1
|
Zhang Y, Chang Y, Cheng L, Wang J, Wei X, Xue M. IL-25 expression in induced sputum may serve as a reliable biomarker in children with bronchial asthma. Clin Chim Acta 2025; 575:120366. [PMID: 40398554 DOI: 10.1016/j.cca.2025.120366] [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: 12/27/2024] [Revised: 04/15/2025] [Accepted: 05/14/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Asthma is a chronic respiratory disease characterized by reversible airway obstruction and persistent airway inflammation, presenting as a highly heterogeneous disorder in children. Further understanding of its complexity is essential to identify applicable biomarkers and targeted therapies. Interleukin-25 (IL-25) has been shown to play a critical role in the pathogenesis of asthma. METHODS To investigate the association between IL and 25 expression and clinical characteristics, we enrolled46 children with asthma (age 6-17 years)and15 age-matched healthy controls. Asthma patients were stratified intoGroup A (untreated, n = 24)andGroup B (treatment-controlled, n = 22). IL-25 protein levels in serum and IL-25 mRNA in induced sputum were quantified usingELISA and PCR, respectively. RESULTS No significant intergroup differences existed in age (P = 0.32), sex (P = 0.67), or BMI (P = 0.144).IL-25 mRNA in sputumwas significantly elevated in both groups versus controls (P < 0.001 in Group A and P < 0.05 in Group B).Sputum IL-25 protein levelswerehigher in Group A versus controls (P < 0.001) and Group B (P < 0.05). IL-25 mRNA expression in sputumwas significantly higher in Group A (without anti-asthma drugs) compared to Group B (with controlled asthma treated with anti-asthma drugs) (P < 0.05 in both induced sputum and blood). Furthermore, IL-25 mRNA expression correlated with CRP (P = 0.007), FeNO (P = 0.04), FEV1/FVC (%) (P = 0.01), induced sputum eosinophil count (%) (P = 0.03), disease severity (P = 0.042), and anti-asthma treatment (P < 0.05). Notably, IL-25 levels in induced sputum decreased significantly at both molecular and gene levels following anti-asthma treatment, suggesting its potential as a biomarker for evaluating treatment efficacy and asthma control. CONCLUSION IL-25 expression in induced sputum may serve as a reliable biomarker in children with bronchial asthma, though further large-scale studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Pediatrics, Jinan Children's Hospital, Shandong University, Jinan 250022 Shandong Province, People's Republic of China.
| | - Yuna Chang
- Department of Pediatrics, Jinan Children's Hospital, Shandong University, Jinan 250022 Shandong Province, People's Republic of China
| | - Lu Cheng
- Department of Pediatrics, Jinan Children's Hospital, Shandong University, Jinan 250022 Shandong Province, People's Republic of China
| | - Jing Wang
- Department of Pediatrics, Jinan Children's Hospital, Shandong University, Jinan 250022 Shandong Province, People's Republic of China
| | - Xiaoling Wei
- Department of Pediatrics, Jinan Children's Hospital, Shandong University, Jinan 250022 Shandong Province, People's Republic of China
| | - Min Xue
- Department of Pediatrics, Jinan Children's Hospital, Shandong University, Jinan 250022 Shandong Province, People's Republic of China
| |
Collapse
|
2
|
Ver Heul AM, Mack M, Zamidar L, Tamari M, Yang TL, Trier AM, Kim DH, Janzen-Meza H, Van Dyken SJ, Hsieh CS, Karo JM, Sun JC, Kim BS. RAG suppresses group 2 innate lymphoid cells. eLife 2025; 13:RP98287. [PMID: 40326866 PMCID: PMC12055012 DOI: 10.7554/elife.98287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025] Open
Abstract
Antigen specificity is the central trait distinguishing adaptive from innate immune function. Assembly of antigen-specific T cell and B cell receptors occurs through V(D)J recombination mediated by the Recombinase Activating Gene endonucleases RAG1 and RAG2 (collectively called RAG). In the absence of RAG, mature T and B cells do not develop and thus RAG is critically associated with adaptive immune function. In addition to adaptive T helper 2 (Th2) cells, group 2 innate lymphoid cells (ILC2s) contribute to type 2 immune responses by producing cytokines like Interleukin-5 (IL-5) and IL-13. Although it has been reported that RAG expression modulates the function of innate natural killer (NK) cells, whether other innate immune cells such as ILC2s are affected by RAG remains unclear. We find that in RAG-deficient mice, ILC2 populations expand and produce increased IL-5 and IL-13 at steady state and contribute to increased inflammation in atopic dermatitis (AD)-like disease. Furthermore, we show that RAG modulates ILC2 function in a cell-intrinsic manner independent of the absence or presence of adaptive T and B lymphocytes. Lastly, employing multiomic single cell analyses of RAG1 lineage-traced cells, we identify key transcriptional and epigenomic ILC2 functional programs that are suppressed by a history of RAG expression. Collectively, our data reveal a novel role for RAG in modulating innate type 2 immunity through suppression of ILC2s.
Collapse
Affiliation(s)
- Aaron M Ver Heul
- Division of Allergy and Immunology, Department of Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Madison Mack
- Immunology and Inflammation Research Therapeutic Area, SanofiCambridgeUnited States
| | - Lydia Zamidar
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Masato Tamari
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Ting-Lin Yang
- Division of Dermatology, Department of Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Anna M Trier
- Division of Dermatology, Department of Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Do-Hyun Kim
- Department of Pathology and Immunology, Washington University School of MedicineSt. LouisUnited States
- Department of Life Science, College of Natural Sciences, Hanyang UniversitySeoulRepublic of Korea
| | - Hannah Janzen-Meza
- Division of Allergy and Immunology, Department of Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Steven J Van Dyken
- Department of Pathology and Immunology, Washington University School of MedicineSt. LouisUnited States
| | - Chyi-Song Hsieh
- Division of Rheumatology, Department of Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Jenny M Karo
- Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical CollegeNew YorkUnited States
- Immunology Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Joseph C Sun
- Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical CollegeNew YorkUnited States
- Immunology Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Brian S Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| |
Collapse
|
3
|
Kornfield JM, Bright H, Drake MG. Touching a Nerve: Neuroimmune Interactions in Asthma. Immunol Rev 2025; 331:e70025. [PMID: 40186378 PMCID: PMC12121487 DOI: 10.1111/imr.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
Asthma is an inflammatory airway disease characterized by excessive bronchoconstriction and airway hyperresponsiveness. Airway nerves play a crucial role in regulating these processes. In asthma, interactions between inflammatory cells and nerves result in nerve dysfunction, which worsens airway function. This review discusses new insights regarding the role of airway nerves in healthy lungs and examines how communication between nerves and leukocytes, including eosinophils, mast cells, dendritic cells, and innate lymphoid cells, contributes to nerve dysfunction and the worsening of airway disease. Clinical implications and therapeutic opportunities presented by neuroimmune interactions are also addressed.
Collapse
Affiliation(s)
- James M Kornfield
- Division of Pulmonary, Allergy, and Critical Care, Oregon Health and Science University, Portland, Oregon, USA
| | - Hoyt Bright
- Division of Pulmonary, Allergy, and Critical Care, Oregon Health and Science University, Portland, Oregon, USA
| | - Matthew G Drake
- Division of Pulmonary, Allergy, and Critical Care, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
4
|
Gomez RA, Hou J, Gersuk VH, Chow IT, Farrington ML, Robinson D, Kwok WW. Ara h 2 105-124-Specific TH2A Cells Drive Peanut Allergy in DRB1*15:01 Individuals: A Detailed Epitope Analysis. Clin Exp Allergy 2025. [PMID: 40308027 DOI: 10.1111/cea.70072] [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: 08/23/2024] [Revised: 03/03/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND The IgE-mediated CD4 T-cell response to peanut (Arachis hypogaea) is heterogeneous, yet TH2 cells remain central drivers of pathology. This study aimed to dissect this complexity at the epitope level by focusing on the HLA-DRB1*15:01-DRB5*01:01 haplotype. Specifically, we examined how distinct epitope-specific T-cell subsets shape the immunological landscape of peanut allergy in peanut-allergic (PA) versus non-peanut-allergic (NPA) individuals. METHODS Using in vitro and ex vivo MHC-II tetramer approaches, the phenotype, frequency, function, and transcriptome of CD4 T-cell responses to novel Ara h epitopes were assessed. Bulk RNA sequencing further characterised these T cells, allowing identification of subsets associated with TH2 polarisation in PA individuals. RESULTS Eleven HLA-DRB1*15:01 and DRB5*01:01-restricted epitopes were identified in Ara h 1, 2, 3, 6, 7, and 8 using tetramer-guided epitope mapping on cell lines, followed by ex vivo validation in peripheral blood. T-cell phenotype was epitope-dependent, with a distinct TH2A population specific to the epitope Ara h 2105-124 (Ara h 2 p14) detected only in PA donors. These TH2A cells were phenotypically and transcriptionally distinct, marked by high CRTH2/CD161, low CD27, IL-5 production, and gene enrichment in cytokine signalling and lipid metabolism. Other epitope-specific T-cell subsets displayed more heterogeneous gene profiles related to immune activation, differentiation, and antigen presentation, underscoring the complexity of peanut-specific responses even within a single HLA haplotype. CONCLUSION These findings reveal that the strong TH2 bias in DRB1*15:01-DRB5*01:01 PA individuals arises from a distinct subset of Ara h 2 p14-specific TH2A cells characterised by a specialised metabolic and cytokine signalling program. At the same time, the functional diversity observed in non-Ara h 2 p14 subsets highlights the potential for leveraging these populations in tolerance-promoting therapies. Understanding the epitope-level heterogeneity of peanut-specific T-cells provides insight into the epitope-specific mechanisms driving peanut allergy and has potential implications for therapeutic interventions.
Collapse
Affiliation(s)
- Rebecca A Gomez
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - Jue Hou
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Vivian H Gersuk
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - I-Ting Chow
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA
- Immune Medicine Division, Adaptive Biotechnologies, Seattle, Washington, USA
| | - Mary L Farrington
- Allergy and Immunology, Virginia Mason Franciscan Health, Seattle, Washington, USA
| | - David Robinson
- Allergy and Immunology, Virginia Mason Franciscan Health, Seattle, Washington, USA
| | - William W Kwok
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
5
|
Jayasinghe RG, Hollingsworth D, Schedler NC, Landy E, Boonchalermvichian C, Gupta B, Yan H, Baker J, Dejene B, Weinberg KI, Negrin RS, Mavers M. Single-cell transcriptomic profiling reveals diversity in human iNKT cells across hematologic tissues. Cell Rep 2025; 44:115587. [PMID: 40305288 DOI: 10.1016/j.celrep.2025.115587] [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: 10/07/2024] [Revised: 12/20/2024] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Invariant natural killer T (iNKT) cells are evolutionarily conserved innate lymphocytes important for protection against pathogens, malignancies, and graft-versus-host disease, with potential for universal donor cellular therapies. While mouse studies reveal transcriptionally and functionally distinct subsets, a comprehensive understanding of human iNKT cell heterogeneity is limited. Herein, we delineate the transcriptomic diversity of human iNKT cells from multiple immunologically relevant hematologic tissues. Human iNKT cells express naive/precursor, transitional, and T helper (Th)1/17/NK-like transcriptional profiles, partially contrasting with findings in mice. Additionally, these data uncover transcription factor dynamics not previously described in mice and reveal a T effector memory RA+-like population. Further, two distinct expression patterns of human CD8+ iNKT cells are described-one resembling naive/precursor cells and another resembling Th1/17/NK-like cells, with predominant expression of CD8αα protein. These critical insights into the transcriptional heterogeneity of human iNKT cells will facilitate future functional studies and inform iNKT-based cellular therapy development.
