc17 cells in active disease, mainly due to induction of conventional T cells. Mass cytometry shows that Tc17 cells express a distinct immune signature (CD6high, CD39, CD69, PD-1, CD27low) which was validated in an independent patient cohort. This signature stratifies patients into groups with distinct flare-free survival associated with differential CD6 expression. Targeting of CD6 in vitro reduces IL-17, IFN-γ and TNF production. These results identify a distinct Tc17 cell population in Crohn’s disease with proinflammatory features linked to disease activity. The Tc17 signature informs clinical outcomes and may guide personalized treatment decisions. The T cell compartment in patients with Crohn's disease is dysregulated. Here the authors use cytometric profiling to reveal an enrichment of distinct Tc17 cells during active Crohn's disease and may suggest CD6 as a potential target for therapeutic studies.
Collapse
|
35
|
Kabeerdoss J, Danda D, Goel R, Mohan H, Danda S, Scofield RH. Genome-Wide DNA Methylation Profiling in CD8 T-Cells and Gamma Delta T-Cells of Asian Indian Patients With Takayasu Arteritis. Front Cell Dev Biol 2022; 10:843413. [PMID: 35813204 PMCID: PMC9259853 DOI: 10.3389/fcell.2022.843413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Takayasu’s Arteritis (TA) is a chronic inflammatory disease that affects aorta and its main branches at their origin. Genetic, pathological and functional studies have shown that CD8 and Gamma delta (γ/δ) T-lymphocytes are involved in inflammatory processes in affected regions of arteries causing vascular damage. The molecular function of these lymphocytes remains unclear and currently no epigenetic studies are available in TA. We primarily performed genome wide methylation analysis in CD8 T cells and γδ T cells of patients with TA and compared with healthy controls. Methods: We recruited 12 subjects in each group namely TA patient and healthy controls. Blood samples were collected after obtaining informed written consent. CD8 T cells and γδ T cells were separated from whole blood. DNA extracted from these cells and were subjected to bisulfite treatment. Finally, bisulfite treated DNA was loaded in Infinium Methylation EPIC array. Bioinformatics analysis was used to identify differential methylation regions which were then mapped to genes. Results: Interleukin (IL)-32 and Lymphotoxin-A were genes significantly hypomethylated in CD8 T-cells. Anti-inflammatory cytokine genes, IL-10, IL-1RN and IL-27 were hypomethylated in γδ T cells of TA patients as compared to healthy controls. Gene enrichment analysis using Gene Ontology (GO) database and Kyoto Encyclopaedia of Genes and Genomes (KEGG) identified that genes involved in T-cell receptor signalling pathways were hypomethylated in CD8 T-cells and hypermethylated in γδ T cells of TA patients. Conclusion: CD8 T-cells might play a major role in immunopathogenesis of inflammation in TA, whereas γδ T cells may play a regulatory role.
