1
|
Akenroye A, Boyce JA, Kita H. Targeting alarmins in asthma: From bench to clinic. J Allergy Clin Immunol 2025; 155:1133-1148. [PMID: 39855362 DOI: 10.1016/j.jaci.2025.01.017] [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: 09/20/2024] [Revised: 12/24/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Over the past 2 decades, mechanistic studies of allergic and type 2 (T2)-mediated airway inflammation have led to multiple approved therapies for the treatment of moderate-to-severe asthma. The approval and availability of these monoclonal antibodies targeting IgE, a T2 cytokine (IL-5) and/or cytokine receptors (IL-5Rα, IL-4Rα) has been central to the progresses made in the management of moderate-to-severe asthma over this period. However, there are persistent gaps in clinician's ability to provide precise care, given that many patients with T2-high asthma do not respond to IgE- or T2 cytokine-targeting therapies and that patients with T2-low asthma have few therapeutic options. The new frontier of precision medicine in asthma, as well as in other allergic diseases, includes the targeting of epithelium-derived cytokines known as alarmins, including thymic stromal lymphopoietin, IL-25, IL-33, and their receptors. The effects of these alarmins, which can act upstream of immune cells, involve both the innate and adaptive systems and hold potential for the treatment of both T2-high and -low disease. Tezepelumab, an anti-thymic stromal lymphopoietin antibody, has already been approved for the treatment of severe asthma. In this review, we discuss our current understanding of alarmin biology with a primary focus on allergic airway diseases. We link the mechanistic corollaries to the clinical implications and advances in drug development targeting alarmins, with a particular focus on currently approved treatments, those under study, and future potential targets in alarmin signaling pathways.
Collapse
Affiliation(s)
- Ayobami Akenroye
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| | - Joshua A Boyce
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, the Department of Medicine, and the Department of Immunology, Mayo Clinic Arizona, Scottsdale, Ariz; Department of Immunology, Mayo Clinic Rochester, Rochester, Minn
| |
Collapse
|
2
|
Turkbey M, Karaguzel D, Uzunkaya AD, Aracagok YD, Karaaslan C. The immune response of upper and lower airway epithelial cells to Aspergillus fumigatus and Candida albicans-derived β-glucan in Th17 type cytokine environment. Arch Microbiol 2025; 207:70. [PMID: 39992431 DOI: 10.1007/s00203-025-04266-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/25/2025]
Abstract
The fungal cell wall component β-glucan activates inflammation via the Dectin-1 receptor and IL-17 coordinates the antifungal immunity. However, the molecular crosstalk between IL-17, Dectin-1, and β-glucan in epithelial cells and fungal immunity remains unclear. We investigated the impact of A.fumigatus-derived β-glucan (AFBG) and C.albicans-derived β-glucan (CABG) on Dectin-1 and cytokines in nasal epithelial cells (NECs) and bronchial epithelial cells (BECs) in the presence of IL-17. CABG reduced BEC viability more than AFBG despite similar Dectin-1 expression. IL-17 reduced β-glucan-dependent Dectin-1 expression in NECs but increased it in BECs after 12 h. AFBG synergized with IL-17, enhancing pro-inflammatory cytokines and chemokine expressions. IL-6 and IL-8 production increased in the presence of IL-17. Th17 cytokine influenced the Dectin-1 response to fungal β-glucan in NECs and BECs, impacting the initiation and nature of epithelial cell reactions to AFBG and CABG. Uncovering the molecular mechanisms of fungal β-glucans in the respiratory tract could lead to novel strategies for preventing fungal diseases.
Collapse
Affiliation(s)
- Murat Turkbey
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye
| | - Dilara Karaguzel
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye
| | - Ali Doruk Uzunkaya
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye
| | - Yusuf Doruk Aracagok
- Department of Biology, Biotechnology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye
| | - Cagatay Karaaslan
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye.
| |
Collapse
|
3
|
Hou C, Yu Z, Song Q, Chu X, Wei G, Zhu J, Yang L, Zhao Y, Zhang P, Li Q. Identification of the key role of IL-17RB in the treatment of osteoarthritis with Shaoyao Gancao decoction: Verification based on RNA-seq and bioinformatics analysis. PLoS One 2025; 20:e0315913. [PMID: 39964979 PMCID: PMC11835320 DOI: 10.1371/journal.pone.0315913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/01/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Shaoyao Gancao Decoction (SGD) is a classic and representative oral administration of traditional Chinese medicine formula. It is composed of two Chinese herbal medicines, Paeoniae Radix Alba [Paeonia lactiflora Pall] and Glycyrrhizae Radix et Rhizoma. The clinical study found SGD could effectively reduce clinical symptoms and improve the level of inflammation in osteoarthritis (OA) patients. PURPOSE The aim of this study is to identify the efficacy and molecular mechanism of SGD in the treatment of OA, and find the new therapeutic target through RNA sequencing (RNA-Seq) to provide theoretical support for its clinical application. METHODS Destabilization of the medial meniscus (DMM) OA rat model was established in vivo. Hematoxylineosin staining, safranin O/fast green staining and immunohistochemistry were used to observe changes of cartilage Histology and extracellular matrix (ECM) of cartilage cells. In vitro, the chondrocyte-like cells were derived from ATDC5 cells and induced by interleukin-1 beta to establish the model. The medial meniscotibial ligament (MTT) test was used to identify the effects of SGD on chondrocyte-like cell proliferation, and immunocytochemistry was used to assess changes in chondrocyte ECM. The differentially expressed genes (DEGs) were obtained by RNA-Seq. Meanwhile, the core targets were found through bioinformatics analysis, and then verified by qRT-PCR and Western Blotting. The inflammatory factors IL-1β, IL-6 and TNF-α were detected by ELISA. RESULTS SGD could alleviate cartilage degeneration, and reduce ECM degradation in OA by upregulating COL2A1 and downregulating MMP-13. 120 key targets were screened from DEGs by RNA-Seq. Based on further bioinformatics analysis, interleukin 17 receptor B (IL-17RB), interleukin 23 receptor and growth differentiation factor 5 were finally selected as core targets. IL-17RB has rarely been reported in previous studies about OA, and worthy of further study. Subsequently, it was found that the gene and protein expressions of IL-17RB were significantly reversed in model group after SGD treatment. Moreover, SGD could inhibit the release of inflammatory factors by mediating IL-17RB in OA. CONCLUSIONS SGD reduced the release of inflammatory factors IL-1β, IL-6 and TNF-α, upregulated COL2A1 and downregulated MMP-13 to alleviate degradation of ECM, and reduced the cartilage degeneration and progression of OA by reducing IL-17RB in articular cartilage.
Collapse
Affiliation(s)
- Chengzhi Hou
- Pharmacological Laboratory of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Sports Medicine Department 2, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhangjingze Yu
- Pharmacological Laboratory of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Education, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qinghui Song
- Pharmacological Laboratory of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuelei Chu
- Department of Education, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guangcheng Wei
- Department of Education, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jia Zhu
- Pharmacological Laboratory of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Pharmacological Laboratory of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Zhao
- Sports Medicine Department 2, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Zhang
- Pharmacological Laboratory of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiuyue Li
- Pharmacological Laboratory of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Huynh TNM, Yamazaki F, Konrad RJ, Nishikawa Y, Tanaka A, Son Y, Ozaki Y, Takehana K, Tanizaki H. Circulating CD31 and resistin levels reflect different stages of coronary atherosclerosis in patients with psoriasis. J Dermatol 2025; 52:67-78. [PMID: 39436026 DOI: 10.1111/1346-8138.17450] [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: 03/25/2024] [Revised: 07/24/2024] [Accepted: 08/18/2024] [Indexed: 10/23/2024]
Abstract
Psoriasis is a skin disease with a complicated pathophysiology that includes an extensive inflammatory cytokine network. Nevertheless, the relationship between psoriasis severity, cytokine levels, and coronary artery atherosclerosis remains poorly understood. Our aim was to find serum markers as potential candidates for cardiovascular disease (CVD) risk monitoring in patients with psoriasis. Therefore, we examined coronary artery atherosclerosis via coronary computed tomography angiography (CCTA), serum cytokine levels via multiple immunoassays, and the patients' psoriasis state. Our findings reveal for the first time that the mainstream psoriasis cytokines interleukin 17A (IL-17A), IL-19, and IL-36 in the sera of Japanese patients with psoriasis showed a linear regression association with the Psoriasis Area and Severity Index score. Furthermore, the serum level of IL-19 was remarkably correlated to Th2-related serum cytokines such as IL-4 and IL-17E. When we investigated potential markers to monitor CVD in patients with psoriasis, circulating cluster of differentiation 31 (CD31) and resistin, but not psoriasis-related cytokines, were expressed differently at each stage of coronary atherosclerosis by CCTA. CD31 and resistin levels rose dramatically in individuals with psoriasis vulgaris (PV) and noncalcified atherosclerosis. In contrast, CD31 was negatively correlated with the coronary artery calcification score (CACS) in patients with PV, whereas resistin was inversely correlated with CACS in patients with psoriatic arthritis. In conclusion, the axis of IL-17A, IL-19, and IL-36 remains associated with the severity of psoriasis during the chronic phase of the disease, regardless of the application of topical or systemic treatment. Monitoring the levels of these cytokines can accurately determine the severity of skin inflammation. Resistin and CD31 are linked to coronary artery lesions and might be good candidates for tracking the progression of coronary atherosclerosis in patients with psoriasis.
Collapse
Affiliation(s)
| | - Fumikazu Yamazaki
- Psoriasis Center, Kansai Medical University, Osaka, Japan
- Department of Dermatology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Robert J Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Yumiko Nishikawa
- Japan Drug Development and Medical Affairs, Eli Lilly Japan K.K, Hyogo, Japan
| | - Akihiro Tanaka
- Psoriasis Center, Kansai Medical University, Osaka, Japan
- Division of Rheumatology, Department of Medicine I, Kansai Medical University, Osaka, Japan
| | - Yonsu Son
- Psoriasis Center, Kansai Medical University, Osaka, Japan
- Division of Rheumatology, Department of Medicine I, Kansai Medical University, Osaka, Japan
| | - Yoshio Ozaki
- Psoriasis Center, Kansai Medical University, Osaka, Japan
- Division of Rheumatology, Department of Medicine I, Kansai Medical University, Osaka, Japan
| | - Kazuya Takehana
- Psoriasis Center, Kansai Medical University, Osaka, Japan
- Division of Cardiology, Department of Medicine II, Kansai Medical University, Osaka, Japan
| | - Hideaki Tanizaki
- Department of Dermatology, Kansai Medical University, Osaka, Japan
- Psoriasis Center, Kansai Medical University, Osaka, Japan
| |
Collapse
|
5
|
Jafari N, Abediankenari S. Role of microRNAs in immunoregulatory functions of epithelial cells. BMC Immunol 2024; 25:84. [PMID: 39707170 PMCID: PMC11662810 DOI: 10.1186/s12865-024-00675-3] [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: 07/16/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
Epithelial cells (ECs) provide the first line of defense against microbial threats and environmental challenges. They participate in the host's immune responses via the expression and secretion of various immune-related molecules such as cytokines and chemokines, as well as interaction with immune cells. A growing body of evidence suggests that the dysregulated function of ECs can be involved in the pathophysiology of a broad range of infectious, autoimmune, and inflammatory diseases, including inflammatory bowel disease (IBD), asthma, multiple sclerosis, and rheumatoid arthritis. To maintain a substantial immunoregulatory function of ECs, precise expression of different molecules and their regulatory effects are indispensable. MicroRNAs (miRNAs, miRs) are small non-coding RNAs that regulate gene expression commonly at post-transcriptional level through degradation of target messenger RNAs (mRNAs) or suppression of protein translation. MiRNAs implicate as critical regulators in many cellular processes, including apoptosis, growth, differentiation, and immune response. Due to the crucial roles of miRNAs in such a vast range of biological processes, they have become the spotlight of biological research for more than two decades, but we are still at the beginning stages of the use of miRNA-based therapies in the improvement of human health. Hence, in the present paper, attempts are made to provide a comprehensive overview with regard to the roles of miRNAs in the immunoregulatory functions of ECs. A better understanding of the molecular mechanisms through which immunoregulatory properties of ECs are manifested, could aid the development of efficient strategies to prevent and treat multiple human diseases.