Collapse
Affiliation(s)
- Reyka G Jayasinghe
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Derek Hollingsworth
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nathan C Schedler
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Emily Landy
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chaiyaporn Boonchalermvichian
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Biki Gupta
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Hao Yan
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeanette Baker
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Beruh Dejene
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kenneth I Weinberg
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert S Negrin
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Melissa Mavers
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
6
|
Stacchiotti C, Mazzella di Regnella S, Cinotti M, Spalloni A, Volpe E. Neuroinflammation and Amyotrophic Lateral Sclerosis: Recent Advances in Anti-Inflammatory Cytokines as Therapeutic Strategies. Int J Mol Sci 2025; 26:3854. [PMID: 40332510 PMCID: PMC12028049 DOI: 10.3390/ijms26083854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Neuroinflammation is an inflammatory response occurring within the central nervous system (CNS). The process is marked by the production of pro-inflammatory cytokines, chemokines, small-molecule messengers, and reactive oxygen species. Microglia and astrocytes are primarily involved in this process, while endothelial cells and infiltrating blood cells contribute to neuroinflammation when the blood-brain barrier (BBB) is damaged. Neuroinflammation is increasingly recognized as a pathological hallmark of several neurological diseases, including amyotrophic lateral sclerosis (ALS), and is closely linked to neurodegeneration, another key feature of ALS. In fact, neurodegeneration is a pathological trigger for inflammation, and neuroinflammation, in turn, contributes to motor neuron (MN) degeneration through the induction of synaptic dysfunction, neuronal death, and inhibition of neurogenesis. Importantly, resolution of acute inflammation is crucial for avoiding chronic inflammation and tissue destruction. Inflammatory processes are mediated by soluble factors known as cytokines, which are involved in both promoting and inhibiting inflammation. Cytokines with anti-inflammatory properties may exert protective roles in neuroinflammatory diseases, including ALS. In particular, interleukin (IL)-10, transforming growth factor (TGF)-β, IL-4, IL-13, and IL-9 have been shown to exert an anti-inflammatory role in the CNS. Other recently emerging immune regulatory cytokines in the CNS include IL-35, IL-25, IL-37, and IL-27. This review describes the current understanding of neuroinflammation in ALS and highlights recent advances in the role of anti-inflammatory cytokines within CNS with a particular focus on their potential therapeutic applications in ALS. Furthermore, we discuss current therapeutic strategies aimed at enhancing the anti-inflammatory response to modulate neuroinflammation in this disease.
Collapse
Affiliation(s)
- Costanza Stacchiotti
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Simona Mazzella di Regnella
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
| | - Miriam Cinotti
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
| | - Alida Spalloni
- Molecular Neurobiology Unit, Santa Lucia Foundation, 00143 Rome, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
| |
Collapse
|
7
|
Nouari W, Aribi M. Innate lymphoid cells, immune functional dynamics, epithelial parallels, and therapeutic frontiers in infections. Int Rev Immunol 2025:1-28. [PMID: 40242974 DOI: 10.1080/08830185.2025.2490233] [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: 04/03/2024] [Revised: 02/19/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Innate lymphoid cells (ILCs) have emerged as pivotal players in the field of immunology, expanding our understanding of innate immunity beyond conventional paradigms. This comprehensive review delves into the multifaceted world of ILCs, beginning with their serendipitous discovery and traversing their ontogeny and heterogeneity. We explore the distinct subsets of ILCs unraveling their intriguing plasticity, which adds a layer of complexity to their functional repertoire. As we journey through the functional activities of ILCs, we address their role in immune responses against various infections, categorizing their interactions with helminthic parasites, bacterial pathogens, fungal infections, and viral invaders. Notably, this review offers a detailed examination of ILCs in the context of specific infections, such as Mycobacterium tuberculosis, Citrobacter rodentium, Clostridium difficile, Salmonella typhimurium, Helicobacter pylori, Listeria monocytogenes, Staphylococcus aureus, Pseudomonas aeruginosa, Influenza virus, Cytomegalovirus, Herpes simplex virus, and severe acute respiratory syndrome coronavirus 2. This selection aimed for a comprehensive exploration of ILCs in various infectious contexts, opting for microorganisms based on extensive research findings rather than considerations of virulence or emergence. Furthermore, we raise intriguing questions about the potential immune functional resemblances between ILCs and epithelial cells, shedding light on their interconnectedness within the mucosal microenvironment. The review culminates in a critical assessment of the therapeutic prospects of targeting ILCs during infection, emphasizing their promise as novel immunotherapeutic targets. Nevertheless, due to their recent discovery and evolving understanding, effectively manipulating ILCs is challenging. Ensuring specificity and safety while evaluating long-term effects in clinical settings will be crucial.
Collapse
Affiliation(s)
- Wafa Nouari
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| |
Collapse
|
8
|
Li H, Shan C, Zhu Y, Yao X, Lin L, Zhang X, Qian Y, Wang Y, Xu J, Zhang Y, Li H, Zhao L, Chen K. Helminth-induced immune modulation in colorectal cancer: exploring therapeutic applications. Front Immunol 2025; 16:1484686. [PMID: 40297577 PMCID: PMC12034720 DOI: 10.3389/fimmu.2025.1484686] [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: 08/22/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Colorectal cancer is one of the most lethal tumors, posing a financial and healthcare burden. This study investigates how helminths and pre-existing diseases such as colitis, obesity, diabetes, and gut microbiota issues influence colon cancer development and prognosis. The immune system's protective immunosuppressive response to helminth invasion minimizes inflammation-induced cell damage and DNA mutations, lowering the risk of colorectal cancer precursor lesions. Helminth infection-mediated immunosuppression can hasten colorectal cancer growth and metastasis, which is detrimental to patient outcomes. Some helminth derivatives can activate immune cells to attack cancer cells, making them potentially useful as colorectal cancer vaccines or therapies. This review also covers gene editing approaches. We discovered that using CRISPR/Cas9 to inhibit live helminths modulates miRNA, which limits tumor growth. We propose more multicenter studies into helminth therapy's long-term effects and immune regulation pathways. We hope to treat colorectal cancer patients with helminth therapy and conventional cancer treatments in an integrative setting.
Collapse
Affiliation(s)
- Hongyu Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Ocean College, Beibu Gulf University, Qinzhou, China
| | - Chaojun Shan
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yunhuan Zhu
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xiaodong Yao
- School of Marxism, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lijun Lin
- School of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Xiaofen Zhang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuncheng Qian
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuqing Wang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jialu Xu
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yijie Zhang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Hairun Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ling Zhao
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| |
Collapse
|
9
|
Perez Umana ER, Mendes E, Casaro MC, Lazarini M, Oliveira FA, Sperling AI, Ferreira CM. Exogenous acetate mitigates later enhanced allergic airway inflammation in a menopausal mouse model. Front Cell Infect Microbiol 2025; 15:1543822. [PMID: 40292217 PMCID: PMC12023485 DOI: 10.3389/fcimb.2025.1543822] [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: 12/11/2024] [Accepted: 03/06/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Asthma, an inflammatory lung disease, disproportionately affects women in adulthood and is associated with a decline in estrogen levels during the menstrual cycle and menopause. To study asthma symptoms during menopause, we used a mouse model of postmenopausal asthma via ovariectomy (OVx). Similar to human menopause, we previously discovered that re-exposure of allergic OVx mice to allergen exacerbates lung inflammation. Surprisingly, we found that probiotic treatment alleviates this inflammatory exacerbation and produces acetate as one of its metabolites. Here, we investigate whether exogenous acetate alone can inhibit the exacerbation of experimental asthma in menopause. Methods Mice received acetate administration before and during sensitization. After challenge and OVx the mice were subjected to a second challenge to test whether acetate protected against airway inflammation after menopause induction. Results Acetate administration reduced all lung T2 inflammatory responses, as well as the serum immunoglobulin (IgE) level. Early acetate treatment led to an increase in regulatory T cells, even 3 weeks after cessation of the treatment, suggesting that the increase in Treg percentage is associated with the reduction of type 2 inflammation in the airways after menopause induction, indicating its potential role in this process. Given the significant role of the lung-gut axis in asthma and the association of asthma and menopause with intestinal dysfunctions, this finding is particularly relevant; we also analyzed several markers of intestinal integrity. Compared with sham-operated mice, rechallenged allergic menopausal mice had a reduction in the intestinal epithelial genes, MUC2 and OCLN, and preventive supplementation with acetate returned their expression to normal. No change was found in menopausal mice without allergic inflammation. Conclusion In conclusion, treatment with acetate prior to estrogen level decline protects sensitized and challenged mice against later airway T2 inflammation and may restore gut homeostasis.
Collapse
Affiliation(s)
- Evelyn Roxana Perez Umana
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| | - Eduardo Mendes
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| | - Mateus Campos Casaro
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| | - Mariana Lazarini
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| | - Fernando A. Oliveira
- Cellular and Molecular Neurobiology Laboratory (LaNeC), Center of Mathematics, Computing and Cognition (CMCC), Federal University of ABC, São Bernando do Campo, Brazil
| | - Anne I. Sperling
- Pulmonary and Critical Care Laboratory, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Caroline Marcantonio Ferreira
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| |
Collapse
|
10
|
Abdu S, Xia J, Yuan H, Tan TJ, Layhadi JA, Shamji MH, McKenzie ANJ, Haloob N, Hopkins C, Woszczek G, Till SJ. IL-25 Enhances B Cell Responses in Type 2 Inflammation Through IL-17RB Receptor. Allergy 2025; 80:965-975. [PMID: 39829150 PMCID: PMC11969324 DOI: 10.1111/all.16472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 11/25/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Alarmin cytokine IL-25 promotes type 2 inflammatory responses in disorders such as asthma and chronic rhinosinusitis with nasal polyps (CRSwNP) and known targets include ILC2 and Th2 cells. However, other cellular targets for IL-25 remain poorly defined. OBJECTIVE To investigate induction and expression of IL-25 receptor (IL-17RB) by B cells and evaluate responsiveness of IL-17RB-expressing B cells to IL-25 in vitro. METHODS IL-17RB expression, regulation and function on B cells were evaluated in peripheral blood-derived B cells by flow cytometry and RT-PCR, including in response to IgE-inducing stimuli (anti-CD40 mAb and IL-4). Single-cell RNA sequencing was used to compare IL-17RB+ and IL-17RB-activated peripheral blood-derived B cells. To evaluate B cell IL-17RB expression within type 2 inflamed tissue, B cells were compared from nasal polyps, control turbinate tissue and matched peripheral blood. RESULTS Activation of B cells with anti-CD40 and IL-4 increased IL-17RB expression at both protein and mRNA level, which was further upregulated by IL-25. B cells induced to express IL-17RB responded to IL-25 with enhanced antibody production. Single-cell RNA-sequencing showed that IL17RB+ activated B cells expressed higher levels of IGHE, CCL17 and CCL22 compared to IL17RB- B cells. B cells from nasal polyp tissue expressed higher levels of surface IL-17RB compared with control tissue, correlating with patient-reported CRSwNP severity (SNOT-22). CONCLUSION Peripheral blood B cells activated under IgE-inducing conditions express surface IL-17RB, and tissue IL-17RB+ B cells are increased in type 2 inflammation. IL-17RB+ cells have a distinct transcriptional profile and respond to IL-25 with enhanced antibody production, highlighting the IL-25/IL-17RB pathway as a potential therapeutic target for CRSwNP and other type 2 inflammatory disorders.