Collapse
Affiliation(s)
- Jayakanthan Kabeerdoss
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, India
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
- *Correspondence: Debashish Danda, ; Jayakanthan Kabeerdoss,
| | - Debashish Danda
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, India
- *Correspondence: Debashish Danda, ; Jayakanthan Kabeerdoss,
| | - Ruchika Goel
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, India
| | - Hindhumathi Mohan
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, India
| | - Sumita Danda
- Department of Medical Genetics, Christian Medical College, Vellore, India
| | - R. Hal Scofield
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Veterans Affairs, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
36
|
Liu L, Chen X, Lu Y, Sun XY, Ze K, Zhou YQ, Li W, Li X, Li HJ, Li B. Celastrol gel ameliorates imiquimod-induced psoriasis-like dermatitis in mice by targeting Langerhans cells. Biomed Pharmacother 2022; 147:112644. [DOI: 10.1016/j.biopha.2022.112644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 01/03/2023] Open
|
37
|
Zhang W, Pajulas A, Kaplan MH. γδ T Cells in Skin Inflammation. Crit Rev Immunol 2022; 42:43-56. [PMID: 37075018 PMCID: PMC10439530 DOI: 10.1615/critrevimmunol.2022047288] [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] [Indexed: 01/05/2023]
Abstract
Gamma delta (γδ) T cells are a subset of T lymphocytes that express T cell receptor γ and 5 chains and display structural and functional heterogeneity. γδ T cells are typically of low abundance in the body and account for 1-5% of the blood lymphocytes and peripheral lymphoid tissues. As a bridge between innate and adaptive immunity, γδ T cells are uniquely poised to rapidly respond to stimulation and can regulate immune responses in peripheral tissues. The dendritic epidermal T cells in the skin epidermis can secrete growth factors to regulate skin homeostasis and re-epithelization and release inflammatory factors to mediate wound healing during skin inflammatory responses. Dermal γδ T cells can regulate the inflammatory process by producing interleukin-17 and other cytokines or chemokines. Here, we offer a review of the immune functions of γδ T cells, intending to understand their role in regulating skin barrier integrity and skin wound healing, which may be crucial for the development of novel therapeutics in skin diseases like atopic dermatitis and psoriasis.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Microbiology & Immunology, Indiana University School Medicine, Indianapolis, IN 46202
| | - Abigail Pajulas
- Department of Microbiology & Immunology, Indiana University School Medicine, Indianapolis, IN 46202
| | - Mark H Kaplan
- Department of Microbiology & Immunology, Indiana University School Medicine, Indianapolis, IN 46202
| |
Collapse
|
38
|
Pu X, Li F, Lin X, Wang R, Chen Z. Oxidative stress and expression of inflammatory factors in lung tissue of acute mountain sickness rats. Mol Med Rep 2021; 25:49. [PMID: 34913080 PMCID: PMC8711020 DOI: 10.3892/mmr.2021.12565] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/27/2021] [Indexed: 11/12/2022] Open
Abstract
The aim of the present study was to investigate the changes in lung histomorphology and oxidative stress, as well as the expression of interleukin (IL)-17C and other inflammatory factors during acute mountain sickness (AMS) in male Sprague-Dawley rats and to explore the underlying mechanism. Rats were randomly divided into a control group (0 h) and three hypoxia stress groups, exposed to low-pressure oxygen storage at a simulated altitude of 6,000 m for 24, 48 and 72 h, respectively. Morphological changes in lung tissue were observed by hematoxylin and eosin staining under light microscopy and transmission electron microscopy. The expression of inflammatory factors IL-17C, nuclear factor-κB (NF-κB), IL-1β, IL-6 and tumor necrosis factor-α (TNF-α) in lung tissue was assessed by RNA sequencing and verified by reverse transcription-quantitative PCR (RT-qPCR) and western blotting (WB). Superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) enzyme activity and malondialdehyde (MDA) expression were also measured. Experimental groups were compared to the control group following 24, 48 and 72 h of hypoxic stress. Lung tissue suffered from different degrees of injury, and the damage was the most severe after 48 h of hypoxic stress. RNA sequencing data from the lung tissue of rats from each group suggested that the expression of IL-17C, NF-κB, IL-1β, IL-6, and TNF-α increased significantly after hypoxic stress. RT-qPCR and WB demonstrated that the expression of IL-17C and NF-κB increased significantly after hypoxia lasting 48 and 72 h. IL-1β expression increased significantly after hypoxia stress lasting 24 and 48 h, and the expressions of TNF-α and IL-6 increased significantly after hypoxia stress lasting 24, 48 and 72 h (P<0.01). The enzyme activity of SOD and GSH-Px decreased significantly after lasting 24, 48 and 72 h of hypoxia (P<0.01), and MDA increased significantly after hypoxic stress lasting 48 and 72 h (P<0.01). In conclusion, under hypoxic stress, rats quickly initiate oxidative stress and immune responses. However, with prolonged hypoxic stress time, excessive oxidative stress can further stimulate the immune system in vivo, and release a large quantity of inflammatory factors accumulating in the body. This, in turn, may lead to the occurrence of inflammatory storms and further damage the lung tissue resulting in AMS.