Collapse
Affiliation(s)
- Narjes Jafari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid Abediankenari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
6
|
Ullah MN, Rowan NR, Lane AP. Neuroimmune interactions in the olfactory epithelium: maintaining a sensory organ at an immune barrier interface. Trends Immunol 2024; 45:987-1000. [PMID: 39550314 PMCID: PMC11624989 DOI: 10.1016/j.it.2024.10.005] [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: 09/06/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/18/2024]
Abstract
While primarily a sensory organ, the mammalian olfactory epithelium (OE) also plays a critical role as an immune barrier. Mechanisms governing interactions between the immune system and this specialized chemosensory tissue are gaining interest, in part sparked by the COVID-19 pandemic. Regulated inflammation is intrinsic to normal mucosal healing and homeostasis, but prolonged OE inflammation is associated with persistent loss of smell, belying the intertwining of local mucosal immunology and olfactory function. Evidence supports bidirectional communication between OE cells and the immune system in health and disease. Recent investigations suggest that neuro-immune cross-talk modulates olfactory stem cell behavior and neuronal regeneration dynamics, prioritizing the epithelial-like non-neuronal framework with immune barrier function at the expense of the neurosensory organ in chronic inflammation.
Collapse
Affiliation(s)
- Mohammed N Ullah
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas R Rowan
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew P Lane
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
7
|
Li L, Zhang Y, Wang Z, Chen X, Fang M. Glycyrrhizin attenuates renal inflammation in a mouse Con A-hepatitis model via the IL-25/M2 axis. Ren Fail 2024; 46:2356023. [PMID: 38785317 PMCID: PMC11133957 DOI: 10.1080/0886022x.2024.2356023] [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: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
Glycyrrhizin (GL) has immunoregulatory effects on various inflammatory diseases including hepatitis and nephritis. However, the mechanisms underlying the anti-inflammatory effect of GL on renal inflammation are not fully understood. Hepatorenal syndrome (HRS) is a functional acute renal impairment that occurs in severe liver disease, and we found that kidney injury also occurs in Con A-induced experimental hepatitis in mice. We previously found that GL can alleviate Con A-induced hepatitis by regulating the expression of IL-25 in the liver. We wanted to investigate whether GL can alleviate Con A-induced nephritis by regulating IL-25. IL-25 regulates inflammation by modulating type 2 immune responses, but the mechanism by which IL-25 affects kidney disease remains unclear. In this study, we found that the administration of GL enhanced the expression of IL-25 in renal tissues; the latter promoted the generation of type 2 macrophages (M2), which inhibited inflammation in the kidney caused by Con A challenge. IL-25 promoted the secretion of the inhibitory cytokine IL-10 by macrophages but inhibited the expression of the inflammatory cytokine IL-1β by macrophages. Moreover, IL-25 downregulated the Con A-mediated expression of Toll-like receptor (TLR) 4 on macrophages. By comparing the roles of TLR2 and TLR4, we found that TLR4 is required for the immunoregulatory effect of IL-25 on macrophages. Our data revealed that GL has anti-inflammatory effects on Con A-induced kidney injury and that the GL/IL-25/M2 axis participates in the anti-inflammatory process. This study suggested that GL is a potential therapeutic for protecting against acute kidney injury.
Collapse
Affiliation(s)
- Lingyun Li
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, China
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyue Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongyan Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyu Chen
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China
| | - Min Fang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Appleton LK, Hanania NA, Adrish M. Personalized COPD Care: The Future of Precision-Based Therapies. J Clin Med 2024; 13:6339. [PMID: 39518477 PMCID: PMC11546703 DOI: 10.3390/jcm13216339] [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: 09/05/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive respiratory illness characterized by long-standing respiratory symptoms and airflow limitation. It is a major contributor to respiratory disease-related deaths and currently ranked as the sixth leading cause of mortality in the United States. Approved pharmacological therapies for the stable disease primarily consist of inhaled short and long-acting bronchodilators, inhaled corticosteroids, azithromycin, and roflumilast. In recent years, significant progress has been made in the management of COPD through the identification of different COPD phenotypes and endotypes, which allows for a more personalized treatment approach. While earlier studies investigating targeted therapies were less promising, recent data on drugs targeting type 2 inflammatory pathways have shown promising results in carefully selected patients. In this article, we will review the available data on targeted therapies as well as the ongoing clinical studies of novel targeted therapies for COPD. Understanding and implementing these advancements hold promise for improving outcomes and quality of life for individuals living with COPD.
Collapse
Affiliation(s)
| | | | - Muhammad Adrish
- Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, 1504 Taub Loop, Houston, TX 77030, USA (N.A.H.)
| |
Collapse
|
9
|
Kao CJ, Charmsaz S, Alden SL, Brancati M, Li HL, Balaji A, Munjal K, Howe K, Mitchell S, Leatherman J, Griffin E, Nakazawa M, Tsai HL, Danilova L, Thoburn C, Gizzi J, Gross NE, Hernandez A, Coyne EM, Shin SM, Babu JS, Apostol GW, Durham J, Christmas BJ, Konig MF, Lipson EJ, Naidoo J, Cappelli LC, Pabani A, Ged Y, Baretti M, Brahmer J, Hoffman-Censits J, Seiwert TY, Garonce-Hediger R, Guha A, Bansal S, Tang L, Jaffee EM, Chandler GS, Mohindra R, Ho WJ, Yarchoan M. Immune-related events in individuals with solid tumors on immunotherapy associate with Th17 and Th2 signatures. J Clin Invest 2024; 134:e176567. [PMID: 39403935 PMCID: PMC11473156 DOI: 10.1172/jci176567] [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: 11/20/2023] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUNDImmune-related adverse events (irAEs) and their associated morbidity/mortality are a key concern for patients receiving immune checkpoint inhibitors (ICIs). Prospective evaluation of the drivers of irAEs in a diverse pan-tumor cohort is needed to identify patients at greatest risk and to develop rational treatment and interception strategies.METHODSIn an observational study, we prospectively collected blood samples and performed regular clinical evaluations for irAEs in patients receiving ICI therapy as standard of care for solid tumors. We performed in-parallel analysis of cytokines by Luminex immunoassay and circulating immune cells by cytometry by time-of-flight (CyTOF) at baseline and on treatment to investigate mechanisms of irAEs.RESULTSWe enrolled 111 patients, of whom 40.5% developed a symptomatic irAE (grade ≥ 2). Development of a grade ≥ 2 irAE was positively associated with the use of combination ICI and a history of an autoimmune disorder. Early changes in T helper 17 (Th17) (IL-6, IL-17f), type 2 (IL-5, IL-13, IL-25), and type 1 (TNF-α) cytokine signatures and congruent on-treatment expansions of Th17 and Th2 effector memory (Th2EM) T cell populations in peripheral blood were positively associated with the development of grade ≥2 irAEs. IL-6 levels were also associated with inferior cancer-specific survival and overall survival.CONCLUSIONSIn a diverse, prospective pan-tumor cohort, Th17 and Th2 skewing during early ICI treatment was associated with the development of clinically relevant irAEs but not antitumor responses, providing possible targets for monitoring and therapeutic interventions.FUNDINGJohns Hopkins Bloomberg-Kimmel Institute for Cancer Immunotherapy, the NCI SPORE in Gastrointestinal Cancers (P50 CA062924), NCI grant (R50CA243627 to LD), the NIH Center Core Grant (P30 CA006973), Swim Across America (to MY), NIAMS (K23AR075872 to LC), and imCORE-Genentech grant 137515 (to Johns Hopkins Medicine on behalf of MY).
Collapse
Affiliation(s)
- Chester J. Kao
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Soren Charmsaz
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | | | - Madelena Brancati
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Howard L. Li
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aanika Balaji
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kabeer Munjal
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Kathryn Howe
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Sarah Mitchell
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - James Leatherman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Ervin Griffin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Mari Nakazawa
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Hua-Ling Tsai
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ludmila Danilova
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chris Thoburn
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Gizzi
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicole E. Gross
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Alexei Hernandez
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Erin M. Coyne
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Sarah M. Shin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Jayalaxmi Suresh Babu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - George W. Apostol
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Jennifer Durham
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Brian J. Christmas
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Maximilian F. Konig
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Evan J. Lipson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jarushka Naidoo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Beaumont Hospital, Dublin, Ireland
- RCSI University of Health Sciences, Dublin, Ireland
| | - Laura C. Cappelli
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aliyah Pabani
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yasser Ged
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marina Baretti
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julie Brahmer
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jean Hoffman-Censits
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tanguy Y. Seiwert
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Aditi Guha
- Genentech Inc., a member of the imCORE network, South San Francisco, California, USA
| | - Sanjay Bansal
- Genentech Inc., a member of the imCORE network, South San Francisco, California, USA
| | - Laura Tang
- Genentech Inc., a member of the imCORE network, South San Francisco, California, USA
| | - Elizabeth M. Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | - G. Scott Chandler
- F. Hoffmann-La Roche Ltd., a member of the imCORE network, Basel, Switzerland
| | - Rajat Mohindra
- F. Hoffmann-La Roche Ltd., a member of the imCORE network, Basel, Switzerland
| | - Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
| | - Mark Yarchoan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Muñiz P, Martínez-García M, Bailén R, Chicano M, Oarbeascoa G, Triviño JC, de la Iglesia-San Sebastian I, Fernández de Córdoba S, Anguita J, Kwon M, Díez-Martín JL, Olmos PM, Martínez-Laperche C, Buño I. Identification of predictive models including polymorphisms in cytokines genes and clinical variables associated with post-transplant complications after identical HLA-allogeneic stem cell transplantation. Front Immunol 2024; 15:1396284. [PMID: 39247183 PMCID: PMC11377344 DOI: 10.3389/fimmu.2024.1396284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/08/2024] [Indexed: 09/10/2024] Open
Abstract
Backgrounds Although allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative therapy for hematological malignancies, it can be associated with relevant post-transplant complications. Several reports have shown that polymorphisms in immune system genes are correlated with the development of post-transplant complications. Within this context, this work focuses on identifying novel polymorphisms in cytokine genes and developing predictive models to anticipate the risk of developing graft-versus-host disease (GVHD), transplantation-related mortality (TRM), relapse and overall survival (OS). Methods Our group developed a 132-cytokine gene panel which was tested in 90 patients who underwent an HLA-identical sibling-donor allo-HSCT. Bayesian logistic regression (BLR) models were used to select the most relevant variables. Based on the cut-off points selected for each model, patients were classified as being at high or low-risk for each of the post-transplant complications (aGVHD II-IV, aGVHD III-IV, cGVHD, mod-sev cGVHD, TRM, relapse and OS). Results A total of 737 polymorphisms were selected from the custom panel genes. Of these, 41 polymorphisms were included in the predictive models in 30 cytokine genes were selected (17 interleukins and 13 chemokines). Of these polymorphisms, 5 (12.2%) were located in coding regions, and 36 (87.8%) in non-coding regions. All models had a statistical significance of p<0.0001. Conclusion Overall, genomic polymorphisms in cytokine genes make it possible to anticipate the development all complications studied following allo-HSCT and, consequently, to optimize the clinical management of patients.