Collapse
Affiliation(s)
- Semah Abdu
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- King's Centre for Lung HealthKing's College LondonLondonUK
| | - Jiao Xia
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- ENT Department, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Huihui Yuan
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- Department of Immunology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina
| | - Tiak Ju Tan
- National Heart and Lung InstituteImperial College LondonLondonUK
| | | | | | | | - Nora Haloob
- Department of ENT SurgeryGuy's and St Thomas' NHS Foundation TrustLondonUK
| | - Claire Hopkins
- Department of ENT SurgeryGuy's and St Thomas' NHS Foundation TrustLondonUK
| | - Grzegorz Woszczek
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- King's Centre for Lung HealthKing's College LondonLondonUK
| | - Stephen J. Till
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- King's Centre for Lung HealthKing's College LondonLondonUK
| |
Collapse
|
11
|
Luo YF, Deng Y, Yang F, Meiduosiji, Xiong X, Yuan YL, Ao SH. The role of ILC2s in asthma combined with atopic dermatitis: bridging the gap from research to clinical practice. Front Immunol 2025; 16:1567817. [PMID: 40236701 PMCID: PMC11996653 DOI: 10.3389/fimmu.2025.1567817] [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: 01/28/2025] [Accepted: 03/11/2025] [Indexed: 04/17/2025] Open
Abstract
Asthma, a complex and heterogeneous respiratory disease, is often accompanied by various comorbidities, notably atopic dermatitis (AD). AD characterized by recurrent eczematous lesions and severe itching, can trigger or exacerbate asthma. Individuals with AD are 2.16 times more likely to develop asthma compared to the reference population. Furthermore, asthmatics with AD experience more severe and frequent emergency department visits and hospital admissions compared to patients with asthma alone. The close connection between asthma and AD indicates there are overlap pathophysiologic mechanisms. It is well-known that dysregulated type 2 (T2) immune inflammation is pivotal in the development of both AD and asthma, traditionally attributed to CD4+ type 2 helper T (Th2) cells. Over the past decade, group 2 innate lymphoid cells (ILC2s), as potent innate immune cells, have been demonstrated to be the key drivers of T2 inflammation, playing a crucial role in the pathogenesis of both asthma and AD. ILC2s not only trigger T2 immune-inflammation but also coordinate the recruitment and activation of innate and adaptive immune cells, thereby intensifying the inflammatory response. They are rapidly activated by epithelium alarmins producing copious amounts of T2 cytokines such as interleukin (IL) -5 and IL-13 that mediate the airway inflammation, hyperresponsiveness, and cutaneous inflammation in asthma and AD, respectively. The promising efficiency of targeted ILC2s in asthma and AD has further proven their essential roles in the pathogenesis of both conditions. However, to the best of our knowledge, there is currently no review article specifically exploring the role of ILC2s in asthma combined with AD and their potential as future therapeutic targets. Hence, we hypothesize that ILC2s may play a role in the pathogenesis of asthma combined with AD, and targeting ILC2s could be a promising therapeutic approach for this complex condition in the future. In this review, we discuss recent insights in ILC2s biology, focus on the current knowledge of ILC2s in asthma, AD, particularly in asthma combined with AD, and suggest how this knowledge might be used for improved treatments of asthma combined with AD.
Collapse
Affiliation(s)
- Yan-fang Luo
- Department of Respirology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Deng
- Department of Respirology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Feng Yang
- Department of Respirology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Meiduosiji
- Department of Respirology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xia Xiong
- College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Dermatology, The First Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yu-lai Yuan
- Department of Respirology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Su-hua Ao
- Department of Respirology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
12
|
Palazzo E, Lotti R, Quadri M, Pincelli C, Marconi A. IL-17 Ligand and Receptor Family Members Are Differentially Expressed by Keratinocyte Subpopulations and Modulate Their Differentiation and Inflammatory Phenotype. Int J Mol Sci 2025; 26:2989. [PMID: 40243580 PMCID: PMC11988555 DOI: 10.3390/ijms26072989] [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: 03/10/2025] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by dysregulation of the interleukin 17 (IL-17) signaling axis. Given that psoriasis development depends on keratinocyte stem cells and early progenitors' sensitivity to differentiation, we analyzed IL-17 ligands and the expression and function of in a novel subset of keratinocyte subpopulations: keratinocyte stem cells (KSC) and early and late Transit Amplifying (ETA or LTA, respectively) cells. We found that all subpopulations expressed all IL-17 variants, predominantly in ETA and LTA. Conversely, IL-17 receptor expression resulted in more heterogeneity, with IL-17RA, -C, and -E being the most differentially regulated. Stimulus with IL-17A, IL-17-F, IL-17-A/F, and IL-17C promotes the upregulation of CXCL1, CXCL8, and DEFB4 mRNAs expression in both KSC and ETA. Moreover, IL-17A and IL-17A/F mainly decrease KSC proliferation and promote cell cycle block. Globally, IL-17A and IL-17A/F modulated the expression of proliferation, differentiation, and psoriasis-associated markers. Furthermore, KSC- and ETA-derived 3D reconstructions displayed increased epidermal thickness and upregulated KRT16 expression after treatment with IL-17A or IL-17A/F. Therefore, our data demonstrated that IL-17 family members perform distinctive functions in a specific keratinocyte subpopulation and define IL-17 signaling as a critical modulator of KSC behavior, proving its role in epidermal homeostasis dysregulation of psoriasis.
Collapse
Affiliation(s)
- Elisabetta Palazzo
- DermoLab, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (R.L.); (M.Q.); (C.P.); (A.M.)
| | | | | | | | | |
Collapse
|
13
|
Ver Heul AM, Mack M, Zamidar L, Tamari M, Yang TL, Trier AM, Kim DH, Janzen-Meza H, Van Dyken SJ, Hsieh CS, Karo JM, Sun JC, Kim BS. RAG suppresses group 2 innate lymphoid cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.23.590767. [PMID: 38712036 PMCID: PMC11071423 DOI: 10.1101/2024.04.23.590767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Antigen specificity is the central trait distinguishing adaptive from innate immune function. Assembly of antigen-specific T cell and B cell receptors occurs through V(D)J recombination mediated by the Recombinase Activating Gene endonucleases RAG1 and RAG2 (collectively called RAG). In the absence of RAG, mature T and B cells do not develop and thus RAG is critically associated with adaptive immune function. In addition to adaptive T helper 2 (Th2) cells, group 2 innate lymphoid cells (ILC2s) contribute to type 2 immune responses by producing cytokines like Interleukin-5 (IL-5) and IL-13. Although it has been reported that RAG expression modulates the function of innate natural killer (NK) cells, whether other innate immune cells such as ILC2s are affected by RAG remains unclear. We find that in RAG-deficient mice, ILC2 populations expand and produce increased IL-5 and IL-13 at steady state and contribute to increased inflammation in atopic dermatitis (AD)-like disease. Further, we show that RAG modulates ILC2 function in a cell-intrinsic manner independent of the absence or presence of adaptive T and B lymphocytes. Lastly, employing multiomic single cell analyses of RAG1 lineage-traced cells, we identify key transcriptional and epigenomic ILC2 functional programs that are suppressed by a history of RAG expression. Collectively, our data reveal a novel role for RAG in modulating innate type 2 immunity through suppression of ILC2s.
Collapse
Affiliation(s)
- Aaron M. Ver Heul
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Madison Mack
- Immunology & Inflammation Research Therapeutic Area, Sanofi, Cambridge, MA 02141, USA
| | - Lydia Zamidar
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Masato Tamari
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ting-Lin Yang
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Anna M. Trier
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Do-Hyun Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea
| | - Hannah Janzen-Meza
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Steven J. Van Dyken
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Chyi-Song Hsieh
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jenny M. Karo
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY 10065, USA
| | - Joseph C. Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY 10065, USA
| | - Brian S. Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai 10019
| |
Collapse
|
14
|
Zhu Z, Chen X, Luo Y, Feng R, Zhou Z, Chen R. An evaluation of mepolizumab as an add-on maintenance treatment for severe eosinophilic asthma. Expert Opin Biol Ther 2025; 25:209-220. [PMID: 39861980 DOI: 10.1080/14712598.2025.2457779] [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: 10/29/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
INTRODUCTION Clinical experience with anti-interleukin (IL)-5 biologic therapies for severe asthma has been increasing, alongside deeper and broader research focusing on the role of IL-5 and the IL-5 targeted mepolizumab. This review aims to provide an update of the evidence on the role of IL-5 and mepolizumab, with discussions of the benefits of mepolizumab and its future potential, to promote the comprehension of the pathophysiology and therapeutic approaches to asthma. AREAS COVERED For this narrative review, we conducted a database search in PubMed and Embase using the keywords 'IL-5' and 'mepolizumab,' focusing on randomized controlled trials and real-world studies up to September 2024. An overview of the pathogenesis of severe asthma, new insights on the role of IL-5 and mepolizumab, and the evidence on the efficacy and safety of mepolizumab in the treatment of severe eosinophilic asthma is provided, and its benefits in clinical remission and future applications are also discussed. EXPERT OPINION Mepolizumab holds considerable promise in asthma treatment due to its mechanism of action and multiple potential benefits. In clinical practice, it may be worth considering the exploratory initiation of mepolizumab add-on treatment from the time of medium-dose inhaled glucocorticosteroid use.
Collapse
Affiliation(s)
- Zheng Zhu
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| | - Xiaoying Chen
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| | - Yiting Luo
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| | - Rui Feng
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| | - Zicong Zhou
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| | - Ruchong Chen
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
- Guangzhou National Laboratory, Guangzhou, P.R. China
| |
Collapse
|
15
|
Mannion JM, Rahimi RA. Tissue-Resident Th2 Cells in Type 2 Immunity and Allergic Diseases. Immunol Rev 2025; 330:e70006. [PMID: 39981858 PMCID: PMC11897987 DOI: 10.1111/imr.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
Type 2 immunity represents a unique immune module that provides host protection against macro-parasites and noxious agents such as venoms and toxins. In contrast, maladaptive type 2 immune responses cause allergic diseases. While multiple cell types play important roles in type 2 immunity, recent studies in humans and murine models of chronic allergic diseases have shown that a distinct population of tissue-resident, CD4+ T helper type 2 (Th2) cells play a critical role in chronic allergic inflammation. The rules regulating Th2 cell differentiation have remained less well defined than other T cell subsets, but recent studies have shed new light into the specific mechanisms controlling Th2 cell biology in vivo. Here, we review our current understanding of the checkpoints regulating the development and function of tissue-resident Th2 cells with a focus on chronic allergic diseases. We discuss evidence for a barrier tissue checkpoint in initial Th2 cell priming, including the role of neuropeptides, damage-associated molecular patterns, and dendritic cell macro-clusters. Furthermore, we review the evidence for a second barrier tissue checkpoint that instructs the development of multi-cytokine producing, tissue-resident Th2 cells that orchestrate allergic inflammation. Lastly, we discuss potential approaches to therapeutically target tissue-resident Th2 cells in chronic allergic diseases.
Collapse
Affiliation(s)
- Jenny M Mannion
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rod A Rahimi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Kar S, Verma D, Mehrotra S, Prajapati VK. Reconfiguring the immune system to target cancer: Therapies based on T cells, cytokines, and vaccines. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:77-150. [PMID: 39978976 DOI: 10.1016/bs.apcsb.2024.10.017] [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: 02/22/2025]
Abstract
Over the years, extensive research has been dedicated to performing in-depth analysis of cancer to uncover the intricate details of its nature - including the types of cancer, causative agents, stimulators of disease progression, factors contributing to poor prognosis, and efficient therapies to restrict the metastatic aggressiveness. This chapter highlights the mechanisms through which different arms of the host immune system - namely cytokines, lymphocytes, antigen-presenting cells (APCs) -can be mobilized to eradicate cancer. Most malignant tumors are either poorly immunogenic, or are harbored in a highly immuno-suppressive microenvironment. This is why reinforcing the host's anti-tumor defenses, through infusion of pro-inflammatory cytokines, tumor antigen-loaded APCs, and anti-tumor cytotoxic cells has emerged as a viable treatment option against cancer. The chapter also highlights the ongoing preclinical and clinical studies in different malignancies and the outcome of various therapies. Although these methods are not foolproof, and antigen escape variants can still evade or develop resistance to customized therapies, they achieve disease stabilization in several cases when conventional treatments fail. In many instances, combination therapies involving cytokines, T cells, and vaccinations prove more effective than monotherapies. The limitations of the current therapies are also discussed, along with ongoing modifications aimed at improving efficacy.