Collapse
Affiliation(s)
- Xiaoyan Pu
- Qinghai Normal University, Xining, Qinghai 810001, P.R. China
| | - Fuxin Li
- College of Medicine, Qinghai University, Xining, Qinghai 810001, P.R. China
| | - Xue Lin
- College of Medicine, Qinghai University, Xining, Qinghai 810001, P.R. China
| | - Rong Wang
- College of Medicine, Qinghai University, Xining, Qinghai 810001, P.R. China
| | - Zhi Chen
- Qinghai Normal University, Xining, Qinghai 810001, P.R. China
| |
Collapse
|
39
|
Wang S, Kozai M, Mita H, Cai Z, Masum MA, Ichii O, Takada K, Inaba M. REV-ERB agonist suppresses IL-17 production in γδT cells and improves psoriatic dermatitis in a mouse model. Biomed Pharmacother 2021; 144:112283. [PMID: 34628169 DOI: 10.1016/j.biopha.2021.112283] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 11/26/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by epidermal hyperplasia and cellular infiltration. Studies have shown that disease development depends on proinflammatory cytokines, such as interleukin (IL)-23 and IL-17. It has been suggested that IL-23 produced by innate immune cells, such as macrophages, stimulates a subset of helper T cells to release IL-17, promoting neutrophil recruitment and keratinocyte proliferation. However, recent studies have revealed the crucial role of γδT cells in psoriasis pathogenesis as the primary source of dermal IL-17. The nuclear receptors REV-ERBs are ligand-dependent transcription factors recognized as circadian rhythm regulators. REV-ERBs negatively regulate IL-17-producing helper T cells, whereas the involvement of REV-ERBs in regulating IL-17-producing γδT (γδT17) cells remains unclear. Here we revealed the regulatory mechanism involving γδT17 cells through REV-ERBs. γδT17 cell levels were remarkably elevated in the secondary lymphoid organs of mice that lacked an isoform of REV-ERBs. A synthetic REV-ERB agonist, SR9009, suppressed γδT17 cells in vitro and in vivo. Topical application of SR9009 to the skin reduced the inflammatory symptoms of psoriasiform dermatitis in mice. The results of this study provide a novel therapeutic approach for psoriasis targeting REV-ERBs in γδT17 cells.
Collapse
MESH Headings
- Administration, Cutaneous
- Animals
- Anti-Inflammatory Agents/administration & dosage
- Anti-Inflammatory Agents/pharmacology
- Cells, Cultured
- Disease Models, Animal
- Down-Regulation
- Female
- Interleukin-17/metabolism
- Intraepithelial Lymphocytes/drug effects
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
- Mice, Knockout
- Nuclear Receptor Subfamily 1, Group D, Member 1/agonists
- Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
- Psoriasis/drug therapy
- Psoriasis/immunology
- Psoriasis/metabolism
- Psoriasis/pathology
- Pyrrolidines/administration & dosage
- Pyrrolidines/pharmacology
- Signal Transduction
- Skin/drug effects
- Skin/immunology
- Skin/metabolism
- Thiophenes/administration & dosage
- Thiophenes/pharmacology
- Mice
Collapse
Affiliation(s)
- Shangyi Wang
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mina Kozai
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hironobu Mita
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Zimeng Cai
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Md Abdul Masum
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Agrobiomedical Science, Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Kensuke Takada
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| | - Mutsumi Inaba
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
40
|
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: 0.5] [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
|
41
|
Wilson PA, Santos Franco S, He L, Galwey NW, Meakin J, McIntyre R, McHugh SM, Nolan MA, Spain SL, Carlson T, Lobera M, Rubio JP, Davis B, McCarthy LC. Transcriptomic effects of rs4845604, an IBD and allergy-associated RORC variant, in stimulated ex vivo CD4+ T cells. PLoS One 2021; 16:e0258316. [PMID: 34673799 PMCID: PMC8530322 DOI: 10.1371/journal.pone.0258316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/23/2021] [Indexed: 11/18/2022] Open
Abstract