Collapse
Affiliation(s)
- Paula Muñiz
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
| | - María Martínez-García
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
- Department of Signal Theory and Communications, University Carlos III of Madrid, Madrid, Spain
| | - Rebeca Bailén
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
| | - María Chicano
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
| | - Gillen Oarbeascoa
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
| | | | - Ismael de la Iglesia-San Sebastian
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
| | - Sara Fernández de Córdoba
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
| | - Javier Anguita
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
| | - Mi Kwon
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
| | - José Luis Díez-Martín
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
- Department of Medicine, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Pablo M Olmos
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
- Department of Signal Theory and Communications, University Carlos III of Madrid, Madrid, Spain
| | - Carolina Martínez-Laperche
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
| | - Ismael Buño
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain
- Signal Theory and Communications Department, School of Engineering, Carlos III University, Leganés, Madrid, Spain
- Genomics Unit, Gregorio Marañón General University Hospital, IiSGM, Madrid, Spain
- Department of Cell Biology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
11
|
Nastasa V, Minea B, Pasca AS, Bostanaru-Iliescu AC, Stefan AE, Gologan D, Capota R, Foia LG, Mares M. Long-Term Oral Administration of Hyperimmune Egg-Based IgY-Rich Formulations Induces Mucosal Immune Response and Systemic Increases of Cytokines Involved in Th2- and Th17-Type Immune Responses in C57BL/6 Mice. Int J Mol Sci 2024; 25:8701. [PMID: 39201385 PMCID: PMC11354499 DOI: 10.3390/ijms25168701] [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: 06/12/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Three hyperimmune egg-based formulations rich in immunoglobulin Y (IgY) were orally administered (daily, for up to 90 days) to C57BL/6 mice that were not microbially challenged. The serum levels of 32 cytokines were quantified every 30 days. Histopathology, hematology, and serum biochemistry investigations were also performed. As a sign of increased immune activity, lymphohistiocytic infiltrates were detected in the digestive tract and the liver after 30, 60, and 90 days of treatment. These infiltrates were also present in the lungs after 30 and 60 days, but not at 90 days. Blood analysis indicated systemic inflammation after 30 days of treatment: increases in pro-inflammatory cytokines, glycemia, total serum proteins, ALT, and ALP. After 60 and 90 days of treatment, the analyzed blood parameters showed mixed signs of both increased and decreased inflammation. The increased cytokines, which varied with formulation and time of exposure, indicated a combination of mostly Th17- and Th2-type immune responses. As the mice were healthy and housed in standardized sanitary conditions, and were not microbially challenged, the data were consistent with an interaction of IgY with the gut-associated lymphoid tissue as the main mechanism of action. This interaction generated a local immune response, which subsequently induced a systemic response.
Collapse
Affiliation(s)
- Valentin Nastasa
- Laboratory of Antimicrobial Chemotherapy, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (V.N.); (A.-S.P.); (A.-C.B.-I.); (R.C.); (M.M.)
| | - Bogdan Minea
- Department of Surgery, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, 16 Universitatii Street, 700115 Iasi, Romania;
| | - Aurelian-Sorin Pasca
- Laboratory of Antimicrobial Chemotherapy, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (V.N.); (A.-S.P.); (A.-C.B.-I.); (R.C.); (M.M.)
| | - Andra-Cristina Bostanaru-Iliescu
- Laboratory of Antimicrobial Chemotherapy, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (V.N.); (A.-S.P.); (A.-C.B.-I.); (R.C.); (M.M.)
| | - Alina-Elena Stefan
- Doctoral School, Department of Pathology, Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, 59 Mărăşti Boulevard, 011464 Bucharest, Romania;
- Department of Research and Development, Themis Pathology SRL, 56F 1 Decembrie 1918 Boulevard, 032468 Bucharest, Romania;
| | - Daniela Gologan
- Department of Research and Development, Themis Pathology SRL, 56F 1 Decembrie 1918 Boulevard, 032468 Bucharest, Romania;
- Doctoral School, Department of Organic Chemistry, Faculty of Chemical Engineering and Biotechnologies, Politehnica University, 313 Splaiul Independenţei, 060042 Bucharest, Romania
| | - Robert Capota
- Laboratory of Antimicrobial Chemotherapy, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (V.N.); (A.-S.P.); (A.-C.B.-I.); (R.C.); (M.M.)
| | - Liliana-Georgeta Foia
- Department of Surgery, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, 16 Universitatii Street, 700115 Iasi, Romania;
| | - Mihai Mares
- Laboratory of Antimicrobial Chemotherapy, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (V.N.); (A.-S.P.); (A.-C.B.-I.); (R.C.); (M.M.)
| |
Collapse
|
12
|
He Y, Lin T, Liang R, Xiang Q, Tang T, Ge N, Yue J. Interleukin 25 promotes muscle regeneration in sarcopenia by regulating macrophage-mediated Sonic Hedgehog signaling. Int Immunopharmacol 2024; 139:112662. [PMID: 39038385 DOI: 10.1016/j.intimp.2024.112662] [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: 04/09/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024]
Abstract
OBJECTIVE Sarcopenia manifests as a chronic, low-level inflammation along with multiple inflammatory cells infiltration. Interleukin (IL)-25 can regulate the function of macrophages. However, the specific role and mechanisms through which IL-25 functions in sarcopenia are still not fully understood and require further investigation. METHODS Aged mice were utilized as sarcopenia models and examined the expression of inflammatory factors. To investigate the effects of IL-25 on sarcopenia, the model mice received IL-25 treatment and underwent in vivo adoptive transfer of IL-25-induced macrophages. Meanwhile, RAW264.7 cells, bone marrow-derived macrophages, satellite cells and C2C12 cells were used in vitro. Shh insufficiency was induced through intramuscular administration of SHH-shRNA adenoviruses. Then, various assays including scratch wound, cell counting kit-8 and Transwell assays, as well as histological staining and molecular biological methods, were conducted. RESULTS Aged mice exhibited an accelerated decline in muscle strength and mass, along with an increased muscle lipid droplets and macrophage infiltration, and decreased IL-25 levels compared to the young group. IL-25 therapy and the transfer of IL-25-preconditioned macrophages could improve these conditions by promoting M2 macrophage polarization in vivo as well as in vitro. M2 macrophage conditioned medium enhanced satellite cell proliferation and migration, as well as the vitality, migration, and differentiation of C2C12 cells in vitro. Furthermore, IL-25 enhanced Shh expression in macrophages in vitro, and activated the Shh signaling pathway in muscle tissue of aged mice, which could be suppressed by either the inhibitor cyclopamine or Shh knockdown. Mechanistic studies showed that Shh insufficiency suppressed the activation of Akt/mTOR signaling pathway in muscle tissue of aged mice. CONCLUSION IL-25 promotes the secretion of Shh by M2 macrophages and activates the Shh/Akt/mTOR signaling pathway, which improves symptoms and function in sarcopenia mice. This suggests that IL-25 has potential as a therapeutic agent for treating sarcopenia.
Collapse
Affiliation(s)
- Yan He
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Geriatrics, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Taiping Lin
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Liang
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiao Xiang
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianjiao Tang
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ning Ge
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jirong Yue
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
13
|
Newman M, Connery H, Kannan S, Gautam A, Hammamieh R, Chakraborty N, Boyd J. Fentanyl Overdose Causes Prolonged Cardiopulmonary Dysregulation in Male SKH1 Mice. Pharmaceuticals (Basel) 2024; 17:941. [PMID: 39065791 PMCID: PMC11279777 DOI: 10.3390/ph17070941] [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: 06/18/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Fentanyl overdose is a survivable condition that commonly resolves without chronic overt changes in phenotype. While the acute physiological effects of fentanyl overdose, such as opioid-induced respiratory depression (OIRD) and Wooden Chest Syndrome, represent immediate risks of lethality, little is known about longer-term systemic or organ-level impacts for survivors. In this study, we investigated the effects of a single, bolus fentanyl overdose on components of the cardiopulmonary system up to one week post. SKH1 mice were administered subcutaneous fentanyl at the highest non-lethal dose (62 mg/kg), LD10 (110 mg/kg), or LD50 (135 mg/kg), before euthanasia at 40 min, 6 h, 24 h, or 7 d post-exposure. The cerebral cortex, heart, lungs, and plasma were assayed using an immune monitoring 48-plex panel. The results showed significantly dysregulated cytokine, chemokine, and growth factor concentrations compared to time-matched controls, principally in hearts, then lungs and plasma to a lesser extent, for the length of the study, with the cortex largely unaffected. Major significant analytes contributing to variance included eotaxin-1, IL-33, and betacellulin, which were generally downregulated across time. The results of this study suggest that cardiopulmonary toxicity may persist from a single fentanyl overdose and have wide implications for the endurance of the expanding population of survivors.