Collapse
Affiliation(s)
- Sramona Kar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Divya Verma
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
17
|
Wang J, Wang X, Zhuo E, Chen B, Chan S. Gut‑liver axis in liver disease: From basic science to clinical treatment (Review). Mol Med Rep 2025; 31:10. [PMID: 39450549 PMCID: PMC11541166 DOI: 10.3892/mmr.2024.13375] [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: 11/29/2023] [Accepted: 06/14/2024] [Indexed: 10/26/2024] Open
Abstract
Incidence of a number of liver diseases has increased. Gut microbiota serves a role in the pathogenesis of hepatitis, cirrhosis and liver cancer. Gut microbiota is considered 'a new virtual metabolic organ'. The interaction between the gut microbiota and liver is termed the gut‑liver axis. The gut‑liver axis provides a novel research direction for mechanism of liver disease development. The present review discusses the role of the gut‑liver axis and how this can be targeted by novel treatments for common liver diseases.
Collapse
Affiliation(s)
- Jianpeng Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Department of Clinical Medicine, The First Clinical Medical College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xinyi Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Enba Zhuo
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Bangjie Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Shixin Chan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
18
|
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: 1] [Impact Index Per Article: 1.0] [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
|
19
|
Xu G, Paglialunga S, Qian X, Ding R, Webster K, van Haarst A, Engel C, Hui CW, Lam LH, Li W, Wu WC, Rasmussen S, Hunt A, Leung SO. Evaluation of the safety, tolerability, pharmacokinetics and pharmacodynamics of SM17 in healthy volunteers: results from pre-clinical models and a first-in-human, randomized, double blinded clinical trial. Front Immunol 2024; 15:1495540. [PMID: 39717777 PMCID: PMC11663749 DOI: 10.3389/fimmu.2024.1495540] [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: 10/15/2024] [Accepted: 11/11/2024] [Indexed: 12/25/2024] Open
Abstract
Background Alarmins mediate type 2 T helper cell (Th2) inflammation and serve as upstream signaling elements in allergic inflammation and autoimmune responses. The alarmin interleukin (IL)-25 binds to a multi-domain receptor consisting of IL-17RA and IL-17RB subunits, resulting in the release of Th2 cytokines IL-4, IL-5, IL-9 and IL-13 to drive an inflammatory response. Therefore, the blockage of IL-17RB via SM17, a novel humanized monoclonal antibody, offers an attractive therapeutic target for Th2-mediated diseases, such as asthma. Methods Wild-type mice were stimulated with house dust mite (HDM) extracts for evaluation of SM17's pre-clinical efficacy in allergic asthma. The safety, pharmacokinectics (PK), pharmacodynamics (PD), and immunogenicity of intravenous (IV) doses of SM17 were assessed in a 2-part clinical study in healthy adult subjects. In Part A, 53 healthy participants were enrolled to receive a single IV dose of SM17 (2, 20, 70, 200, 400, 600, 1200 mg) or placebo. In Part B, 24 healthy subjects were enrolled to receive a single IV dose of SM17 every two weeks (Q2W; 200, 400, 600 mg) or placebo for a total of 3 doses. Results Animal studies demonstrated that SM17 significantly suppressed Th2 inflammation in the bronchoalveolar lavage fluid and infiltration of immune cells into the lungs. In the Phase I clinical study, no drug-related serious adverse events were observed. Total SM17 exposure increased by approximately 60- to 188-fold with a 60-fold increase in dose from 20 to 1200 mg SM17. Upon administration of the third dose, mean accumulation ratios over 200-600 mg was 1.5 to 2.1, which confirms moderate accumulation of SM17. After Q2W dosing of SM17 over 4 weeks, total exposure increased in a dose-proportional manner from 200 mg to 600 mg SM17. Conclusion In the pre-clinical studies, we demonstrated that SM17 is a potential therapeutic agent to treat allergic asthma. In the Phase 1 clinical trial, a single IV dose of SM17 up to 1200 mg and three Q2W doses up to 600 mg were well tolerated in healthy participants and demonstrated a favorable safety profile. The pre-clinical efficacy and clinical PK and immunogenicity results of SM17 support further clinical development. Clinical trial registration https://clinicaltrials.gov/, identifier NCT05332834.
Collapse
Affiliation(s)
- Guolin Xu
- SinoMab BioScience Limited, Hong Kong, Hong Kong SAR, China
| | | | - Xuchen Qian
- SinoMab BioScience Limited, Hong Kong, Hong Kong SAR, China
| | - Ru Ding
- SinoMab BioScience Limited, Hong Kong, Hong Kong SAR, China
| | | | | | | | - Chin Wai Hui
- SinoMab BioScience Limited, Hong Kong, Hong Kong SAR, China
| | - Lik Hang Lam
- SinoMab BioScience Limited, Hong Kong, Hong Kong SAR, China
| | - Weimin Li
- SinoMab BioScience Limited, Hong Kong, Hong Kong SAR, China
| | - Wai Chung Wu
- SinoMab BioScience Limited, Hong Kong, Hong Kong SAR, China
| | | | - Allen Hunt
- Celerion Inc., Lincoln, NE, United States
| | - Shui-on Leung
- SinoMab BioScience Limited, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
20
|
Yue C, Zhou H, Wang X, Yu J, Hu Y, Zhou P, Zhao F, Zeng F, Li G, Li Y, Feng Y, Sun X, Huang S, He M, Wu W, Huang N, Li J. Atopic dermatitis: pathogenesis and therapeutic intervention. MedComm (Beijing) 2024; 5:e70029. [PMID: 39654684 PMCID: PMC11625510 DOI: 10.1002/mco2.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024] Open
Abstract
The skin serves as the first protective barrier for nonspecific immunity and encompasses a vast network of skin-associated immune cells. Atopic dermatitis (AD) is a prevalent inflammatory skin disease that affects individuals of all ages and races, with a complex pathogenesis intricately linked to genetic, environmental factors, skin barrier dysfunction as well as immune dysfunction. Individuals diagnosed with AD frequently exhibit genetic predispositions, characterized by mutations that impact the structural integrity of the skin barrier. This barrier dysfunction leads to the release of alarmins, activating the type 2 immune pathway and recruiting various immune cells to the skin, where they coordinate cutaneous immune responses. In this review, we summarize experimental models of AD and provide an overview of its pathogenesis and the therapeutic interventions. We focus on elucidating the intricate interplay between the immune system of the skin and the complex regulatory mechanisms, as well as commonly used treatments for AD, aiming to systematically understand the cellular and molecular crosstalk in AD-affected skin. Our overarching objective is to provide novel insights and inform potential clinical interventions to reduce the incidence and impact of AD.
Collapse
Affiliation(s)
- Chengcheng Yue
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Hong Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Xiaoyan Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Jiadong Yu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Yawen Hu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Pei Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Fulei Zhao
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Fanlian Zeng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Guolin Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Ya Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Yuting Feng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Xiaochi Sun
- Department of CardiologyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Shishi Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Mingxiang He
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Wenling Wu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Nongyu Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| |
Collapse
|
21
|
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
|
22
|
Huang YM, Shih LJ, Hsieh TW, Tsai KW, Lu KC, Liao MT, Hu WC. Type 2 hypersensitivity disorders, including systemic lupus erythematosus, Sjögren's syndrome, Graves' disease, myasthenia gravis, immune thrombocytopenia, autoimmune hemolytic anemia, dermatomyositis, and graft-versus-host disease, are THαβ-dominant autoimmune diseases. Virulence 2024; 15:2404225. [PMID: 39267271 PMCID: PMC11409508 DOI: 10.1080/21505594.2024.2404225] [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: 02/29/2024] [Revised: 08/21/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024] Open
Abstract
The THαβ host immunological pathway contributes to the response to infectious particles (viruses and prions). Furthermore, there is increasing evidence for associations between autoimmune diseases, and particularly type 2 hypersensitivity disorders, and the THαβ immune response. For example, patients with systemic lupus erythematosus often produce anti-double stranded DNA antibodies and anti-nuclear antibodies and show elevated levels of type 1 interferons, type 3 interferons, interleukin-10, IgG1, and IgA1 throughout the disease course. These cytokines and antibody isotypes are associated with the THαβ host immunological pathway. Similarly, the type 2 hypersensitivity disorders myasthenia gravis, Graves' disease, graft-versus-host disease, autoimmune hemolytic anemia, immune thrombocytopenia, dermatomyositis, and Sjögren's syndrome have also been linked to the THαβ pathway. Considering the potential associations between these diseases and dysregulated THαβ immune responses, therapeutic strategies such as anti-interleukin-10 or anti-interferon α/β could be explored for effective management.
Collapse
Affiliation(s)
- Yao-Ming Huang
- Department of Emergency Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Li-Jane Shih
- Department of Medical Laboratory, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei city, Taiwan
| | - Teng-Wei Hsieh
- Division of Immunology, Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Kuo-Wang Tsai
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Medical Tzu Chi Foundation, New Taipei City, Taiwan
- Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan
| |
Collapse
|
23
|
Fernandes AMS, da Silva ES, Silva RC, Silveira EF, Santiago LF, de Andrade Belitardo EMM, Alves VDS, Bôas DSV, de Freitas LAR, Ferreira F, Jacquet A, Pacheco LGC, Alcantara-Neves NM, Pinheiro CS. Therapeutic potential of a novel hybrid protein: Mitigating allergy and airway remodeling in chronic asthma models induced by Dermatophagoides pteronyssinus. Mol Immunol 2024; 175:121-131. [PMID: 39357098 DOI: 10.1016/j.molimm.2024.09.005] [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: 07/17/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND The house-dust mite Dermatophagoides pteronyssinus is a key trigger of allergic asthma. Therefore, it is essential to develop new vaccines that can alter inflammatory processes and airway remodeling. The goal of this study was to test the hypoallergenic and immunogenic characteristics of the hypoallergen rDer p 2231 in a murine model of chronic asthma induced by D. pteronyssinus. METHODS For this, we measured the levels of IgE, IgG1, IgG2a, and cytokines produced by mice receiving the rDer p 2231 protein. Histopathological parameters of the chronic inflammatory response were also investigated by assessing inflammation and airway remodeling. RESULTS rDer p 2231 given as a therapeutic vaccine, led to a reduction in the production of IgE, eosinophils, and neutrophils, a lower activity of eosinophilic peroxidase in the airways, and an increase in the production of IgG1 and IgG2a antibodies. IgG antibodies blocked IgE binding to parental allergens in sera from atopic patients. Splenocytes, BALF, and lung from mice treated with rDer p 2231 secreted higher levels of Th1 and regulatory cytokines, as well as reduced levels of Th2 cytokines. Histopathological investigation of the lower airways demonstrated reductions in the thickness of the bronchiolar smooth muscle layer, in the subepithelial fibrosis, and in the goblet cells hyperplasia. CONCLUSIONS Our preclinical studies suggest that rDer p 2231 is a promising candidate for the treatment of D. pteronyssinus allergy, as the hypoallergen has demonstrated the ability to reduce IgE production, induce specific blocking antibodies, restore and balance Th1/Th2 immune responses, and significantly reduce airway remodeling factors. However, additional clinical studies are needed to more accurately assess the efficacy and safety of rDer p 2231 as a vaccine against D. pteronyssinus-induced allergy.