RORγt is an isoform of RORC, preferentially expressed in Th17 cells, that functions as a critical regulator of type 3 immunity. As murine Th17-driven inflammatory disease models were greatly diminished in RORC knock-out mice, this receptor was prioritised as an attractive therapeutic target for the treatment of several autoimmune diseases. Human genetic studies indicate a significant contributory role for RORC in several human disease conditions. Furthermore, genome-wide association studies (GWAS) report a significant association between inflammatory bowel disease (IBD) and the RORC regulatory variant rs4845604. To investigate if the rs4845604 variant may affect CD4+ T cell differentiation events, naïve CD4+ T cells were isolated from eighteen healthy subjects homozygous for the rs4845604 minor (A) or major (G) allele). Isolated cells from each subject were differentiated into distinct T cell lineages by culturing in either T cell maintenance medium or Th17 driving medium conditions for six days in the presence of an RORC inverse agonist (to prevent constitutive receptor activity) or an inactive diastereomer (control). Our proof of concept study indicated that genotype had no significant effect on the mean number of naïve CD4 T cells isolated, nor the frequency of Th1-like and Th17-like cells following six days of culture in any of the four culture conditions. Analysis of the derived RNA-seq count data identified genotype-driven transcriptional effects in each of the four culture conditions. Subsequent pathway enrichment analysis of these profiles reported perturbation of metabolic signalling networks, with the potential to affect the cellular detoxification response. This investigation reveals that rs4845604 genotype is associated with transcriptional effects in CD4+ T cells that may perturb immune and metabolic pathways. Most significantly, the rs4845604 GG, IBD risk associated, genotype may be associated with a differential detoxification response. This observation justifies further investigation in a larger cohort of both healthy and IBD-affected individuals.
Collapse
Affiliation(s)
- Paul A. Wilson
- Human Genetics, GlaxoSmithKline Medicine Research Centre, Stevenage, England
| | - Sara Santos Franco
- Clinical Unit Cambridge, Addenbrooke’s Centre for Clinical Investigation, GlaxoSmithKline, Cambridge, England
| | - Liu He
- Wellcome Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Nicholas W. Galwey
- Research Statistics, GlaxoSmithKline Medicines Research Centre, Stevenage, England
| | - Jackie Meakin
- Functional Genomics, GlaxoSmithKline Medicines Research Centre, Stevenage, England
| | | | - Simon M. McHugh
- Clinical Unit Cambridge, Addenbrooke’s Centre for Clinical Investigation, GlaxoSmithKline, Cambridge, England
| | | | | | - Thaddeus Carlson
- Adaptive Immunity, GSK Pharma Research & Development, Cambridge, MA, United States of America
| | - Mercedes Lobera
- Adaptive Immunity, GSK Pharma Research & Development, Cambridge, MA, United States of America
| | - Justin P. Rubio
- Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia
| | - Bill Davis
- Clinical Unit Cambridge, Addenbrooke’s Centre for Clinical Investigation, GlaxoSmithKline, Cambridge, England
| | - Linda C. McCarthy
- Human Genetics, GlaxoSmithKline Medicine Research Centre, Stevenage, England
- * E-mail:
| |
Collapse
|
42
|
Li G, Chen H, Liu L, Xiao P, Xie Y, Geng X, Zhang T, Zhang Y, Lu T, Tan H, Li L, Sun B. Role of Interleukin-17 in Acute Pancreatitis. Front Immunol 2021; 12:674803. [PMID: 34594321 PMCID: PMC8476864 DOI: 10.3389/fimmu.2021.674803] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/31/2021] [Indexed: 11/24/2022] Open
Abstract
Acute pancreatitis (AP) is a leading cause of death and is commonly accompanied by systemic manifestations that are generally associated with a poor prognosis. Many cytokines contribute to pancreatic tissue damage and cause systemic injury. Interleukin-17 (IL-17) is a cytokine that may play a vital role in AP. Specifically, IL-17 has important effects on the immune response and causes interactions between different inflammatory mediators in the AP-related microenvironment. In this literature review, we will discuss the existing academic understanding of IL-17 and the impacts of IL-17 in different cells (especially in acinar cells and immune system cells) in AP pathogenesis. The clinical significance and potential mechanisms of IL-17 on AP deterioration are emphasized. The evidence suggests that inhibiting the IL-17 cytokine family could alleviate the pathogenic process of AP, and we highlight therapeutic strategies that directly or indirectly target IL-17 cytokines in acute pancreatitis.