Collapse
Affiliation(s)
- Mackenzie Newman
- Department of Orthopaedic Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA;
- Department of Physiology, Pharmacology and Toxicology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA
| | - Heather Connery
- Department of Physiology, Pharmacology and Toxicology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA
| | - Swapna Kannan
- Walter Reed Army Institute of Research, Silver Spring, MD 20907, USA
| | - Aarti Gautam
- Walter Reed Army Institute of Research, Silver Spring, MD 20907, USA
| | - Rasha Hammamieh
- Walter Reed Army Institute of Research, Silver Spring, MD 20907, USA
| | | | - Jonathan Boyd
- Department of Orthopaedic Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA;
- Department of Physiology, Pharmacology and Toxicology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
14
|
Lam LH, Li W, Wu WC, Chow KC, Au WYD, Xu G, Wang G, Qian X, Ng KM, Nawabjan SA, Gandhi Sethuraman MI, Choi WY, Chow BKC, Leung SO, Hui CW. SM17, a new IL-17RB-targeting antibody, ameliorates disease progression in a mouse model of atopic dermatitis. Allergy 2024; 79:1625-1628. [PMID: 38590291 DOI: 10.1111/all.16120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/10/2024]
Affiliation(s)
| | - Weimin Li
- SinoMab BioScience Limited, Hong Kong, China
| | | | | | | | - Guolin Xu
- MediNexus Pharma (Suzhou) Limited, Suzhou, Jiangsu, China
| | - Ge Wang
- MediNexus Pharma (Suzhou) Limited, Suzhou, Jiangsu, China
| | - Xuchen Qian
- MediNexus Pharma (Suzhou) Limited, Suzhou, Jiangsu, China
| | - Ka Man Ng
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | | | | | - Wai Yan Choi
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
15
|
Bagnasco D, De Ferrari L, Bondi B, Candeliere MG, Mincarini M, Riccio AM, Braido F. Thymic Stromal Lymphopoietin and Tezepelumab in Airway Diseases: From Physiological Role to Target Therapy. Int J Mol Sci 2024; 25:5972. [PMID: 38892164 PMCID: PMC11172531 DOI: 10.3390/ijms25115972] [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: 02/20/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Thymic stromal lymphopoietin (TSLP), is a protein belonging to a class of epithelial cytokines commonly called alarmins, which also includes IL-25 and IL-33. Functionally, TSLP is a key player in the immune response to environmental insults, initiating a number of downstream inflammatory pathways. TSLP performs its role by binding to a high-affinity heteromeric complex composed of the thymic stromal lymphopoietin receptor (TSLPR) chain and IL-7Rα. In recent years, the important role of proinflammatory cytokines in the etiopathogenesis of various chronic diseases such as asthma, chronic rhinosinusitis with nasal polyposis (CRSwNP), chronic obstructive pulmonary diseases (COPDs), and chronic spontaneous urticaria has been studied. Although alarmins have been found to be mainly implicated in the mechanisms of type 2 inflammation, studies on monoclonal antibodies against TSLP demonstrate partial efficacy even in patients whose inflammation is not definable as T2 and the so-called low T2. Tezepelumab is a human anti-TSLP antibody that prevents TSLP-TSLPR interactions. Several clinical trials are evaluating the safety and efficacy of Tezepelumab in various inflammatory disorders. In this review, we will highlight major recent advances in understanding the functional role of TSLP, its involvement in Th2-related diseases, and its suitability as a target for biological therapies.
Collapse
Affiliation(s)
- Diego Bagnasco
- Allergy and Respiratory Diseases, IRCCS Policlinico San Martino, University of Genoa, 16132 Genoa, Italy; (L.D.F.); (B.B.); (M.G.C.); (M.M.); (A.M.R.); (F.B.)
- Department of Internal Medicine (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Laura De Ferrari
- Allergy and Respiratory Diseases, IRCCS Policlinico San Martino, University of Genoa, 16132 Genoa, Italy; (L.D.F.); (B.B.); (M.G.C.); (M.M.); (A.M.R.); (F.B.)
- Department of Internal Medicine (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Benedetta Bondi
- Allergy and Respiratory Diseases, IRCCS Policlinico San Martino, University of Genoa, 16132 Genoa, Italy; (L.D.F.); (B.B.); (M.G.C.); (M.M.); (A.M.R.); (F.B.)
- Department of Internal Medicine (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Maria Giulia Candeliere
- Allergy and Respiratory Diseases, IRCCS Policlinico San Martino, University of Genoa, 16132 Genoa, Italy; (L.D.F.); (B.B.); (M.G.C.); (M.M.); (A.M.R.); (F.B.)
- Department of Internal Medicine (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Marcello Mincarini
- Allergy and Respiratory Diseases, IRCCS Policlinico San Martino, University of Genoa, 16132 Genoa, Italy; (L.D.F.); (B.B.); (M.G.C.); (M.M.); (A.M.R.); (F.B.)
- Department of Internal Medicine (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Anna Maria Riccio
- Allergy and Respiratory Diseases, IRCCS Policlinico San Martino, University of Genoa, 16132 Genoa, Italy; (L.D.F.); (B.B.); (M.G.C.); (M.M.); (A.M.R.); (F.B.)
- Department of Internal Medicine (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Fulvio Braido
- Allergy and Respiratory Diseases, IRCCS Policlinico San Martino, University of Genoa, 16132 Genoa, Italy; (L.D.F.); (B.B.); (M.G.C.); (M.M.); (A.M.R.); (F.B.)
- Department of Internal Medicine (DIMI), University of Genoa, 16132 Genoa, Italy
| |
Collapse
|
16
|
Ashraf MI, Mengwasser J, Reutzel-Selke A, Polenz D, Führer K, Lippert S, Tang P, Michaelis E, Catar R, Pratschke J, Witzel C, Sauer IM, Tullius SG, Kern B. Depletion of donor dendritic cells ameliorates immunogenicity of both skin and hind limb transplants. Front Immunol 2024; 15:1395945. [PMID: 38799435 PMCID: PMC11116604 DOI: 10.3389/fimmu.2024.1395945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Acute cellular rejection remains a significant obstacle affecting successful outcomes of organ transplantation including vascularized composite tissue allografts (VCA). Donor antigen presenting cells (APCs), particularly dendritic cells (DCs), orchestrate early alloimmune responses by activating recipient effector T cells. Employing a targeted approach, we investigated the impact of donor-derived conventional DCs (cDCs) and APCs on the immunogenicity of skin and skin-containing VCA grafts, using mouse models of skin and hind limb transplantation. By post-transplantation day 6, skin grafts demonstrated severe rejections, characterized by predominance of recipient CD4 T cells. In contrast, hind limb grafts showed moderate rejection, primarily infiltrated by CD8 T cells. Notably, the skin component exhibited heightened immunogenicity when compared to the entire VCA, evidenced by increased frequencies of pan (CD11b-CD11c+), mature (CD11b-CD11c+MHCII+) and active (CD11b-CD11c+CD40+) DCs and cDC2 subset (CD11b+CD11c+ MHCII+) in the lymphoid tissues and the blood of skin transplant recipients. While donor depletion of cDC and APC reduced frequencies, maturation and activation of DCs in all analyzed tissues of skin transplant recipients, reduction in DC activities was only observed in the spleen of hind limb recipients. Donor cDC and APC depletion did not impact all lymphocyte compartments but significantly affected CD8 T cells and activated CD4 T in lymph nodes of skin recipients. Moreover, both donor APC and cDC depletion attenuated the Th17 immune response, evident by significantly reduced Th17 (CD4+IL-17+) cells in the spleen of skin recipients and reduced levels of IL-17E and lymphotoxin-α in the serum samples of both skin and hind limb recipients. In conclusion, our findings underscore the highly immunogenic nature of skin component in VCA. The depletion of donor APCs and cDCs mitigates the immunogenicity of skin grafts while exerting minimal impact on VCA.
Collapse
Affiliation(s)
- Muhammad Imtiaz Ashraf
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Joerg Mengwasser
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Anja Reutzel-Selke
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Dietrich Polenz
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Kirsten Führer
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Steffen Lippert
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Peter Tang
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Edward Michaelis
- Department of Pathology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy, Berlin, Germany
| | - Johann Pratschke
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Christian Witzel
- Department of Plastic Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Igor M. Sauer
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Stefan G. Tullius
- Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Einstein Berlin Institute of Health Visiting Fellow, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Barbara Kern
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Plastic Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Biomedical Innovation Academy, Berlin Institute of Health (BIH) Charité Clinician Scientist Program, Berlin, Germany
| |
Collapse
|
17
|
Osman HC, Moreno R, Rose D, Rowland ME, Ciernia AV, Ashwood P. Impact of maternal immune activation and sex on placental and fetal brain cytokine and gene expression profiles in a preclinical model of neurodevelopmental disorders. J Neuroinflammation 2024; 21:118. [PMID: 38715090 PMCID: PMC11077729 DOI: 10.1186/s12974-024-03106-7] [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: 12/21/2023] [Accepted: 04/21/2024] [Indexed: 05/12/2024] Open
Abstract
Maternal inflammation during gestation is associated with a later diagnosis of neurodevelopmental disorders including autism spectrum disorder (ASD). However, the specific impact of maternal immune activation (MIA) on placental and fetal brain development remains insufficiently understood. This study aimed to investigate the effects of MIA by analyzing placental and brain tissues obtained from the offspring of pregnant C57BL/6 dams exposed to polyinosinic: polycytidylic acid (poly I: C) on embryonic day 12.5. Cytokine and mRNA content in the placenta and brain tissues were assessed using multiplex cytokine assays and bulk-RNA sequencing on embryonic day 17.5. In the placenta, male MIA offspring exhibited higher levels of GM-CSF, IL-6, TNFα, and LT-α, but there were no differences in female MIA offspring. Furthermore, differentially expressed genes (DEG) in the placental tissues of MIA offspring were found to be enriched in processes related to synaptic vesicles and neuronal development. Placental mRNA from male and female MIA offspring were both enriched in synaptic and neuronal development terms, whereas females were also enriched for terms related to excitatory and inhibitory signaling. In the fetal brain of MIA offspring, increased levels of IL-28B and IL-25 were observed with male MIA offspring and increased levels of LT-α were observed in the female offspring. Notably, we identified few stable MIA fetal brain DEG, with no male specific difference whereas females had DEG related to immune cytokine signaling. Overall, these findings support the hypothesis that MIA contributes to the sex- specific abnormalities observed in ASD, possibly through altered neuron developed from exposure to inflammatory cytokines. Future research should aim to investigate how interactions between the placenta and fetal brain contribute to altered neuronal development in the context of MIA.
Collapse
Affiliation(s)
- Hadley C Osman
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA
| | - Rachel Moreno
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA
| | - Destanie Rose
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA
| | - Megan E Rowland
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Annie Vogel Ciernia
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA.
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA.
| |
Collapse
|
18
|
Polverino F, Sin DD. Type 2 airway inflammation in COPD. Eur Respir J 2024; 63:2400150. [PMID: 38485148 DOI: 10.1183/13993003.00150-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/07/2024] [Indexed: 05/30/2024]
Abstract
Globally, nearly 400 million persons have COPD, and COPD is one of the leading causes of hospitalisation and mortality across the world. While it has been long-recognised that COPD is an inflammatory lung disease, dissimilar to asthma, type 2 inflammation was thought to play a minor role. However, recent studies suggest that in approximately one third of patients with COPD, type 2 inflammation may be an important driver of disease and a potential therapeutic target. Importantly, the immune cells and molecules involved in COPD-related type 2 immunity may be significantly different from those observed in severe asthma. Here, we identify the important molecules and effector immune cells involved in type 2 airway inflammation in COPD, discuss the recent therapeutic trial results of biologicals that have targeted these pathways and explore the future of therapeutic development of type 2 immune modulators in COPD.