Collapse
Affiliation(s)
| | - Eduardo Santos da Silva
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| | - Raphael Chagas Silva
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| | - Elisânia Fontes Silveira
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Leonardo Freire Santiago
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | | | - Vítor Dos Santos Alves
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Deise Souza Vilas Bôas
- Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil; Laboratory of Histotechnology, Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Luiz Antônio Rodrigues de Freitas
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FioCruz), Salvador, BA, Brazil; Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Salvador, BA, Brazil.
| | - Fatima Ferreira
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria.
| | - Alain Jacquet
- Department of Biochemistry, Chulalongkorn University, Bangkok, Thailand.
| | - Luis Gustavo Carvalho Pacheco
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Neuza Maria Alcantara-Neves
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| | - Carina Silva Pinheiro
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| |
Collapse
|
24
|
Danto SI, Tsamandouras N, Reddy P, Gilbert SA, Mancuso JY, Page K, Beebe JS, Peeva E, Vincent MS. Exploratory pharmacodynamics and efficacy of PF-06817024 in a Phase 1 study of patients with chronic rhinosinusitis and atopic dermatitis. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2024; 20:46. [PMID: 39215351 PMCID: PMC11365161 DOI: 10.1186/s13223-024-00894-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/23/2024] [Indexed: 09/04/2024]
Abstract
PF-06817024 is a humanized antibody against interleukin-33 that has the potential to inhibit type 2 inflammation. An exploratory analysis of the pharmacodynamics and clinical effects of single and repeat doses of PF-06817024 was assessed in patients with chronic rhinosinusitis with nasal polyps (CRSwNP) and patients with moderate-to-severe atopic dermatitis (AD), respectively, as part of a Phase 1, first-in-human study. Rhinosinusitis symptoms were improved, and nasal polyps were decreased in size following treatment with PF-06817024 in patients with CRSwNP. In patients with AD, PF-06817024, in aggregate, reduced disease severity and improved symptoms, as demonstrated by greater percentage decrease from baseline in Eczema Area and Severity Index (EASI) scores and reduced pruritus numerical rating scores, compared with placebo. The efficacy in AD appeared to be bimodal with a sub-group of participants exhibiting high levels of improvement (EASI75 and EASI90) for a sustained period of time after dosing. In patients with CRSwNP, a consistent trend of decrease in eosinophil levels was observed in the PF-06817024 group, compared with placebo. Further research would be needed to confirm the clinical benefit and safety of PF-06817024 as a treatment for allergic diseases. Trial registration ClinicalTrials.gov, NCT02743871. Registered 15 April 2016, https://clinicaltrials.gov/study/NCT02743871?term=NCT02743871&rank=1 .
Collapse
Affiliation(s)
| | | | - Padma Reddy
- Pfizer Inc, 1 Portland Street, Cambridge, MA, 02151, USA
| | | | | | - Karen Page
- Pfizer Inc, 1 Portland Street, Cambridge, MA, 02151, USA
| | - Jean S Beebe
- Pfizer Inc, 1 Portland Street, Cambridge, MA, 02151, USA
| | - Elena Peeva
- Pfizer Inc, 1 Portland Street, Cambridge, MA, 02151, USA
| | | |
Collapse
|
25
|
Kong B, Lai Y. IL-17 family cytokines in inflammatory or autoimmune skin diseases. Adv Immunol 2024; 163:21-49. [PMID: 39271258 DOI: 10.1016/bs.ai.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
As potent pro-inflammatory mediators, IL-17 family cytokines play crucial roles in the pathogenesis of various inflammatory and autoimmune skin disorders. Although substantial progress has been achieved in understanding the pivotal role of IL-17A signaling in psoriasis, leading to the development of highly effective biologics, the functions of other IL-17 family members in inflammatory or autoimmune skin diseases remain less explored. In this review, we provide a comprehensive overview of IL-17 family cytokines and their receptors, with a particular focus on the recent advancements in identifying cellular sources, receptors and signaling pathways regulated by these cytokines. At the end, we discuss how the aberrant functions of IL-17 family cytokines contribute to the pathogenesis of diverse inflammatory or autoimmune skin diseases.
Collapse
Affiliation(s)
- Baida Kong
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, P.R. China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, School of Life Sciences, East China Normal University, Shanghai, P.R. China
| | - Yuping Lai
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, P.R. China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, School of Life Sciences, East China Normal University, Shanghai, P.R. China.
| |
Collapse
|
26
|
Giangrazi F, Buffa D, Lloyd AT, Redmond AK, Glover LE, O'Farrelly C. Evolutionary Analysis of the Mammalian IL-17 Cytokine Family Suggests Conserved Roles in Female Fertility. Am J Reprod Immunol 2024; 92:e13907. [PMID: 39177066 DOI: 10.1111/aji.13907] [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: 05/06/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 08/24/2024] Open
Abstract
PROBLEM The interleukin-17 (IL-17) family includes pro-inflammatory cytokines IL-17A-F with important roles in mucosal defence, barrier integrity and tissue regeneration. IL-17A can be dysregulated in fertility complications, including pre-eclampsia, endometriosis and miscarriage. Because mammalian subclasses (eutherian, metatherian, and prototherian) have different related reproductive strategies, IL-17 genes and proteins were investigated in the three mammalian classes to explore their involvement in female fertility. METHOD OF STUDY Gene and protein sequences for IL-17s are found in eutherian, metatherian and prototherian mammals. Through synteny and multiple sequence protein alignment, the relationships among mammalian IL-17s were inferred. Publicly available datasets of early pregnancy stages and female fertility in therian mammals were collected and analysed to retrieve information on IL-17 expression. RESULTS Synteny mapping and phylogenetic analyses allowed the classification of mammalian IL-17 family orthologs of human IL-17. Despite differences in their primary amino acid sequence, metatherian and prototherian IL-17s share the same tertiary structure as human IL-17s, suggesting similar functions. The analysis of available datasets for female fertility in therian mammals shows up-regulation of IL-17A and IL-17D during placentation. IL-17B and IL-17D are also found to be over-expressed in human fertility complication datasets, such as endometriosis or recurrent implantation failure. CONCLUSIONS The conservation of the IL-17 gene and protein across mammals suggests similar functions in all the analysed species. Despite significant differences, the upregulation of IL-17 expression is associated with the establishment of pregnancy in eutherian and metatherian mammals. The dysregulation of IL-17s in human reproductive disorders suggests them as a potential therapeutic target.
Collapse
Affiliation(s)
- Federica Giangrazi
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Dafne Buffa
- Department of Biology, Maynooth University, Maynooth, Ireland
| | - Andrew T Lloyd
- Department of Science and Health, Institute of Technology, Carlow, Ireland
| | | | - Louise E Glover
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Reproductive Medicine, Merrion Fertility Clinic, Dublin 2, Ireland
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
27
|
López DA, Griffin A, Aguilar LM, Deering-Rice C, Myers EJ, Warren KJ, Welner RS, Beaudin AE. Prenatal inflammation remodels lung immunity and function by programming ILC2 hyperactivation. Cell Rep 2024; 43:114365. [PMID: 38909363 DOI: 10.1016/j.celrep.2024.114365] [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: 01/11/2024] [Revised: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 06/25/2024] Open
Abstract
Here, we examine how prenatal inflammation shapes tissue function and immunity in the lung by reprogramming tissue-resident immune cells from early development. Maternal, but not fetal, type I interferon-mediated inflammation provokes expansion and hyperactivation of group 2 innate lymphoid cells (ILC2s) seeding the developing lung. Hyperactivated ILC2s produce increased IL-5 and IL-13 and are associated with acute Th2 bias, decreased Tregs, and persistent lung eosinophilia into adulthood. ILC2 hyperactivation is recapitulated by adoptive transfer of fetal liver precursors following prenatal inflammation, indicative of developmental programming at the fetal progenitor level. Reprogrammed ILC2 hyperactivation and subsequent lung immune remodeling, including persistent eosinophilia, is concomitant with worsened histopathology and increased airway dysfunction equivalent to papain exposure, indicating increased asthma susceptibility in offspring. Our data elucidate a mechanism by which early-life inflammation results in increased asthma susceptibility in the presence of hyperactivated ILC2s that drive persistent changes to lung immunity during perinatal development.
Collapse
Affiliation(s)
- Diego A López
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Aleah Griffin
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Lorena Moreno Aguilar
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | | | - Elizabeth J Myers
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Kristi J Warren
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Robert S Welner
- Department of Medicine, University of Alabama, Birmingham, AL, USA
| | - Anna E Beaudin
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; Department of Internal Medicine and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
28
|
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
|
29
|
Rajeev S, Li S, Leon-Coria A, Wang A, Kraemer L, Wang SJ, Boim A, Flannigan K, Shute A, Baggio CH, Callejas BE, MacNaughton WK, Finney CAM, McKay DM. Enteric tuft cells coordinate timely expulsion of the tapeworm Hymenolepis diminuta from the murine host by coordinating local but not systemic immunity. PLoS Pathog 2024; 20:e1012381. [PMID: 39083533 PMCID: PMC11290655 DOI: 10.1371/journal.ppat.1012381] [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: 09/06/2023] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
Recognizing that enteric tuft cells can signal the presence of nematode parasites, we investigated whether tuft cells are required for the expulsion of the cestode, Hymenolepis diminuta, from the non-permissive mouse host, and in concomitant anti-helminthic responses. BALB/c and C57BL/6 mice infected with H. diminuta expelled the worms by 11 days post-infection (dpi) and displayed DCLK1+ (doublecortin-like kinase 1) tuft cell hyperplasia in the small intestine (not the colon) at 11 dpi. This tuft cell hyperplasia was dependent on IL-4Rα signalling and adaptive immunity, but not the microbiota. Expulsion of H. diminuta was slowed until at least 14 dpi, but not negated, in tuft cell-deficient Pou2f3-/- mice and was accompanied by delayed goblet cell hyperplasia and slowed small bowel transit. Worm antigen and mitogen evoked production of IL-4 and IL-10 by splenocytes from wild-type and Pou2f3-/- mice was not appreciably different, suggesting similar systemic immune reactivity to infection with H. diminuta. Wild-type and Pou2f3-/- mice infected with H. diminuta displayed partial protection against subsequent infection with the nematode Heligmosomoides bakeri. We speculate that, with respect to H. diminuta, enteric tuft cells are important for local immune events driving the rapidity of H. diminuta expulsion but are not critical in initiating or sustaining systemic Th2 responses that provide concomitant immunity against secondary infection with H. bakeri.
Collapse
Affiliation(s)
- Sruthi Rajeev
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - ShuHua Li
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Aralia Leon-Coria
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
- Department of Biology, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Arthur Wang
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Lucas Kraemer
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Susan Joanne Wang
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Annaliese Boim
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Kyle Flannigan
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Adam Shute
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Cristiane H. Baggio
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Blanca E. Callejas
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K. MacNaughton
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Constance A. M. Finney
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
- Department of Biology, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Derek M. McKay
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
30
|
Marriott H, Duchesne M, Moitra S, Okoye I, Gerla L, Mayers I, Moolji J, Adatia A, Lacy P. Upper Airway Alarmin Cytokine Expression in Asthma of Different Severities. J Clin Med 2024; 13:3721. [PMID: 38999286 PMCID: PMC11242732 DOI: 10.3390/jcm13133721] [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: 06/04/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Background: The secretion of alarmin cytokines by epithelial cells, including thymic stromal lymphopoietin (TSLP), interleukin (IL)-25, and IL-33, initiates inflammatory cascades in asthma. However, alarmin cytokine expression in the upper airways in asthma remains largely unknown. Methods: We recruited 40 participants with asthma into four groups as per the Global Initiative for Asthma (GINA) steps (10 in each group of GINA 1/2, 3, 4, and 5). Cells were derived from nasal, buccal, and throat brushings. Intracellular cytokine expression (TSLP, IL-25, and IL-33) was assessed by flow cytometry in cytokeratin 8+ (Ck8+) epithelial cells immediately following collection. Results: TSLP was significantly increased (p < 0.001) in GINA 5 patients across nasal, buccal, and throat Ck8+ epithelial cells, while IL-25 was elevated in nasal and throat samples (p < 0.003), and IL-33 levels were variable, compared with GINA 1-4 patients. Individual GINA subgroup comparison showed that TSLP levels in nasal samples from GINA 5 patients were significantly (p = 0.03) elevated but did not differ between patients with and without nasal comorbidities. IL-25 and IL-33 (obtained from nasal, buccal, and throat samples) were not significantly different in individual groups. Conclusions: Our study demonstrates for the first time that Ck8+ nasal epithelial cells from GINA 5 asthma patients express elevated levels of TSLP.