Collapse
Affiliation(s)
- Guanqun Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongze Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liwei Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng Xiao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Xie
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinglong Geng
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tao Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianqi Lu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongtao Tan
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| |
Collapse
|
43
|
Zhou Q, Xiang H, Liu H, Qi B, Shi X, Guo W, Zou J, Wan X, Wu W, Wang Z, Liu W, Xia S, Shang D. Emodin Alleviates Intestinal Barrier Dysfunction by Inhibiting Apoptosis and Regulating the Immune Response in Severe Acute Pancreatitis. Pancreas 2021; 50:1202-1211. [PMID: 34714285 PMCID: PMC8565508 DOI: 10.1097/mpa.0000000000001894] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/17/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The intestinal barrier injury caused by severe acute pancreatitis (SAP) can induce enterogenous infection, further aggravating the inflammatory reactions and immune responses. This study aimed to test the hypothesis that emodin protects the intestinal function and is involved in the immune response in SAP. METHODS The network pharmacology was established using the Swiss target prediction and pathway enrichment analysis. The SAP mice model was induced by cerulein (50 μg/kg) and lipopolysaccharide (10 mg/kg) hyperstimulation. The pharmacological effect of emodin in treating SAP was evaluated at mRNA and protein levels by various methods. RESULTS The network analysis provided the connectivity between the targets of emodin and the intestinal barrier-associated proteins and predicted the BAX/Bcl-2/caspase 3 signaling pathway. Emodin alleviated the pathological damages to the pancreas and intestine and reduced the high concentrations of serum amylase and cytokines in vivo. Emodin increased the expression of intestinal barrier-related proteins and reversed the changes in the apoptosis-related proteins in the intestine. Simultaneously, emodin regulated the ratio of T helper type 1 (TH1), TH2, TH17, γδ T cells, and interferon γ/interleukin 17 producing γδ T cells. CONCLUSIONS These findings partly verified the mechanism underlying the regulation of the intestinal barrier and immune response by emodin.
Collapse
Affiliation(s)
- Qi Zhou
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
- Institute (College) of Integrative Medicine
| | - Hong Xiang
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
| | - Han Liu
- Department of Oral Pathology, Dalian Medical University
| | - Bing Qi
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xueying Shi
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
- Institute (College) of Integrative Medicine
| | - Wenhui Guo
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
| | - Jiacheng Zou
- Department of Molecular, Cellular, & Developmental Biology, University of California, Santa Barbara, Goleta, CA
| | - Xueting Wan
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
| | - Wenjing Wu
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
| | - Zhengpeng Wang
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
| | - Wenhui Liu
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
| | - Shilin Xia
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
| | - Dong Shang
- From the Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University
- Institute (College) of Integrative Medicine
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
44
|
Byrne EM, Llorián-Salvador M, Tang M, Margariti A, Chen M, Xu H. IL-17A Damages the Blood-Retinal Barrier through Activating the Janus Kinase 1 Pathway. Biomedicines 2021; 9:831. [PMID: 34356895 PMCID: PMC8301352 DOI: 10.3390/biomedicines9070831] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/06/2021] [Accepted: 07/14/2021] [Indexed: 12/16/2022] Open
Abstract