Collapse
Affiliation(s)
- Francesca Polverino
- Pulmonary and Critical Care Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia Division of Respiratory Medicine, Vancouver, BC, Canada
| |
Collapse
|
19
|
Dong Y, Zhang X, Wang Y. Interleukins in Epilepsy: Friend or Foe. Neurosci Bull 2024; 40:635-657. [PMID: 38265567 PMCID: PMC11127910 DOI: 10.1007/s12264-023-01170-2] [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: 08/19/2023] [Accepted: 10/28/2023] [Indexed: 01/25/2024] Open
Abstract
Epilepsy is a chronic neurological disorder with recurrent unprovoked seizures, affecting ~ 65 million worldwide. Evidence in patients with epilepsy and animal models suggests a contribution of neuroinflammation to epileptogenesis and the development of epilepsy. Interleukins (ILs), as one of the major contributors to neuroinflammation, are intensively studied for their association and modulatory effects on ictogenesis and epileptogenesis. ILs are commonly divided into pro- and anti-inflammatory cytokines and therefore are expected to be pathogenic or neuroprotective in epilepsy. However, both protective and destructive effects have been reported for many ILs. This may be due to the complex nature of ILs, and also possibly due to the different disease courses that those ILs are involved in. In this review, we summarize the contributions of different ILs in those processes and provide a current overview of recent research advances, as well as preclinical and clinical studies targeting ILs in the treatment of epilepsy.
Collapse
Affiliation(s)
- Yuan Dong
- Neuropsychiatry Research Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China.
| | - Xia Zhang
- Neuropsychiatry Research Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China
| | - Ying Wang
- Neuropsychiatry Research Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
20
|
Jou E. Clinical and basic science aspects of innate lymphoid cells as novel immunotherapeutic targets in cancer treatment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 209:1-60. [PMID: 39461748 DOI: 10.1016/bs.pmbts.2024.03.036] [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: 10/29/2024]
Abstract
Immunotherapy has revolutionised cancer treatment over the past decade, demonstrating remarkable efficacy across a broad range of cancer types. However, not all patients or cancer types respond to contemporary clinically-utilised immunotherapeutic strategies, which largely focus on harnessing adaptive immune T cells for cancer treatment. Accordingly, it is increasingly recognised that upstream innate immune pathways, which govern and orchestrate the downstream adaptive immune response, may prove critical in overcoming cancer immunotherapeutic resistance. Innate lymphoid cells (ILCs) are the most recently discovered major innate immune cell population. They have overarching roles in homeostasis and orchestrating protective immunity against pathogens. As innate immune counterparts of adaptive immune T cells, ILCs exert effector functions through the secretion of cytokines and direct cell-to-cell contact, with broad influence on the overall immune response. Importantly, dysregulation of ILC subsets have been associated with a range of diseases, including immunodeficiency disorders, allergy, autoimmunity, and more recently, cancer. ILCs may either promote or inhibit cancer initiation and progression depending on the cancer type and the specific ILC subsets involved. Critically, therapeutic targeting of ILCs and their associated cytokines shows promise against a wide range of cancer types in both preclinical models and early phase oncology clinical trials. This chapter provides a comprehensive overview of the current understanding of ILC subsets and the associated cytokines they produce in cancer pathogenesis, with specific focus on how these innate pathways are, or can be targeted, therapeutically to overcome therapeutic resistance and ultimately improve patient care.
Collapse
Affiliation(s)
- Eric Jou
- Department of Oncology, Oxford University Hospitals, University of Oxford, Oxford, United Kingdom; Kellogg College, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
21
|
Chuang KF, Wang CH, Chen HK, Lin YY, Lin CH, Lin YC, Shih CP, Kuo CY, Chen YC, Chen HC. GRAIL gene knockout mice protect against aging-related and noise-induced hearing loss. J Chin Med Assoc 2023; 86:1101-1108. [PMID: 37820291 DOI: 10.1097/jcma.0000000000001005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Hearing loss is a global health issue and its etiopathologies involve complex molecular pathways. The ubiquitin-proteasome system has been reported to be associated with cochlear development and hearing loss. The gene related to anergy in lymphocytes ( GRAIL ), as an E3 ubiquitin ligase, has not, as yet, been examined in aging-related and noise-induced hearing loss mice models. METHODS This study used wild-type (WT) and GRAIL knockout (KO) mice to examine cochlear hair cells and synaptic ribbons using immunofluorescence staining. The hearing in WT and KO mice was detected using auditory brainstem response. Gene expression patterns were compared using RNA-sequencing to identify potential targets during the pathogenesis of noise-induced hearing loss in WT and KO mice. RESULTS At the 12-month follow-up, GRAIL KO mice had significantly less elevation in threshold level and immunofluorescence staining showed less loss of outer hair cells and synaptic ribbons in the hook region compared with GRAIL WT mice. At days 1, 14, and 28 after noise exposure, GRAIL KO mice had significantly less elevation in threshold level than WT mice. After noise exposure, GRAIL KO mice showed less loss of outer hair cells in the cochlear hook and basal regions compared with WT mice. Moreover, immunofluorescence staining showed less loss of synaptic ribbons in the hook regions of GRAIL KO mice than of WT mice. RNA-seq analysis results showed significant differences in C-C motif chemokine ligand 19 ( CCL19 ), C-C motif chemokine ligand 21 ( CCL21 ), interleukin 25 ( IL25 ), glutathione peroxidase 6 ( GPX6 ), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 ( NOX1 ) genes after noise exposure. CONCLUSION The present data demonstrated that GRAIL deficiency protects against aging-related and noise-induced hearing loss. The mechanism involved needs to be further clarified from the potential association with synaptic modulation, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Kai-Fen Chuang
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chih-Hung Wang
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
- Division of Otolaryngology, Taipei Veterans General Hospital, Taoyuan Branch, Taoyuan, Taiwan
| | - Hang-Kang Chen
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yuan-Yung Lin
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chia-Hsin Lin
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-Chun Lin
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Cheng-Ping Shih
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chao-Yin Kuo
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Ying-Chuan Chen
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hsin-Chien Chen
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
22
|
Chiesa Fuxench ZC, Wan J, Wang S, Syed MN, Shin DB, Abuabara K, Gelfand JM. Risk of Inflammatory Bowel Disease in Patients With Atopic Dermatitis. JAMA Dermatol 2023; 159:1085-1092. [PMID: 37647058 PMCID: PMC10469290 DOI: 10.1001/jamadermatol.2023.2875] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/27/2023] [Indexed: 09/01/2023]
Abstract
Importance Data on the association between atopic dermatitis (AD) and inflammatory bowel disease (IBD) are inconsistent. Few studies have examined the association of AD or AD severity with risk of ulcerative colitis (UC) and Crohn disease (CD) separately. Objectives To examine the risk of new-onset IBD, UC, and CD in children and adults with AD. Design, Setting, and Participants This population-based cohort study assessed patients with AD matched with up to 5 controls on age, practice, and index date. Treatment exposure was used as a proxy for AD severity. Data were retrieved from The Health Improvement Network, a UK electronic medical record database, for January 1, 1994, to February 28, 2015. Data analysis was performed from January 8, 2020, to June 30, 2023. Main Outcomes and Measures Outcomes of interest were incident IBD, UC, and CD. Logistic regression was used to examine the risk for each outcome in children and adults with AD compared with controls. Results A total of 1 809 029 pediatric controls were matched to 409 431 children with AD (93.2% mild, 5.5% moderate, and 1.3% severe). The pediatric cohort ranged in median age from 4 to 5 years (overall range, 1-10 years), was predominantly male (936 750 [51.8%] controls, 196 996 [51.6%] with mild AD, 11 379 [50.7%] with moderate AD, and 2985 [56.1%] with severe AD), and with similar socioeconomic status. A total of 2 678 888 adult controls were matched to 625 083 adults with AD (65.7% mild, 31.4% moderate, and 2.9% severe). The adult cohort ranged in median age from 45 to 50 years (overall range, 30-68 years) and was predominantly female (1 445 589 [54.0%] controls, 256 071 [62.3%] with mild AD, 109 404 [55.8%] with moderate AD, and 10 736 [59.3%] with severe AD). In fully adjusted models, children with AD had a 44% increased risk of IBD (hazard ratio [HR], 1.44; 95% CI, 1.31-1.58) and a 74% increased risk of CD (HR, 1.74; 95% CI, 1.54-1.97), which increased with worsening AD; however, they did not have increased risk of UC (HR, 1.09; 95% CI, 0.94-1.27) except for those with severe AD (HR, 1.65; 95% CI, 1.02-2.67). Adults with AD had a 34% (HR, 1.34; 95% CI, 1.27-1.40) increased risk of IBD, a 36% (HR, 1.36; 95% CI, 1.26-1.47) increased risk of CB, and a 32% (HR, 1.32; 95% CI, 1.24-1.41) increased risk of UC, with risk increasing with worsening AD. Conclusion and Relevance In this cohort study, children and adults with AD had an increased risk of IBD, with risk varying by age, AD severity, and IBD subtype. These findings provide new insights into the association between AD and IBD. Clinicians should be aware of these risks, particularly when selecting systemic treatments for AD in patients who may have coincident gastrointestinal symptoms.
Collapse
Affiliation(s)
- Zelma C. Chiesa Fuxench
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Joy Wan
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sonia Wang
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Maha N. Syed
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Daniel B. Shin
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Katrina Abuabara
- Department of Dermatology, University of California, San Francisco
| | - Joel M. Gelfand
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| |
Collapse
|
23
|
Mortier C, Gracey E, Coudenys J, Manuello T, Decruy T, Maelegheer M, Stappers F, Gilis E, Gaublomme D, Van Hoorebeke L, Van Welden S, Ambler C, Hegen M, Symanowicz P, Steyn S, Berstein G, Elewaut D, Venken K. RORγt inhibition ameliorates IL-23 driven experimental psoriatic arthritis by predominantly modulating γδ-T cells. Rheumatology (Oxford) 2023; 62:3169-3178. [PMID: 36661300 DOI: 10.1093/rheumatology/kead022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/19/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Divergent therapeutic outcomes on different disease domains have been noted with IL-23 and IL-17A-blockade in PsA. Therefore, elucidating the role of RORγt, the master regulator of type 17 immune responses, is of potential therapeutic interest. To this end, RORγt inhibition was assessed in combined skin, joint and gut inflammation in vivo, using a PsA model. METHODS We tested the efficacy of a RORγt antagonist in B10.RIII mice challenged with systemic overexpression of IL-23 by hydrodynamic injection of IL-23 enhanced episomal vector (IL-23 EEV). Clinical outcomes were evaluated by histopathology. Bone density and surface erosions were examined using micro-computed tomography. Cytokine production was measured in serum and by intracellular flow cytometry. Gene expression in PsA-related tissues was analysed by qPCR. RESULTS RORγt-blockade significantly ameliorated psoriasis, peripheral arthritis and colitis development in IL-23 EEV mice (improvement of clinical scores and weight loss respectively by 91.8%, 58.2% and 7.0%, P < 0.001), in line with profound suppression of an enhanced type IL-17 immune signature in PsA-affected tissues. Moreover, inflammation-induced bone loss and bone erosions were reduced (P < 0.05 in calcaneus, P < 0.01 in tibia). Sustained IL-23 overexpression resulted in only mild signs of sacroiliitis. Gamma-delta (γδ)-T cells, the dominant source of T cell-derived IL-17A and IL-22, were expanded during IL-23 overexpression, and together with Th17 cells, clearly countered by RORγt inhibition (P < 0.001). CONCLUSION RORγt-blockade shows therapeutic efficacy in a preclinical PsA model with protection towards extra-musculoskeletal manifestations, reflected by a clear attenuation of type 17 cytokine responses by γδ-T cells and Th17 cells.