Collapse
Affiliation(s)
- Hazel Marriott
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Marc Duchesne
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Subhabrata Moitra
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Isobel Okoye
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Luke Gerla
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Irvin Mayers
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jalal Moolji
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Adil Adatia
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Paige Lacy
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
31
|
Liu J, Liu F, Liang T, Zhou Y, Su X, Li X, Zeng J, Qu P, Wang Y, Chen F, Lei Q, Li G, Cheng P. The roles of Th cells in myocardial infarction. Cell Death Discov 2024; 10:287. [PMID: 38879568 PMCID: PMC11180143 DOI: 10.1038/s41420-024-02064-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/19/2024] Open
Abstract
Myocardial infarction, commonly known as a heart attack, is a serious condition caused by the abrupt stoppage of blood flow to a part of the heart, leading to tissue damage. A significant aspect of this condition is reperfusion injury, which occurs when blood flow is restored but exacerbates the damage. This review first addresses the role of the innate immune system, including neutrophils and macrophages, in the cascade of events leading to myocardial infarction and reperfusion injury. It then shifts focus to the critical involvement of CD4+ T helper cells in these processes. These cells, pivotal in regulating the immune response and tissue recovery, include various subpopulations such as Th1, Th2, Th9, Th17, and Th22, each playing a unique role in the pathophysiology of myocardial infarction and reperfusion injury. These subpopulations contribute to the injury process through diverse mechanisms, with cytokines such as IFN-γ and IL-4 influencing the balance between tissue repair and injury exacerbation. Understanding the interplay between the innate immune system and CD4+ T helper cells, along with their cytokines, is crucial for developing targeted therapies to mitigate myocardial infarction and reperfusion injury, ultimately improving outcomes for cardiac patients.
Collapse
Affiliation(s)
- Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Tingting Liang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Xiaohan Su
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xue Li
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jiao Zeng
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Peng Qu
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yali Wang
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Fuli Chen
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qian Lei
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Gang Li
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
32
|
Kinane DF, Lappin DF, Culshaw S. The role of acquired host immunity in periodontal diseases. Periodontol 2000 2024. [PMID: 38641953 DOI: 10.1111/prd.12562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/06/2024] [Accepted: 03/04/2024] [Indexed: 04/21/2024]
Abstract
The aim of this narrative review is to relate the contribution of European researchers to the complex topic of the host immune system in periodontal disease, focusing on acquired immunity. Other chapters in this volume will address the genetics and autoantibody responses and other forms of immunity to periodontal disease. While the contribution of European authors is the focus, global literature is included in this descriptive narrative for contextual clarity, albeit many with European co-authors. The topic is relatively intense and is thus broken down into sections outlined below, tackled as descriptive narratives to enhance understanding. Any attempt at a systematic or scoping review was quickly abandoned given the descriptive nature and marked variation of approach in almost all publications. Even the most uniform area of this acquired periodontal immunology literature, antibody responses to putative pathogens in periodontal diseases, falls short of common structures and common primary outcome variables one would need and expect in clinical studies, where randomized controlled clinical trials (RCTs) abound. Addressing 'the host's role' in immunity immediately requires a discussion of host susceptibility, which necessitates consideration of genetic studies (covered elsewhere in the volume and superficially covered here).
Collapse
|
33
|
Zaiss DMW, Pearce EJ, Artis D, McKenzie ANJ, Klose CSN. Cooperation of ILC2s and T H2 cells in the expulsion of intestinal helminth parasites. Nat Rev Immunol 2024; 24:294-302. [PMID: 37798539 DOI: 10.1038/s41577-023-00942-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/07/2023]
Abstract
Type 2 immune responses form a critical defence against enteric worm infections. In recent years, mouse models have revealed shared and unique functions for group 2 innate lymphoid cells and T helper 2 cells in type 2 immune response to intestinal helminths. Both cell types use similar innate effector functions at the site of infection, whereas each population has distinct roles during different stages of infection. In this Perspective, we review the underlying mechanisms used by group 2 innate lymphoid cells and T helper 2 cells to cooperate with each other and suggest an overarching model of the interplay between these cell types over the course of a helminth infection.
Collapse
Affiliation(s)
- Dietmar M W Zaiss
- Department of Immune Medicine, University Regensburg, Regensburg, Germany.
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany.
- Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany.
| | - Edward J Pearce
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | | | - Christoph S N Klose
- Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
34
|
Yamamura Y, Nakashima C, Otsuka A. Interplay of cytokines in the pathophysiology of atopic dermatitis: insights from Murin models and human. Front Med (Lausanne) 2024; 11:1342176. [PMID: 38590314 PMCID: PMC10999685 DOI: 10.3389/fmed.2024.1342176] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 04/10/2024] Open
Abstract
The pathogenesis of atopic dermatitis (AD) is understood to be crucially influenced by three main factors: dysregulation of the immune response, barrier dysfunction, and pruritus. In the lesional skin of AD, various innate immune cells, including Th2 cells, type 2 innate lymphoid cells (ILC2s), and basophils, produce Th2 cytokines [interleukin (IL)-4, IL-5, IL-13, IL-31]. Alarmins such as TSLP, IL-25, and IL-33 are also produced by epidermal keratinocytes, amplifying type 2 inflammation. In the chronic phase, not only Th2 cells but also Th22 and Th17 cells increase in number, leading to suppression of filaggrin expression by IL-4, IL-13, and IL-22, which further deteriorates the epidermal barrier function. Dupilumab, which targets IL-4 and IL-13, has shown efficacy in treating moderate to severe AD. Nemolizumab, targeting IL-31RA, effectively reduces pruritus in AD patients. In addition, clinical trials with fezakinumab, targeting IL-22, have demonstrated promising results, particularly in severe AD cases. Conversely, in murine models of AD, several cytokines, initially regarded as promising therapeutic targets, have not demonstrated sufficient efficacy in clinical trials. IL-33 has been identified as a potent activator of immune cells, exacerbating AD in murine models and correlating with disease severity in human patients. However, treatments targeting IL-33 have not shown sufficient efficacy in clinical trials. Similarly, thymic stromal lymphopoietin (TSLP), integral to type 2 immune responses, induces dermatitis in animal models and is elevated in human AD, yet clinical treatments like tezepelumab exhibit limited efficacy. Therapies targeting IL-1α, IL-5, and IL-17 also failed to achieve sufficient efficacy in clinical trials. It has become clear that for treating AD, IL-4, IL-13, and IL-31 are relevant therapeutic targets during the acute phase, while IL-22 emerges as a target in more severe cases. This delineation underscores the necessity of considering distinct pathophysiological aspects and therapeutic targets in AD between mouse models and humans. Consequently, this review delineates the distinct roles of cytokines in the pathogenesis of AD, juxtaposing their significance in human AD from clinical trials against insights gleaned from AD mouse models. This approach will improve our understanding of interspecies variation and facilitate a deeper insight into the pathogenesis of AD in humans.
Collapse
Affiliation(s)
| | - Chisa Nakashima
- Department of Dermatology, Faculty of Medicine, Kindai University Hospital, Osaka, Japan
| | | |
Collapse
|
35
|
Roostaee A, Yaghobi R, Afshari A, Jafarinia M. Regulatory role of T helper 9/interleukin-9: Transplantation view. Heliyon 2024; 10:e26359. [PMID: 38420400 PMCID: PMC10900956 DOI: 10.1016/j.heliyon.2024.e26359] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
T helper 9 (Th9) cells, a subset of CD4+ T helper cells, have emerged as a valuable target for immune cell therapy due to their potential to induce immunomodulation and tolerance. The Th9 cells mainly produce interleukin (IL)-9 and are known for their defensive effects against helminth infections, allergic and autoimmune responses, and tumor suppression. This paper explores the mechanisms involved in the generation and differentiation of Th9 cells, including the cytokines responsible for their polarization and stabilization, the transcription factors necessary for their differentiation, as well as the role of Th9 cells in inflammatory and autoimmune diseases, allergic reactions, and cancer immunotherapies. Recent research has shown that the differentiation of Th9 cells is coregulated by the transcription factors transforming growth factor β (TGF-β), IL-4, and PU.1, which are also known to secrete IL-10 and IL-21. Multiple cell types, such as T and B cells, mast cells, and airway epithelial cells, are influenced by IL-9 due to its pleiotropic effects.
Collapse
Affiliation(s)
- Azadeh Roostaee
- Department of Genetics, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Ramin Yaghobi
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afsoon Afshari
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Jafarinia
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| |
Collapse
|
36
|
Kim HY, Jeong D, Kim JH, Chung DH. Innate Type-2 Cytokines: From Immune Regulation to Therapeutic Targets. Immune Netw 2024; 24:e6. [PMID: 38455467 PMCID: PMC10917574 DOI: 10.4110/in.2024.24.e6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 03/09/2024] Open
Abstract
The intricate role of innate type-2 cytokines in immune responses is increasingly acknowledged for its dual nature, encompassing both protective and pathogenic dimensions. Ranging from defense against parasitic infections to contributing to inflammatory diseases like asthma, fibrosis, and obesity, these cytokines intricately engage with various innate immune cells. This review meticulously explores the cellular origins of innate type-2 cytokines and their intricate interactions, shedding light on factors that amplify the innate type-2 response, including TSLP, IL-25, and IL-33. Recent advancements in therapeutic strategies, specifically the utilization of biologics targeting pivotal cytokines (IL-4, IL-5, and IL-13), are discussed, offering insights into both challenges and opportunities. Acknowledging the pivotal role of innate type-2 cytokines in orchestrating immune responses positions them as promising therapeutic targets. The evolving landscape of research and development in this field not only propels immunological knowledge forward but also holds the promise of more effective treatments in the future.
Collapse
Affiliation(s)
- Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Korea
| | - Dongjin Jeong
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Ji Hyung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Doo Hyun Chung
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
37
|
Tsuda T, Suzuki M, Kato Y, Kidoguchi M, Kumai T, Fujieda S, Sakashita M. The current findings in eosinophilic chronic rhinosinusitis. Auris Nasus Larynx 2024; 51:51-60. [PMID: 37574421 DOI: 10.1016/j.anl.2023.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023]
Abstract
Chronic rhinosinusitis (CRS) is a persistent inflammatory disease of the nasal cavity and paranasal sinuses. Traditional classification is denoted by the presence (CRSwNP) or absence of nasal polyps (CRSsNP). Particularly, CRSwNP is distinguished by the presence of infiltrating cells and inflammatory markers in the nasal mucosa. Patients with CRSwNP in Western countries predominantly display a type 2 endotype, whereas those in Asian regions display a mixed type 2 endotype. Nevertheless, recent transcriptome analyses have revealed two types of nasal polyps - type 2 and non-type 2 polyps, suggesting that geographical differences in endotypes likely resulted from the different proportions of each endotype. Moreover, various endotypes of CRSsNP have been identified, making phenotype a crucial factor for predicting treatment efficacy. Type 2 endotypes, designated as eosinophilic CRS (ECRS) in Japan, are characterized by severe eosinophilic infiltration into the paranasal sinus tissue and are particularly refractory. In this review, we discuss the latest developments in ECRS. We also provide recent findings on the involvement of nasal epithelial cells in pathogenesis.