Blood-retinal barrier (BRB) dysfunction underlies macular oedema in many sight-threatening conditions, including diabetic macular oedema, neovascular age-related macular degeneration and uveoretinitis. Inflammation plays an important role in BRB dysfunction. This study aimed to understand the role of the inflammatory cytokine IL-17A in BRB dysfunction and the mechanism involved. Human retinal pigment epithelial (RPE) cell line ARPE19 and murine brain endothelial line bEnd.3 were cultured on transwell membranes to model the outer BRB and inner BRB, respectively. IL-17A treatment (3 days in bEnd.3 cells and 6 days in ARPE19 cells) disrupted the distribution of claudin-5 in bEnd.3 cells and ZO-1 in ARPE19 cells, reduced the transepithelial/transendothelial electrical resistance (TEER) and increased permeability to FITC-tracers in vitro. Intravitreal (20 ng/1 μL/eye) or intravenous (20 ng/g) injection of recombinant IL-17A induced retinal albumin leakage within 48 h in C57BL/6J mice. Mechanistically, IL-17A induced Janus kinase 1 (JAK1) phosphorylation in bEnd.3 but not ARPE19 cells. Blocking JAK1 with Tofacitinib prevented IL-17A-mediated claudin-5 dysmorphia in bEnd.3 cells and reduced albumin leakage in IL-17A-treated mice. Our results suggest that IL-17A can damage the BRB through the activating the JAK1 signaling pathway, and targeting this pathway may be a novel approach to treat inflammation-induced macular oedema.
Collapse
Affiliation(s)
| | | | | | | | | | - Heping Xu
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (E.M.B.); (M.L.-S.); (M.T.); (A.M.); (M.C.)
| |
Collapse
|
45
|
de Sousa E, Lérias JR, Beltran A, Paraschoudi G, Condeço C, Kamiki J, António PA, Figueiredo N, Carvalho C, Castillo-Martin M, Wang Z, Ligeiro D, Rao M, Maeurer M. Targeting Neoepitopes to Treat Solid Malignancies: Immunosurgery. Front Immunol 2021; 12:592031. [PMID: 34335558 PMCID: PMC8320363 DOI: 10.3389/fimmu.2021.592031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 05/07/2021] [Indexed: 12/26/2022] Open
Abstract
Successful outcome of immune checkpoint blockade in patients with solid cancers is in part associated with a high tumor mutational burden (TMB) and the recognition of private neoantigens by T-cells. The quality and quantity of target recognition is determined by the repertoire of ‘neoepitope’-specific T-cell receptors (TCRs) in tumor-infiltrating lymphocytes (TIL), or peripheral T-cells. Interferon gamma (IFN-γ), produced by T-cells and other immune cells, is essential for controlling proliferation of transformed cells, induction of apoptosis and enhancing human leukocyte antigen (HLA) expression, thereby increasing immunogenicity of cancer cells. TCR αβ-dependent therapies should account for tumor heterogeneity and availability of the TCR repertoire capable of reacting to neoepitopes and functional HLA pathways. Immunogenic epitopes in the tumor-stroma may also be targeted to achieve tumor-containment by changing the immune-contexture in the tumor microenvironment (TME). Non protein-coding regions of the tumor-cell genome may also contain many aberrantly expressed, non-mutated tumor-associated antigens (TAAs) capable of eliciting productive anti-tumor immune responses. Whole-exome sequencing (WES) and/or RNA sequencing (RNA-Seq) of cancer tissue, combined with several layers of bioinformatic analysis is commonly used to predict possible neoepitopes present in clinical samples. At the ImmunoSurgery Unit of the Champalimaud Centre for the Unknown (CCU), a pipeline combining several tools is used for predicting private mutations from WES and RNA-Seq data followed by the construction of synthetic peptides tailored for immunological response assessment reflecting the patient’s tumor mutations, guided by MHC typing. Subsequent immunoassays allow the detection of differential IFN-γ production patterns associated with (intra-tumoral) spatiotemporal differences in TIL or peripheral T-cells versus TIL. These bioinformatics tools, in addition to histopathological assessment, immunological readouts from functional bioassays and deep T-cell ‘adaptome’ analyses, are expected to advance discovery and development of next-generation personalized precision medicine strategies to improve clinical outcomes in cancer in the context of i) anti-tumor vaccination strategies, ii) gauging mutation-reactive T-cell responses in biological therapies and iii) expansion of tumor-reactive T-cells for the cellular treatment of patients with cancer.