Collapse
Affiliation(s)
- Céline Mortier
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Eric Gracey
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Julie Coudenys
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Teddy Manuello
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Tine Decruy
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Margaux Maelegheer
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Flore Stappers
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Elisabeth Gilis
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Djoere Gaublomme
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Luc Van Hoorebeke
- UGCT, Department of Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Sophie Van Welden
- IBD Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Intestinal Barrier Signaling in Disease and Therapy, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | | | - Martin Hegen
- Inflammation and Immunology Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Peter Symanowicz
- Inflammation and Immunology Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Stefan Steyn
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Cambridge, MA, USA
| | - Gabriel Berstein
- Inflammation and Immunology Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Dirk Elewaut
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Koen Venken
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
24
|
Yao X, Chen Q, Wang X, Liu X, Zhang L. IL-25 induces airway remodeling in asthma by orchestrating the phenotypic changes of epithelial cell and fibrocyte. Respir Res 2023; 24:212. [PMID: 37635231 PMCID: PMC10463650 DOI: 10.1186/s12931-023-02509-z] [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: 05/10/2023] [Accepted: 08/11/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Previous studies have shown that IL-25 levels are increased in patients with asthma with fixed airflow limitation (FAL). However, the mechanism by which IL-25 contributes to airway remodeling and FAL remains unclear. Here, we hypothesized that IL-25 facilitates pro-fibrotic phenotypic changes in bronchial epithelial cells (BECs) and circulating fibrocytes (CFs), orchestrates pathological crosstalk from BECs to CFs, and thereby contributes to airway remodeling and FAL. METHODS Fibrocytes from asthmatic patients with FAL and chronic asthma murine models were detected using flow cytometry, multiplex staining and multispectral imaging analysis. The effect of IL-25 on BECs and CFs and on the crosstalk between BECs and CFs was determined using cell culture and co-culture systems. RESULTS We found that asthmatic patients with FAL had higher numbers of IL-25 receptor (i.e., IL-17RB)+-CFs, which were negatively correlated with forced expiratory volume in 1 s/forced vital capacity (FEV1/FVC). The number of airway IL-17RB+-fibrocytes was significantly increased in ovalbumin (OVA)- and IL-25-induced asthmatic mice versus the control subjects. BECs stimulated with IL-25 exhibited an epithelial-mesenchymal transition (EMT)-like phenotypic changes. CFs stimulated with IL-25 produced high levels of extracellular matrix (ECM) proteins and connective tissue growth factors (CTGF). These profibrotic effects of IL-25 were partially blocked by the PI3K-AKT inhibitor LY294002. In the cell co-culture system, OVA-challenged BECs facilitated the migration and expression of ECM proteins and CTGF in CFs, which were markedly blocked using an anti-IL-17RB antibody. CONCLUSION These results suggest that IL-25 may serve as a potential therapeutic target for asthmatic patients with FAL.
Collapse
Affiliation(s)
- Xiujuan Yao
- Department of Respiratory and Critical Care Medicine, Beijing Tongren Hospital, Capital Medical University, No.2, Xinanhuan Road, Yizhuang District, Beijing, 100176, China
| | - Qinglin Chen
- Department of Respiratory and Critical Care Medicine, Beijing Tongren Hospital, Capital Medical University, No.2, Xinanhuan Road, Yizhuang District, Beijing, 100176, China
| | - Xiangdong Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery of Ministry of Education of China, Beijing Institute of Otolaryngology, No. 17, Hougou Hutong, Dongcheng District, Beijing, 100005, China
| | - Xiaofang Liu
- Department of Respiratory and Critical Care Medicine, Beijing Tongren Hospital, Capital Medical University, No.2, Xinanhuan Road, Yizhuang District, Beijing, 100176, China.
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
- Key Laboratory of Otolaryngology Head and Neck Surgery of Ministry of Education of China, Beijing Institute of Otolaryngology, No. 17, Hougou Hutong, Dongcheng District, Beijing, 100005, China.
| |
Collapse
|
25
|
Nedeva D, Kowal K, Mihaicuta S, Guidos Fogelbach G, Steiropoulos P, Jose Chong-Neto H, Tiotiu A. Epithelial alarmins: a new target to treat chronic respiratory diseases. Expert Rev Respir Med 2023; 17:773-786. [PMID: 37746733 DOI: 10.1080/17476348.2023.2262920] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
INTRODUCTION In response to injury, epithelial cells release alarmins including thymic stromal lymphopoietin (TSLP), high mobility group-box-1 (HMGB1), interleukin (IL)-33 and -25 that can initiate innate immune responses. These alarmins are recognized as activators of T2-immune responses characteristic for asthma, but recent evidence highlighted their role in non-T2 inflammation, airway remodeling, and pulmonary fibrosis making them an attractive therapeutic target for chronic respiratory diseases (CRD). AREAS COVERED In this review, firstly we discuss the role of TSLP, IL-33, IL-25, and HMGB1 in the pathogenesis of asthma, COPD, idiopathic pulmonary fibrosis, and cystic fibrosis according to the published data. In the second part, we summarize the current evidence concerning the efficacy of the antialarmin therapies in CRD. Recent clinical trials showed that anti-TSLP and IL-33/R antibodies can improve severe asthma outcomes. Blocking the IL-33-mediated pathway decreased the exacerbation rate in COPD patients with more important benefit for former-smokers. EXPERT OPINION Despite progress in the understanding of the alarmins' role in the pathogenesis of CRD, all their mechanisms of action are not yet identified. Blocking IL-33 and TSLP pathways offers an interesting option to treat severe asthma and COPD, but future investigations are needed to establish their place in the treatment strategies.
Collapse
Affiliation(s)
- Denislava Nedeva
- Clinic of Asthma and Allergology, UMBAL Alexandrovska, Medical University Sofia, Sofia, Bulgaria
| | - Krzysztof Kowal
- Department of Experimental Allergology and Immunology, Department of Internal Medicine and Allergology, Medical University of Bialystok, Bialystok, Poland
| | - Stefan Mihaicuta
- Center for Research and Innovation in Precision Medicine and Pharmacy, University of Medicine and Pharmacy, Timisoara, Romania
- Department of Pulmonology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | | | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Herberto Jose Chong-Neto
- Division of Allergy and Immunology, Complexo Hospital de Clinicas Federal University of Paraná, Curitiba, PR, Brazil
| | - Angelica Tiotiu
- Department of Pulmonology, University Hospital of Nancy, Vandœuvre-lès-Nancy, France
- Development, Adaptation and Disadvantage. Cardiorespiratory regulations and motor control (EA 3450 DevAH), University of Lorraine, Vandœuvre-lès-Nancy, France
| |
Collapse
|
26
|
Yuan Q, Peng N, Xiao F, Shi X, Zhu B, Rui K, Tian J, Lu L. New insights into the function of Interleukin-25 in disease pathogenesis. Biomark Res 2023; 11:36. [PMID: 37005677 PMCID: PMC10068183 DOI: 10.1186/s40364-023-00474-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/13/2023] [Indexed: 04/04/2023] Open
Abstract
Interleukin-25 (IL-25), also known as IL-17E, is a cytokine belonging to the IL-17 family. IL-25 is abundantly expressed by Th2 cells and various kinds of epithelial cells. IL-25 is an alarm signal generated upon cell injury or tissue damage to activate immune cells through the interaction with IL-17RA and IL-17RB receptors. The binding of IL-25 to IL-17RA/IL-17RB complex not only initiates and maintains type 2 immunity but also regulates other immune cells (e.g., macrophages and mast cells) via various signaling pathways. It has been well-documented that IL-25 is critically involved in the development of allergic disorders (e.g., asthma). However, the roles of IL-25 in the pathogenesis of other diseases and the underlying mechanisms are still unclear. This review presents current evidence on the roles of IL-25 in cancers, allergic disorders, and autoimmune diseases. Moreover, we discuss the unanswered key questions underlying IL-25-mediated disease pathology, which will provide new insights into the targeted therapy of this cytokine in clinical treatment.
Collapse
Affiliation(s)
- Qingfang Yuan
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Na Peng
- Department of Rheumatology, the Second People's Hospital, Three Gorges University, Yichang, China
| | - Fan Xiao
- Department of Pathology, Shenzhen Institute of Research and Innovation, The University of Hong Kong, Chongqing International Institute for Immunology, Chongqing, China
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, The First Affiliated Hospital, School of Medicine, Henan University of Science and Technology, Luoyang, China
| | - Bo Zhu
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ke Rui
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Jie Tian
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Liwei Lu
- Department of Rheumatology, the Second People's Hospital, Three Gorges University, Yichang, China.
- Department of Pathology, Shenzhen Institute of Research and Innovation, The University of Hong Kong, Chongqing International Institute for Immunology, Chongqing, China.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong, China.
| |
Collapse
|
27
|
The Interplay of Type 1, Type 2, and Type 3 Lymphocytes and Cytokines in Atopic Dermatitis. Int J Mol Sci 2023; 24:ijms24043310. [PMID: 36834723 PMCID: PMC9960233 DOI: 10.3390/ijms24043310] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Atopic dermatitis (AD) is classified as a type 2 disease owing to the majority of type 2 lymphocytes that constitute the skin-infiltrating leukocytes. However, all of the type 1-3 lymphocytes intermingle in inflamed skin lesions. Here, using an AD mouse model where caspase-1 was specifically amplified under keratin-14 induction, we analyzed the sequential changes in type 1-3 inflammatory cytokines in lymphocytes purified from the cervical lymph nodes. Cells were cultured and stained for CD4, CD8, and γδTCR, followed by intracellular cytokines. Cytokine production in innate lymphocyte cells (ILCs) and the protein expression of type 2 cytokine IL-17E (IL-25) were investigated. We observed that, as inflammation progresses, the cytokine-producing T cells increased and abundant IL-13 but low levels of IL-4 are produced in CD4-positive T cells and ILCs. TNF-α and IFN-γ levels increased continuously. The total number of T cells and ILCs peaked at 4 months and decreased in the chronic phase. In addition, IL-25 may be simultaneously produced by IL-17F-producing cells. IL-25-producing cells increased in a time-dependent manner during the chronic phase and may work specifically for the prolongation of type 2 inflammation. Altogether, these findings suggest that inhibition of IL-25 may be a potential target in the treatment of inflammation.
Collapse
|
28
|
Gauvreau GM, Bergeron C, Boulet LP, Cockcroft DW, Côté A, Davis BE, Leigh R, Myers I, O'Byrne PM, Sehmi R. Sounding the alarmins-The role of alarmin cytokines in asthma. Allergy 2023; 78:402-417. [PMID: 36463491 PMCID: PMC10108333 DOI: 10.1111/all.15609] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/14/2022] [Accepted: 11/27/2022] [Indexed: 12/07/2022]
Abstract
The alarmin cytokines thymic stromal lymphopoietin (TSLP), interleukin (IL)-33, and IL-25 are epithelial cell-derived mediators that contribute to the pathobiology and pathophysiology of asthma. Released from airway epithelial cells exposed to environmental triggers, the alarmins drive airway inflammation through the release of predominantly T2 cytokines from multiple effector cells. The upstream positioning of the alarmins is an attractive pharmacological target to block multiple T2 pathways important in asthma. Blocking the function of TSLP inhibits allergen-induced responses including bronchoconstriction, airway hyperresponsiveness, and inflammation, and subsequent clinical trials of an anti-TSLP monoclonal antibody, tezepelumab, in asthma patients demonstrated improvements in lung function, airway responsiveness, inflammation, and importantly, a reduction in the rate of exacerbations. Notably, these improvements were observed in patients with T2-high and with T2-low asthma. Clinical trials blocking IL-33 and its receptor ST2 have also shown improvements in lung function and exacerbation rates; however, the impact of blocking the IL-33/ST2 axis in T2-high versus T2-low asthma is unclear. To date, there is no evidence that IL-25 blockade is beneficial in asthma. Despite the considerable overlap in the cellular functions of IL-25, IL-33, and TSLP, they appear to have distinct roles in the immunopathology of asthma.