Collapse
Affiliation(s)
- Takeshi Tsuda
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masanobu Suzuki
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 14-jo nishi 5, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Yukinori Kato
- Division of Otorhinolaryngology and Head & Neck Surgery, Department of Sensory and Locomotor Medicine Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, Yoshida, Eiheiji, Fukui 910-1193, Japan
| | - Masanori Kidoguchi
- Division of Otorhinolaryngology and Head & Neck Surgery, Department of Sensory and Locomotor Medicine Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, Yoshida, Eiheiji, Fukui 910-1193, Japan
| | - Takumi Kumai
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Shigeharu Fujieda
- Division of Otorhinolaryngology and Head & Neck Surgery, Department of Sensory and Locomotor Medicine Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, Yoshida, Eiheiji, Fukui 910-1193, Japan
| | - Masafumi Sakashita
- Division of Otorhinolaryngology and Head & Neck Surgery, Department of Sensory and Locomotor Medicine Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, Yoshida, Eiheiji, Fukui 910-1193, Japan.
| |
Collapse
|
38
|
Yang H, Huang YX, Xiong PY, Li JQ, Chen JL, Liu X, Gong YJ, Ding WJ. Possible connection between intestinal tuft cells, ILC2s and obesity. Front Immunol 2024; 14:1266667. [PMID: 38283340 PMCID: PMC10811205 DOI: 10.3389/fimmu.2023.1266667] [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: 07/26/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024] Open
Abstract
Intestinal tuft cells (TCs) are defined as chemosensory cells that can "taste" danger and induce immune responses. They play a critical role in gastrointestinal parasite invasion, inflammatory bowel diseases and high-fat diet-induced obesity. Intestinal IL-25, the unique product of TCs, is a key activator of type 2 immunity, especially to promote group 2 innate lymphoid cells (ILC2s) to secret IL-13. Then the IL-13 mainly promotes intestinal stem cell (ISCs) proliferation into TCs and goblet cells. This pathway formulates the circuit in the intestine. This paper focuses on the potential role of the intestinal TC, ILC2 and their circuit in obesity-induced intestinal damage, and discussion on further study and the potential therapeutic target in obesity.
Collapse
Affiliation(s)
- Hong Yang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Xing Huang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Pei-Yu Xiong
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin-Qian Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ji-Lan Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xia Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan-Ju Gong
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei-Jun Ding
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
39
|
Xia CW, Saranchova I, Finkel PL, Besoiu S, Munro L, Pfeifer CG, Haegert A, Lin YY, Le Bihan S, Collins C, Jefferies WA. A diversity of novel type-2 innate lymphoid cell subpopulations revealed during tumour expansion. Commun Biol 2024; 7:12. [PMID: 38172434 PMCID: PMC10764766 DOI: 10.1038/s42003-023-05536-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 11/01/2023] [Indexed: 01/05/2024] Open
Abstract
Type 2 innate lymphoid cells (ILC2s) perform vital functions in orchestrating humoral immune responses, facilitating tissue remodelling, and ensuring tissue homeostasis. Additionally, in a role that has garnered considerably less attention, ILC2s can also enhance Th1-related cytolytic T lymphocyte immune responses against tumours. Studies have thus far generally failed to address the mystery of how one ILC2 cell-type can participate in a multiplicity of functions. Here we utilized single cell RNA sequencing analysis to create the first comprehensive atlas of naïve and tumour-associated lung ILC2s and discover multiple unique subtypes of ILC2s equipped with developmental gene programs that become skewed during tumour expansion favouring inflammation, antigen processing, immunological memory and Th1-related anti-tumour CTL responses. The discovery of these new subtypes of ILC2s challenges current paradigms of ILC2 biology and provides an explanation for their diversity of function.
Collapse
Affiliation(s)
- Clara Wenjing Xia
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
| | - Iryna Saranchova
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Pablo L Finkel
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
| | - Stephanie Besoiu
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
| | - Lonna Munro
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Cheryl G Pfeifer
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Anne Haegert
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Yen-Yi Lin
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Stéphane Le Bihan
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Colin Collins
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Wilfred A Jefferies
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada.
- The Laboratory for Advanced Genome Analysis (LAGA), The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada.
- Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada.
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada.
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada.
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z4, Canada.
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z4, Canada.
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
40
|
Liu Y, Ouyang Y, You W, Liu W, Cheng Y, Mai X, Shen Z. Physiological roles of human interleukin-17 family. Exp Dermatol 2024; 33:e14964. [PMID: 37905720 DOI: 10.1111/exd.14964] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 11/02/2023]
Abstract
Interleukin-17 s (IL-17s) are well-known proinflammatory cytokines, and their antagonists perform excellently in the treatment of inflammatory skin diseases such as psoriasis. However, their physiological functions have not been given sufficient attention by clinicians. IL-17s can protect the host from extracellular pathogens, maintain epithelial integrity, regulate cognitive processes and modulate adipocyte activity through distinct mechanisms. Here, we present a systematic review concerning the physiological functions of IL-17s. Our goal is not to negate the therapeutic effect of IL-17 antagonists, but to ensure their safe use and reasonably explain the possible adverse events that may occur in their application.
Collapse
Affiliation(s)
- Yucong Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ye Ouyang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wanchun You
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wenqi Liu
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yufan Cheng
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xinming Mai
- Medical School, Shenzhen University, Shenzhen, China
| | - Zhu Shen
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
Poto R, Fusco W, Rinninella E, Cintoni M, Kaitsas F, Raoul P, Caruso C, Mele MC, Varricchi G, Gasbarrini A, Cammarota G, Ianiro G. The Role of Gut Microbiota and Leaky Gut in the Pathogenesis of Food Allergy. Nutrients 2023; 16:92. [PMID: 38201921 PMCID: PMC10780391 DOI: 10.3390/nu16010092] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Food allergy (FA) is a growing public health concern, with an increasing prevalence in Western countries. Increasing evidence suggests that the balance of human gut microbiota and the integrity of our intestinal barrier may play roles in the development of FA. Environmental factors, including industrialization and consumption of highly processed food, can contribute to altering the gut microbiota and the intestinal barrier, increasing the susceptibility to allergic sensitization. Compositional and functional alterations to the gut microbiome have also been associated with FA. In addition, increased permeability of the gut barrier allows the translocation of allergenic molecules, triggering Th2 immune responses. Preclinical and clinical studies have highlighted the potential of probiotics, prebiotics, and postbiotics in the prevention and treatment of FA through enhancing gut barrier function and promoting the restoration of healthy gut microbiota. Finally, fecal microbiota transplantation (FMT) is now being explored as a promising therapeutic strategy to prevent FA in both experimental and clinical studies. In this review article, we aim to explore the complex interplay between intestinal permeability and gut microbiota in the development of FA, as well as depict potential therapeutic strategies.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - William Fusco
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Emanuele Rinninella
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Cintoni
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Kaitsas
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
| | - Pauline Raoul
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Cristiano Caruso
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Maria Cristina Mele
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
42
|
Li X, Peng Z, An K, Xue M, Wang Z, Xia J, Qi Z, Shu X. Temsirolimus is a promising immunomodulatory agent for enhanced transplantation outcomes. Transpl Immunol 2023; 81:101952. [PMID: 37918580 DOI: 10.1016/j.trim.2023.101952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Identifying effective immunosuppressive strategies is critical for addressing immunological rejection following organ transplantation. This study explores the potential immunosuppressive effects and mechanisms of temsirolimus, a rapamycin derivative, in organ transplantation. METHODS A mouse cardiac allograft model was established using a cervical cannula technique with BALB/c donors and C57BL/6 recipients. Mice were administered temsirolimus intragastrically and graft survival was evaluated. Histological staining was used to assess pathological changes. The BrdU assay was used to measure splenic T cell proliferation. Flow cytometry was used to quantify regulatory T cells (Tregs), CD4+ T cells, and CD8+ T cells. ELISA and qPCR assays were used to determine Foxp3, IL-4, IFN-γ, and TGF-β expression. RESULTS Temsirolimus displayed potent immunosuppressive effects at 20 mg/kg/day, significantly inhibiting T cell proliferation (84.6%, P < 0.0001) and prolonging graft survival (median 49 days vs. 8.5 days in controls, P < 0.0001). However, median survival decreased to 34.5 days upon withdrawal. Temsirolimus also reduced splenic CD4+ and CD8+ T cells (2.85% and 2.92%, P < 0.001) and antibody levels (IgM, IgG1, IgG2) by 11.85-29.09% (P < 0.0001) and increased Tregs, Foxp3, IL-4 (P < 0.01), and TGF-β (P < 0.05), while decreasing IFN-γ (P < 0.001). CONCLUSIONS Temsirolimus exhibited potent immunosuppressive effects, emerging as a strong candidate to mitigate organ transplant rejection.
Collapse
Affiliation(s)
- Xianguo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zuojie Peng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke An
- Department of Physiology, Xuzhou Medical University, Xuzhou 221009, China
| | - Mengjiao Xue
- Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhenzhen Wang
- Department of Pharmacy, Zhoukou Central Hospital, Zhoukou 466000, China
| | - Junjie Xia
- Organ Transplantation Institute, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen 361100, China.
| | - Zhongquan Qi
- Medical College of Guangxi University, Guangxi University, Nanning 530004, China.
| | - Xiaogang Shu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
43
|
Narsale A, Almanza F, Tran T, Lam B, Seo D, Vu A, Long SA, Cooney L, Serti E, Davies JD. Th2 cell clonal expansion at diagnosis in human type 1 diabetes. Clin Immunol 2023; 257:109829. [PMID: 37907122 DOI: 10.1016/j.clim.2023.109829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023]
Abstract
Soon after diagnosis with type 1 diabetes (T1D), many patients experience a period of partial remission. A longer partial remission is associated with a better response to treatment, but the mechanism is not known. The frequency of CD4+CD25+CD127hi (127-hi) cells, a cell subset with an anti-inflammatory Th2 bias, correlates positively with length of partial remission. The purpose of this study was to further characterize the nature of the Th2 bias in 127-hi cells. Single cell RNA sequencing paired with TCR sequencing of sorted 127-hi memory cells identifies clonally expanded Th2 clusters in 127-hi cells from T1D, but not from healthy donors. The Th2 clusters express GATA3, GATA3-AS1, PTGDR2, IL17RB, IL4R and IL9R. The existence of 127-hi Th2 cell clonal expansion in T1D suggests that disease factors may induce clonal expansion of 127-hi Th2 cells that prolong partial remission and delay disease progression.
Collapse
Affiliation(s)
- Aditi Narsale
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - Francisco Almanza
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - Theo Tran
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA
| | - Breanna Lam
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - David Seo
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA
| | - Alisa Vu
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - S Alice Long
- Benaroya Research Institute, 1201 9(th) Ave, Seattle, WA 98101, USA.
| | | | | | - Joanna D Davies
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| |
Collapse
|
44
|
López DA, Griffin A, Aguilar LM, Rice CD, Myers EJ, Warren KJ, Welner R, Beaudin AE. Prenatal inflammation reprograms hyperactive ILC2s that promote allergic lung inflammation and airway dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567899. [PMID: 38045298 PMCID: PMC10690173 DOI: 10.1101/2023.11.20.567899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Allergic asthma is a chronic respiratory disease that initiates in early life, but causal mechanisms are poorly understood. Here we examined how prenatal inflammation shapes allergic asthma susceptibility by reprogramming lung immunity from early development. Induction of Type I interferon-mediated inflammation during development provoked expansion and hyperactivation of group 2 innate lymphoid cells (ILC2s) seeding the developing lung. Hyperactivated ILC2s produced increased IL-5 and IL-13, and were associated with acute Th2 bias, eosinophilia, and decreased Tregs in the lung. The hyperactive ILC2 phenotype was recapitulated by adoptive transfer of a fetal liver precursor following exposure to prenatal inflammation, indicative of developmental programming. Programming of ILC2 function and subsequent lung immune remodeling by prenatal inflammation led to airway dysfunction at baseline and in response to papain, indicating increased asthma susceptibility. Our data provide a link by which developmental programming of progenitors by early-life inflammation drives lung immune remodeling and asthma susceptibility through hyperactivation of lung-resident ILC2s. One Sentence Summary Prenatal inflammation programs asthma susceptibility by inducing the production of hyperactivated ILC2s in the developing lung.