Collapse
Affiliation(s)
- Eric de Sousa
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana R Lérias
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Antonio Beltran
- Department of Pathology, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Carolina Condeço
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Jéssica Kamiki
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Nuno Figueiredo
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | - Carlos Carvalho
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Zhe Wang
- Jiangsu Industrial Technology Research Institute (JITRI), Applied Adaptome Immunology Institute, Nanjing, China
| | - Dário Ligeiro
- Lisbon Centre for Blood and Transplantation, Instituto Português do Sangue e Transplantação (IPST), Lisbon, Portugal
| | - Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,I Medical Clinic, Johannes Gutenberg University of Mainz, Mainz, Germany
| |
Collapse
|
46
|
Chen ELY, Lee CR, Thompson PK, Wiest DL, Anderson MK, Zúñiga-Pflücker JC. Ontogenic timing, T cell receptor signal strength, and Notch signaling direct γδ T cell functional differentiation in vivo. Cell Rep 2021; 35:109227. [PMID: 34107257 PMCID: PMC8256923 DOI: 10.1016/j.celrep.2021.109227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 02/20/2021] [Accepted: 05/14/2021] [Indexed: 12/29/2022] Open
Abstract
γδ T cells form an integral arm of the immune system and are critical during protective and destructive immunity. However, how γδ T cells are functionally programmed in vivo remains unclear. Here, we employ RBPJ-inducible and KN6-transgenic mice to assess the roles of ontogenic timing, T cell receptor (TCR) signal strength, and Notch signaling. We find skewing of Vγ1+ cells toward the PLZF+Lin28b+ lineage at the fetal stage. Generation of interleukin-17 (IL-17)-producing γδ T cells is favored during, although not exclusive to, the fetal stage. Surprisingly, Notch signaling is dispensable for peripheral γδ T cell IL-17 production. Strong TCR signals, together with Notch, promote IL-4 differentiation. Conversely, less strong TCR signals promote Notch-independent IL-17 differentiation. Single-cell transcriptomic analysis reveals differential programming instilled by TCR signal strength and Notch for specific subsets. Thus, our results precisely define the roles of ontogenic timing, TCR signal strength, and Notch signaling in γδ T cell functional programming in vivo.
Collapse
Affiliation(s)
- Edward L Y Chen
- Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | | | - David L Wiest
- Blood Cell Development and Function, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Michele K Anderson
- Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
47
|
The Association of Psoriasis and Obesity: Focusing on IL-17A-Related Immunological Mechanisms. INTERNATIONAL JOURNAL OF DERMATOLOGY AND VENEREOLOGY 2021. [DOI: 10.1097/jd9.0000000000000155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
48
|
Torres-Cabala CA, Huen A, Iyer SP, Miranda RN. Gamma/Delta Phenotype in Primary Cutaneous T-cell Lymphomas and Lymphoid Proliferations: Challenges for Diagnosis and Classification. Surg Pathol Clin 2021; 14:177-194. [PMID: 34023099 DOI: 10.1016/j.path.2021.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Primary cutaneous T-cell lymphomas pose a diagnostic challenge for dermatopathologists, hematopathologists, and general surgical pathologists. Recognition of gamma/delta phenotype in cutaneous T proliferations has been enhanced by the availability of antibodies against TCRgamma and delta for immunohistochemistry. Thus, reporting gamma/delta phenotype in a cutaneous T-cell lymphoid proliferation may indicate a significant change in therapy and a challenge for dermatologists and oncologists who treat these patients. Herein, we discuss primary cutaneous gamma/delta T-cell lymphoma, its differential diagnosis, and other skin lymphoid proliferations that may show gamma/delta phenotype. Awareness of the occurrence of gamma/delta phenotype in both T-cell lymphomas and benign lymphoid proliferations involving skin is crucial for a better interpretation of histopathologic findings. Integration of clinical presentation, morphology, immunoprofile, and molecular findings is key for a correct diagnosis and appropriate therapy of lesions displaying gamma/delta T-cell phenotype.