Collapse
Affiliation(s)
- Gail M Gauvreau
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Celine Bergeron
- Centre for Lung Health, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Donald W Cockcroft
- Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Andréanne Côté
- Centre for Lung Health, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Beth E Davis
- Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Richard Leigh
- Department of Medicine, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Irvin Myers
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Paul M O'Byrne
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Roma Sehmi
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
29
|
Wang H, Li C, Ren G, Yang C, Sun J, Zhao L, Sun W, Ju J, Xu D. Updated insight into the role of Th2-associated immunity in systemic lupus erythematosus. Autoimmun Rev 2023; 22:103213. [PMID: 36252932 DOI: 10.1016/j.autrev.2022.103213] [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: 09/16/2022] [Accepted: 10/11/2022] [Indexed: 12/27/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with multiple organs involvement, abundant autoantibodies, complement activation, and immune complexes depositions. By regulating inflammation and immune homeostasis, cytokines have been well documented to participate in the pathogenesis of SLE. A number of studies have shown that T helper 2 (Th2)-associated immunity plays an important role in autoimmune diseases, including SLE. Key molecules underlying Th2-related immunity are expected to serve as promising targets for the diagnosis and targeted treatment of SLE. Current progress in SLE pathogenesis and biological treatment strategies has been reviewed, focusing on the latest development in Th2-associated immunity.
Collapse
Affiliation(s)
- Hui Wang
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, China
| | - Chaoran Li
- Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, China
| | - Guifang Ren
- Hospital Office of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, China
| | - Chunjuan Yang
- Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, China
| | - Jiamei Sun
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, China
| | - Lu Zhao
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, China
| | - Wenchang Sun
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, China
| | - Jiyu Ju
- Department of Immunology, Weifang Medical University, Weifang 261053, China.
| | - Donghua Xu
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, China; Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, China.
| |
Collapse
|
30
|
Jou E, Rodriguez-Rodriguez N, McKenzie ANJ. Emerging roles for IL-25 and IL-33 in colorectal cancer tumorigenesis. Front Immunol 2022; 13:981479. [PMID: 36263033 PMCID: PMC9573978 DOI: 10.3389/fimmu.2022.981479] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/15/2022] [Indexed: 12/31/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide, and is largely refractory to current immunotherapeutic interventions. The lack of efficacy of existing cancer immunotherapies in CRC reflects the complex nature of the unique intestinal immune environment, which serves to maintain barrier integrity against pathogens and harmful environmental stimuli while sustaining host-microbe symbiosis during homeostasis. With their expression by barrier epithelial cells, the cytokines interleukin-25 (IL-25) and IL-33 play key roles in intestinal immune responses, and have been associated with inappropriate allergic reactions, autoimmune diseases and cancer pathology. Studies in the past decade have begun to uncover the important roles of IL-25 and IL-33 in shaping the CRC tumour immune microenvironment, where they may promote or inhibit tumorigenesis depending on the specific CRC subtype. Notably, both IL-25 and IL-33 have been shown to act on group 2 innate lymphoid cells (ILC2s), but can also stimulate an array of other innate and adaptive immune cell types. Though sometimes their functions can overlap they can also produce distinct phenotypes dependent on the differential distribution of their receptor expression. Furthermore, both IL-25 and IL-33 modulate pathways previously known to contribute to CRC tumorigenesis, including angiogenesis, tumour stemness, invasion and metastasis. Here, we review our current understanding of IL-25 and IL-33 in CRC tumorigenesis, with specific focus on dissecting their individual function in the context of distinct subtypes of CRC, and the potential prospects for targeting these pathways in CRC immunotherapy.
Collapse
Affiliation(s)
- Eric Jou
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | | |
Collapse
|
31
|
Goepfert A, Barske C, Lehmann S, Wirth E, Willemsen J, Gudjonsson JE, Ward NL, Sarkar MK, Hemmig R, Kolbinger F, Rondeau JM. IL-17-induced dimerization of IL-17RA drives the formation of the IL-17 signalosome to potentiate signaling. Cell Rep 2022; 41:111489. [PMID: 36260993 PMCID: PMC9637376 DOI: 10.1016/j.celrep.2022.111489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/20/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022] Open
Abstract
Signaling through innate immune receptors such as the Toll-like receptor (TLR)/interleukin-1 receptor (IL-1R) superfamily proceeds via the assembly of large membrane-proximal complexes or “signalosomes.” Although structurally distinct, the IL-17 receptor family triggers cellular responses that are typical of innate immune receptors. The IL-17RA receptor subunit is shared by several members of the IL-17 family. Using a combination of crystallographic, biophysical, and mutational studies, we show that IL-17A, IL-17F, and IL-17A/F induce IL-17RA dimerization. X-ray analysis of the heteromeric IL-17A complex with the extracellular domains of the IL-17RA and IL-17RC receptors reveals that cytokine-induced IL-17RA dimerization leads to the formation of a 2:2:2 hexameric signaling assembly. Furthermore, we demonstrate that the formation of the IL-17 signalosome potentiates IL-17-induced IL-36γ and CXCL1 mRNA expression in human keratinocytes, compared with a dimerization-defective IL-17RA variant. IL-17RA is the shared co-receptor for several IL-17 family members. Goepfert et al. show that IL-17 induces IL-17RA dimerization, which then drives the formation of a 2:2:2 hexameric signaling assembly with IL-17RC. Furthermore, IL-17RA dimerization potentiates IL-17 signaling in immortalized primary human keratinocytes, compared with cells expressing a dimerization-defective IL-17RA variant.
Collapse
Affiliation(s)
- Arnaud Goepfert
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Carmen Barske
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Sylvie Lehmann
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Emmanuelle Wirth
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Joschka Willemsen
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | | | - Nicole L Ward
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mrinal K Sarkar
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - René Hemmig
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Frank Kolbinger
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Jean-Michel Rondeau
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland.
| |
Collapse
|
32
|
Comparison of cytokine mediators in type 2 inflammatory conditions on the skin and ocular surface. Curr Opin Allergy Clin Immunol 2022; 22:319-327. [DOI: 10.1097/aci.0000000000000842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Interleukin-17 Family Cytokines in Metabolic Disorders and Cancer. Genes (Basel) 2022; 13:genes13091643. [PMID: 36140808 PMCID: PMC9498678 DOI: 10.3390/genes13091643] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 02/07/2023] Open
Abstract
Interleukin-17 (IL-17) family cytokines are potent drivers of inflammatory responses. Although IL-17 was originally identified as a cytokine that induces protective effects against bacterial and fungal infections, IL-17 can also promote chronic inflammation in a number of autoimmune diseases. Research in the last decade has also elucidated critical roles of IL-17 during cancer development and treatment. Intriguingly, IL-17 seems to play a role in the risk of cancers that are associated with metabolic disorders. In this review, we summarize our current knowledge on the biochemical basis of IL-17 signaling, IL-17′s involvement in cancers and metabolic disorders, and postulate how IL-17 family cytokines may serve as a bridge between these two types of diseases.
Collapse
|
34
|
Wu J, Zhang F, Tao H, Nawaz W, Chen D, Wu Z. The potential roles of interleukin-25 in infectious diseases. Front Immunol 2022; 13:986118. [PMID: 36119076 PMCID: PMC9478665 DOI: 10.3389/fimmu.2022.986118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Interleukin-25 (IL-25), also known as IL-17E, is a recently identified cytokine of the IL-17 family. Numerous studies illustrated that the expression of IL-25 is regulated by multiple pathogens, including parasitic, viral, and bacterial infections. IL-25 has a dual function in infectious diseases. On the one hand, IL-25 activates type 2 immunity via the relevant cytokines, including IL-4, IL-5, and IL-13, which are associated with the development of pathogenic infection-related allergic diseases. On the other hand, IL-25 involves in the recruitment of group 2 innate lymphoid cells (ILC2) to enhanced T helper 2 (Th2) cell differentiation, which are important to the clearance of pathogens. However, the precise roles of IL-25 in infectious diseases remain largely unknown. Thus, the current review will shed light on the pivotal roles of IL-25 in infectious diseases.
Collapse
Affiliation(s)
- Jing Wu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Fang Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hongji Tao
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Waqas Nawaz
- Hôpital Maisonneuve-Rosemont, School of Medicine, University of Montreal, Montréal, Canada
| | - Deyan Chen
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- *Correspondence: Deyan Chen, ; Zhiwei Wu,
| | - Zhiwei Wu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
- *Correspondence: Deyan Chen, ; Zhiwei Wu,
| |
Collapse
|
35
|
Feng W, Zhong XQ, Zheng XX, Liu QP, Liu MY, Liu XB, Lin CS, Xu Q. Study on the effect and mechanism of quercetin in treating gout arthritis. Int Immunopharmacol 2022; 111:109112. [PMID: 35932610 DOI: 10.1016/j.intimp.2022.109112] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022]
Abstract
Quercetin is widely found in natural plants, especially Chinese herbal plants. It has been used to treat arthritis in China for thousands of years. However, the effects and mechanisms of quercetin in the treatment of gout arthritis (GA) remain unclear. We aimed to verify the treatment of GA with quercetin and investigate the underlying mechanism. A combination of network pharmacology and experiments was used to reveal the mechanism of quercetin in the treatment of GA. Potential targets of quercetin and gout were identified. Then, the protein-protein interaction network for the common targets between quercetin and gout was constructed and the core targets were identified. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses for the common targets were performed to elucidate the pharmacological functions and mechanisms associated with quercetin treatment in GA. Finally, a monosodium urate-induced GA rat model was used to validate the predicted mechanisms in network pharmacology. Seventy-two common targets were identified. KEGG analysis revealed that treatment of GA with quercetin predominantly involved the interleukin (IL)-17, tumor necrosis factor (TNF), mitogen-activated protein kinase, and phosphoinositide 3-kinase-Akt signaling pathways. In an experimental validation, quercetin attenuated ankle joint inflammation-induced bone destruction and histological lesions. It also diminished the expression of IL-6, IL-17A, and IL-17F in the IL-17 pathway, and regulated the release of RAR-related orphan receptor gamma t,IL-17E, IL-1β, IL-6, TNF-α, Foxp3, and transforming growth factor-beta 1. The collective findings implicate quercetin as a valuable alternative drug for the treatment of GA.