Collapse
|
45
|
Singh BK, Yokoyama Y, Tanaka Y, Laczkó D, Deshpande DA, Kambayashi T. Diacylglycerol kinase zeta deficiency attenuates papain-induced type 2 airway inflammation. Cell Immunol 2023; 393-394:104780. [PMID: 37918056 DOI: 10.1016/j.cellimm.2023.104780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/10/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Allergic airway diseases are caused by inappropriate immune responses directed against inhaled environmental antigens. We previously reported that the inhibition of diacylglycerol (DAG) kinaseζ (DGKζ),an enzyme that terminates DAG-mediated signaling,protects against T cell-mediated allergic airway inflammation by blocking Th2 cell differentiation.In this study, we tested whether DGKζ deficiency also affects allergic airway disease mediated by type 2 innate lymphoid cells (ILC2)s. DGKζ-deficient mice displayed diminished ILC2 function and reduced papain-induced airway inflammation compared to wildtype mice. Unexpectedly, however, mice with hematopoietic cell-specific deletion ofDGKζ displayed intact airway inflammation upon papain challenge. Rather, bone marrow chimera studies revealed thatDGKζ deficiency in the non-hematopoietic compartment was responsible for the reduction in papain-induced airway inflammation. These data suggest that DGK might represent a novel therapeutic target not only for T cell-dependent but also ILC2-dependent allergic airway inflammation by affecting non-hematopoietic cells.
Collapse
Affiliation(s)
- Brenal K Singh
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Yuichi Yokoyama
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yukinori Tanaka
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dorottya Laczkó
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Deepak A Deshpande
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
46
|
Parrish A, Boudaud M, Grant ET, Willieme S, Neumann M, Wolter M, Craig SZ, De Sciscio A, Cosma A, Hunewald O, Ollert M, Desai MS. Akkermansia muciniphila exacerbates food allergy in fibre-deprived mice. Nat Microbiol 2023; 8:1863-1879. [PMID: 37696941 PMCID: PMC10522492 DOI: 10.1038/s41564-023-01464-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/05/2023] [Indexed: 09/13/2023]
Abstract
Alterations in the gut microbiome, including diet-driven changes, are linked to the rising prevalence of food allergy. However, little is known about how specific gut bacteria trigger the breakdown of oral tolerance. Here we show that depriving specific-pathogen-free mice of dietary fibre leads to a gut microbiota signature with increases in the mucin-degrading bacterium Akkermansia muciniphila. This signature is associated with intestinal barrier dysfunction, increased expression of type 1 and 2 cytokines and IgE-coated commensals in the colon, which result in an exacerbated allergic reaction to food allergens, ovalbumin and peanut. To demonstrate the causal role of A. muciniphila, we employed a tractable synthetic human gut microbiota in gnotobiotic mice. The presence of A. muciniphila within the microbiota, combined with fibre deprivation, resulted in stronger anti-commensal IgE coating and innate type-2 immune responses, which worsened symptoms of food allergy. Our study provides important insights into how gut microbes can regulate immune pathways of food allergy in a diet-dependent manner.
Collapse
Affiliation(s)
- Amy Parrish
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Erica T Grant
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Stéphanie Willieme
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Mareike Neumann
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Mathis Wolter
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sophie Z Craig
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alessandro De Sciscio
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Antonio Cosma
- National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Oliver Hunewald
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
47
|
Laubhahn K, Schaub B. From preschool wheezing to asthma: Immunological determinants. Pediatr Allergy Immunol 2023; 34:e14038. [PMID: 37877843 DOI: 10.1111/pai.14038] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/26/2023]
Abstract
Asthma represents a chronic respiratory disease affecting millions of children worldwide. The transition from preschool wheezing to school-age asthma involves a multifaceted interplay of various factors, including immunological aspects in early childhood. These factors include complex cellular interactions among different immune cell subsets, induction of pro-inflammatory mediators and the molecular impact of environmental factors like allergens or viral infections on the developing immune system. Furthermore, the activation of specific genes and signalling pathways during this early phase plays a pivotal role in the manifestation of symptoms and subsequent development of asthma. Early identification of the propensity or risk for asthma development, for example by allergen sensitisation and viral infections during this critical period, is crucial for understanding the transition from wheeze to asthma. Favourable immune regulation during a critical 'window of opportunity' in early childhood can induce persistent changes in immune cell behaviour. In this context, trained immunity, including memory function of innate immune cells, has significant implications for understanding immune responses, potentially shaping long-term immunological outcomes based on early-life environmental exposures. Exploration of these underlying immune mechanisms that drive disease progression will provide valuable insights to understand childhood asthma development. This will be instrumental to develop preventive strategies at different stages of disease development for (i) inhibiting progression from wheeze to asthma or (ii) reducing disease severity and (iii) uncovering novel therapeutic strategies and contributing to more tailored and effective treatments for childhood asthma. In the long term, this shall empower healthcare professionals to develop evidence-based interventions that reduce the burden of asthma for children, families and society overall.
Collapse
Affiliation(s)
- Kristina Laubhahn
- Department of Pulmonary and Allergy, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Munich, Germany
- Member of German Centre for Lung Research - DZL, LMU Munich, Munich, Germany
| | - Bianca Schaub
- Department of Pulmonary and Allergy, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Munich, Germany
- Member of German Centre for Lung Research - DZL, LMU Munich, Munich, Germany
| |
Collapse
|
48
|
Meloun A, León B. Sensing of protease activity as a triggering mechanism of Th2 cell immunity and allergic disease. FRONTIERS IN ALLERGY 2023; 4:1265049. [PMID: 37810200 PMCID: PMC10552645 DOI: 10.3389/falgy.2023.1265049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
CD4 T-helper cell type 2 (Th2) cells mediate host defense against extracellular parasites, like helminths. However, Th2 cells also play a pivotal role in the onset and progression of allergic inflammatory diseases such as atopic dermatitis, allergic rhinitis, asthma, and food allergy. This happens when allergens, which are otherwise harmless foreign proteins, are mistakenly identified as "pathogenic." Consequently, the encounter with these allergens triggers the activation of specific Th2 cell responses, leading to the development of allergic reactions. Understanding the molecular basis of allergen sensing is vital for comprehending how Th2 cell responses are erroneously initiated in individuals with allergies. The presence of protease activity in allergens, such as house dust mites (HDM), pollen, fungi, or cockroaches, has been found to play a significant role in triggering robust Th2 cell responses. In this review, we aim to examine the significance of protease activity sensing in foreign proteins for the initiation of Th2 cell responses, highlighting how evolving a host protease sensor may contribute to detect invading helminth parasites, but conversely can also trigger unwanted reactions to protease allergens. In this context, we will explore the recognition receptors activated by proteolytic enzymes present in major allergens and their contribution to Th2-mediated allergic responses. Furthermore, we will discuss the coordinated efforts of sensory neurons and epithelial cells in detecting protease allergens, the subsequent activation of intermediary cells, including mast cells and type 2 innate lymphoid cells (ILC2s), and the ultimate integration of all signals by conventional dendritic cells (cDCs), leading to the induction of Th2 cell responses. On the other hand, the review highlights the role of monocytes in the context of protease allergen exposure and their interaction with cDCs to mitigate undesirable Th2 cell reactions. This review aims to provide insights into the innate functions and cell communications triggered by protease allergens, which can contribute to the initiation of detrimental Th2 cell responses, but also promote mechanisms to effectively suppress their development.
Collapse
Affiliation(s)
| | - Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
49
|
Loperfido A, Ciofalo A, Cavaliere C, Begvarfaj E, Cascone F, Alfonzo G, Cadeddu R, Millarelli S, Bellocchi G, Greco A, de Vincentiis M, Masieri S. Dupilumab's Impact on Blood Parameters in Nasal Polyposis: 18-Month Follow-Up in Real Life. J Immunol Res 2023; 2023:4027701. [PMID: 37745203 PMCID: PMC10516700 DOI: 10.1155/2023/4027701] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 07/30/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
Background Dupilumab represents the first approved biological for severe uncontrolled chronic rhinosinusitis with nasal polyps (CRSwNP). Objective Aim of this paper is to provide a multicentric real-life study about treatment with dupilumab for CRSwNP with a special focus on blood parameters and IgE, IgG, and IgA. Method A retrospective data collection was jointly conducted at the Otolaryngology departments of San Camillo Forlanini Hospital and University of Rome "La Sapienza" from December 2020 to January 2023. Results A total of 130 patients were included in the study. Monitoring our patients for 18 months, we observed a reduction in nasal polyposis and an improvement in symptoms and their impact on quality of life. Regarding blood tests, a transient increase in blood eosinophils was found in most cases. Total IgE showed a gradual decrease in values. IgG and IgA also showed a slight reduction of values, while remaining within normal ranges. Conclusion To the best of our knowledge, this is the first study to evaluate the impact of dupilumab on several blood parameters in patients receiving treatment for CRswNP. Further studies are needed to confirm our results and to understand the underlying immunological mechanisms.
Collapse
Affiliation(s)
| | - Andrea Ciofalo
- Department of Sense Organs, Sapienza University, Rome, Italy
| | - Carlo Cavaliere
- Department of Sense Organs, Sapienza University, Rome, Italy
| | - Elona Begvarfaj
- Department of Sense Organs, Sapienza University, Rome, Italy
| | | | - Giacomo Alfonzo
- Department of Sense Organs, Sapienza University, Rome, Italy
| | - Rosalba Cadeddu
- Department of Sense Organs, Sapienza University, Rome, Italy
| | | | | | - Antonio Greco
- Department of Sense Organs, Sapienza University, Rome, Italy
| | | | - Simonetta Masieri
- Department of Oral and Maxillofacial Sciences, Sapienza University, Rome, Italy
| |
Collapse
|
50
|
Chen Z, Qiao S, Yang L, Sun M, Li B, Lu A, Li F. Mechanistic Insights into the Roles of the IL-17/IL-17R Families in Pancreatic Cancer. Int J Mol Sci 2023; 24:13539. [PMID: 37686343 PMCID: PMC10487659 DOI: 10.3390/ijms241713539] [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: 07/24/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The members of the cytokine interleukin 17 (IL-17) family, along with their receptors (IL-17R), are vital players in a range of inflammatory diseases and cancer. Although generally regarded as proinflammatory, the effects they exhibit on cancer progression are a double-edged sword, with both antitumor and protumor activities being discovered. There is growing evidence that the IL-17 signaling pathways have significant impacts on the tumor microenvironment (TME), immune response, and inflammation in various types of cancer, including pancreatic cancer. However, the detailed mechanistic functions of the IL-17/IL-17R families in pancreatic cancer were rarely systematically elucidated. This review considers the role of the IL-17/IL-17R families in inflammation and tumor immunity and elaborates on the mechanistic functions and correlations of these members with pathogenesis, progression, and chemoresistance in pancreatic cancer. By summarizing the advanced findings on the role of IL-17/IL17R family members and IL-17 signaling pathways at the molecular level, cellular level, and disease level in pancreatic cancer, this review provides an in-depth discussion on the potential of IL-17/IL-17R as prognostic markers and therapeutic targets in pancreatic cancer.
Collapse
Affiliation(s)
- Zheng Chen
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shuangying Qiao
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Liu Yang
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Meiheng Sun
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Boyue Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aiping Lu
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Fangfei Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|