Collapse
Affiliation(s)
- Carlos A Torres-Cabala
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 85, Houston, TX 77030, USA; Department of Dermatology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1452, Houston, TX 77030, USA.
| | - Auris Huen
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1452, Houston, TX 77030, USA
| | - Swaminathan P Iyer
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, TX 77030, USA
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 72, Houston, TX 77030, USA
| |
Collapse
|
49
|
Milling S, Siebert S. T cells and cytokines in inflamed psoriatic skin. Who's in charge? Immunology 2021; 160:311-312. [PMID: 32686183 DOI: 10.1111/imm.13237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inflammation in psoriasis is driven through the IL-23/IL-17 pathway. Monoclonal antibodies targeting cytokines in the pathway have proven highly effective and are widely used in clinical practice. There is still much to learn, however, about how these pathogenic responses are controlled, particularly with respect to the highly immunologically active molecules produced by the inflamed skin tissue itself. These tissue-derived molecules, which include IL-33, play important roles in modulating chronic inflammation that we are only beginning to understand. Here we highlight new research indicating a role for IL-33 in modulating the inflammatory Th17 response in psoriasis, which may provide avenues for developing new psoriasis treatments.
Collapse
Affiliation(s)
- Simon Milling
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
50
|
Moon J, Lee SY, Choi JW, Lee AR, Yoo JH, Moon SJ, Park SH, Cho ML. Metformin ameliorates scleroderma via inhibiting Th17 cells and reducing mTOR-STAT3 signaling in skin fibroblasts. J Transl Med 2021; 19:192. [PMID: 33947424 PMCID: PMC8097822 DOI: 10.1186/s12967-021-02860-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
Scleroderma is an autoimmune disease that causes dermal fibrosis. It occurs when collagen accumulates in tissue as a result of persistent inflammation. Th17 cells and pro-inflammatory cytokines such as IL-1β, IL-6, IL-17, and TNF-α play important roles in the pathogenesis of scleroderma. Because metformin, a medication used to treat diabetes, has effective immunoregulatory functions, we investigated its therapeutic function in scleroderma. Mice in a model of bleomycin-induced scleroderma were treated with metformin for 2 weeks. Histological assessment demonstrated protective effects of metformin against scleroderma. Metformin decreased the expression of pro-inflammatory factors in dermal tissue and lymphocytes. It also decreased mRNA expression of pro-inflammatory cytokines (IL-1β, IL-6, IL-17, and TNF-α) and fibrosis-inducing molecules both in vivo and in vitro. These results suggest that metformin treatment has anti-inflammatory effects on lymphocytes via the inhibition of IL-17 and cytokines related to Th17 differentiation, such as IL-1β, IL-6, and TNF-α. To investigate how metformin modulates the inflammatory process in skin fibroblasts, we measured mTOR-STAT3 signaling in skin fibroblasts and found that phosphorylated mTOR and phosphorylated STAT3 protein expression were decreased by metformin treatment. These results suggest that metformin has potential to treat scleroderma by inhibiting pro-inflammatory cytokines and anti-inflammatory activity mediated by mTOR-STAT3 signaling.
Collapse
Affiliation(s)
- Jeonghyeon Moon
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Seon-Yeong Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Jeong Won Choi
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - A Ram Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Jin Hee Yoo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Su-Jin Moon
- Divison of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, 11765, Republic of Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Mi-La Cho
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea. .,Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|