Collapse
Affiliation(s)
- Wei Feng
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiao-Qin Zhong
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xue-Xia Zheng
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qing-Ping Liu
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Min-Ying Liu
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiao-Bao Liu
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Chang-Song Lin
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Qiang Xu
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
36
|
Shankar A, McAlees JW, Lewkowich IP. Modulation of IL-4/IL-13 cytokine signaling in the context of allergic disease. J Allergy Clin Immunol 2022; 150:266-276. [PMID: 35934680 PMCID: PMC9371363 DOI: 10.1016/j.jaci.2022.06.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/29/2022]
Abstract
Aberrant activation of CD4 TH2 cells and excessive production of TH2 cytokines such as IL-4 and IL-13 have been implicated in the pathogenesis of allergic diseases. Generally, IL-4 and IL-13 utilize Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathways for induction of inflammatory gene expression and the effector functions associated with disease pathology in many allergic diseases. However, it is increasingly clear that JAK/STAT pathways activated by IL-4/IL-13 can themselves be modulated in the presence of other intracellular signaling programs, thereby changing the overall tone and/or magnitude of IL-4/IL-13 signaling. Apart from direct activation of the canonic JAK/STAT pathways, IL-4 and IL-13 also induce proinflammatory gene expression and effector functions through activation of additional signaling cascades. These alternative signaling cascades contribute to several specific aspects of IL-4/IL-13-associated cellular and molecular responses. A more complete understanding of IL-4/IL-13 signaling pathways, including the precise conditions under which noncanonic signaling pathways are activated, and the impact of these pathways on cellular- and host-level responses, will better allow us to design agents that target specific pathologic outcomes or tailor therapies for the treatment of uncommon disease endotypes.
Collapse
|
37
|
Targeting Downstream Type 2 Cytokines or Upstream Epithelial Alarmins for Severe Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1497-1505. [PMID: 35131510 DOI: 10.1016/j.jaip.2022.01.040] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 12/29/2022]
Abstract
Biologics, including omalizumab, mepolizumab, benralizumab, and dupilumab, targeting downstream IgE, cytokines IL-5, and IL-4/13, respectively, have shown promising effects in terms of reduction in annualized asthma exacerbation rates (AER), oral corticosteroid-sparing effects, improvements in forced expiratory volume in 1 second, and improved Asthma Control Questionnaire scores. However, despite these welcome advances, approximately 30% of patients with severe asthma receiving biologics tailored to their specific downstream type 2 biomarkers, including total IgE, peripheral blood eosinophils, and fractional exhaled nitric oxide, do not experience meaningful improvements in their AER. Instead of blocking downstream cytokines, targeting upstream epithelial alarmins, including IL-33, thymic stromal lymphopoietin, and IL-25, has been proposed to tackle the immunologic heterogeneity of asthma. This review article aims to pragmatically summarize the latest key clinical data on antialarmin therapies in severe asthma and put these findings into context with regard to currently available downstream cytokine blockers.
Collapse
|
38
|
Hasegawa T, Oka T, Demehri S. Alarmin Cytokines as Central Regulators of Cutaneous Immunity. Front Immunol 2022; 13:876515. [PMID: 35432341 PMCID: PMC9005840 DOI: 10.3389/fimmu.2022.876515] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
Skin acts as the primary interface between the body and the environment. The skin immune system is composed of a complex network of immune cells and factors that provide the first line of defense against microbial pathogens and environmental insults. Alarmin cytokines mediate an intricate intercellular communication between keratinocytes and immune cells to regulate cutaneous immune responses. Proper functions of the type 2 alarmin cytokines, thymic stromal lymphopoietin (TSLP), interleukin (IL)-25, and IL-33, are paramount to the maintenance of skin homeostasis, and their dysregulation is commonly associated with allergic inflammation. In this review, we discuss recent findings on the complex regulatory network of type 2 alarmin cytokines that control skin immunity and highlight the mechanisms by which these cytokines regulate skin immune responses in host defense, chronic inflammation, and cancer.
Collapse
Affiliation(s)
| | - Tomonori Oka
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
39
|
Stark KG, Falkowski NR, Brown CA, McDonald RA, Huffnagle GB. Contribution of the Microbiome, Environment, and Genetics to Mucosal Type 2 Immunity and Anaphylaxis in a Murine Food Allergy Model. FRONTIERS IN ALLERGY 2022; 3:851993. [PMID: 35769569 PMCID: PMC9234882 DOI: 10.3389/falgy.2022.851993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
There is heterogeneity inherent in the immune responses of individual mice in murine models of food allergy, including anaphylaxis, similar to the clinical heterogeneity observed in humans with food allergies to a defined food. One major driver of this heterogeneity may be differences in the microbiome between sensitized individuals. Our laboratory and others have reported that disruption of the microbiome (dysbiosis) by broad spectrum antibiotics and/or yeast colonization can alter systemic immunity and favor the development of mucosal Type 2 immunity to aeroallergens. Our objective was to use a well-characterized murine model (Balb/c mice) of food allergies (chicken egg ovalbumin, OVA) and determine if antibiotic-mediated dysbiosis (including C. albicans colonization) could enhance the manifestation of food allergies. Furthermore, we sought to identify elements of the microbiome and host response that were associated with this heterogeneity in the anaphylactic reaction between individual food allergen-sensitized mice. In our dataset, the intensity of the anaphylactic reactions was most strongly associated with a disrupted microbiome that included colonization by C. albicans, loss of a specific Lachnoclostridium species (tentatively, Lachnoclostridium YL32), development of a highly polarized Type 2 response in the intestinal mucosa and underlying tissue, and activation of mucosal mast cells. Serum levels of allergen-specific IgE were not predictive of the response and a complete absence of a microbiome did not fully recapitulate the response. Conventionalization of germ-free mice resulted in Akkermansia muciniphila outgrowth and a higher degree of heterogeneity in the allergic response. C57BL/6 mice remained resistant even under the same dysbiosis-inducing antibiotic regimens, while changes in the microbiome markedly altered the reactivity of Balb/c mice to OVA, as noted above. Strikingly, we also observed that genetically identical mice from different rooms in our vivarium develop different levels of a Type 2 response, as well as anaphylactic reactions. The intestinal microbiome in these mice also differed between rooms. Thus, our data recapitulate the heterogeneity in anaphylactic reactions, ranging from severe to none, seen in patients that have circulating levels of food allergen-reactive IgE and support the concept that alterations in the microbiome can be one factor underlying this heterogeneity.
Collapse
Affiliation(s)
- Kelsey G. Stark
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Nicole R. Falkowski
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Christopher A. Brown
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
- Institute for Research on Innovation and Science (IRIS), Institute for Social Research (ISR), University of Michigan, Ann Arbor, MI, United States
| | - Roderick A. McDonald
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Gary B. Huffnagle
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Gary B. Huffnagle
| |
Collapse
|
40
|
Chen B, Yang J, Song Y, Zhang D, Hao F. Skin Immunosenescence and Type 2 Inflammation: A Mini-Review With an Inflammaging Perspective. Front Cell Dev Biol 2022; 10:835675. [PMID: 35281103 PMCID: PMC8908007 DOI: 10.3389/fcell.2022.835675] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Skin-resident stromal cells, including keratinocytes, fibroblasts, adipocytes, and immune cells including Langerhans cells, dendritic cells, T cells, and innate lymphoid cells, and their functional products work in concert to ensure the realization of skin barrier immunity. However, aging-induced immunosenescence predisposes the elderly to pruritic dermatoses, including type 2 inflammation-mediated. Inflammaging, characterized by chronic low level of pro-inflammatory cytokines released from senescent cells with the senescence-associated secretory phenotype (SASP), may drive immunosenescence and tangle with type 2 inflammatory dermatoses. The present mini-review summarizes current evidence on immunosenescence and type 2 inflammation in the skin and further focuses on future needs from an inflammaging perspective to clarify their complexity.
Collapse
Affiliation(s)
- Bangtao Chen
- Department of Dermatology, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Jing Yang
- Department of Dermatology, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yao Song
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daojun Zhang
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fei Hao
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Fei Hao,
| |
Collapse
|
41
|
Li S, Ding X, Zhang H, Ding Y, Tan Q. IL-25 improves diabetic wound healing through stimulating M2 macrophage polarization and fibroblast activation. Int Immunopharmacol 2022; 106:108605. [PMID: 35149293 DOI: 10.1016/j.intimp.2022.108605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/22/2022] [Accepted: 01/31/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Persistent chronic inflammation is one of the main pathogenic characteristics of diabetic wounds. The resolution of inflammation is important for wound healing and extracellular matrix (ECM) formation. Interleukin (IL)-25 can modulate the function of macrophage and fibroblast, but its role and mechanism of action in the treatment of diabetic wounds remain largely unclear. METHODS The mice were categorized into diabetic, diabetic + IL-25 and control groups. Human monocytic THP-1 cell line and human dermal fibroblast (HDF) were stimulated under different IL-25 conditions. Then, flow cytometry, real-time quantitative PCR (RT-qPCR), Western blot (WB), and immunofluorescence (IF) assays were carried out. RESULTS The mice in diabetes group (DG) had a slower wound healing rate, more severe inflammation, less blood vessels and more disordered collagen than those in control group (CG). Intradermal injection of IL-25 could improve these conditions. IL-25 promoted M2 macrophage polarization and fibroblast activation in DG and high-glucose environment. The phenomenon, which was dependent on PI3K/AKT/mTOR and TGF-β/SMAD signaling, could be blocked by LY294002 and LY2109761. CONCLUSION IL-25 may serve as a therapeutic target to improve wound healing in diabetic mice.
Collapse
Affiliation(s)
- Shiyan Li
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, NO. 321, Zhongshan Road, Nanjing, Jiangsu, China.
| | - Xiaofeng Ding
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, NO. 321, Zhongshan Road, Nanjing, Jiangsu, China.
| | - Hao Zhang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, NO. 321, Zhongshan Road, Nanjing, Jiangsu, China.
| | - Youjun Ding
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, NO. 321, Zhongshan Road, Nanjing, Jiangsu, China.
| | - Qian Tan
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, NO. 321, Zhongshan Road, Nanjing, Jiangsu, China; Department of Burns and Plastic Surgery, Anqing Shihua Hospital, Nanjing Drum Tower Hospital Group, Anqing 246002, China.
| |
Collapse
|
42
|
Thymic stromal lymphopoietin and alarmins as possible therapeutical targets for asthma. Curr Opin Allergy Clin Immunol 2021; 21:590-596. [PMID: 34608100 PMCID: PMC9722372 DOI: 10.1097/aci.0000000000000793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW Overview of epithelial cytokines, particularly thymic stromal lymphopoietin (TSLP), released by the airway epithelium and the effects of their inhibition on the outcomes of patients with asthma. RECENT FINDINGS The epithelial cytokines are early mediators at the top of the inflammatory cascade and are attractive therapeutic targets to prevent exacerbations and improve lung function in patients with type 2 and nontype 2 asthma. SUMMARY Clinical trials demonstrated that tezepelumab, an anti-TSLP monoclonal antibody, is a promising alternative treatment for asthma that is effective also in nontype 2 asthma. The PATHWAY and NAVIGATOR trials have assessed its effects in improving outcomes on broad clinically diverse populations. The identification of biomarkers will help to predict potential responders and help in asthma treatment personalization.
Collapse
|