1
|
Fonta N, Page N, Klimek B, Piccinno M, Di Liberto G, Lemeille S, Kreutzfeldt M, Kastner AL, Ertuna YI, Vincenti I, Wagner I, Pinschewer DD, Merkler D. Oligodendrocyte-derived IL-33 regulates self-reactive CD8+ T cells in CNS autoimmunity. J Exp Med 2025; 222:e20241188. [PMID: 40227193 PMCID: PMC11995930 DOI: 10.1084/jem.20241188] [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: 07/11/2024] [Revised: 02/11/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
In chronic inflammatory disorders of the central nervous system (CNS), tissue-resident self-reactive T cells perpetuate disease. The specific tissue factors governing the persistence and continuous differentiation of these cells remain undefined but could represent attractive therapeutic targets. In a model of chronic CNS autoimmunity, we find that oligodendrocyte-derived IL-33, an alarmin, is key for locally regulating the pathogenicity of self-reactive CD8+ T cells. The selective ablation of IL-33 from neo-self-antigen-expressing oligodendrocytes mitigates CNS disease. In this context, fewer self-reactive CD8+ T cells persist in the inflamed CNS, and the remaining cells are impaired in generating TCF-1low effector cells. Importantly, interventional IL-33 blockade by locally administered somatic gene therapy reduces T cell infiltrates and improves the disease course. Our study identifies oligodendrocyte-derived IL-33 as a druggable tissue factor regulating the differentiation and survival of self-reactive CD8+ T cells in the inflamed CNS. This finding introduces tissue factors as a novel category of immune targets for treating chronic CNS autoimmune diseases.
Collapse
Affiliation(s)
- Nicolas Fonta
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Nicolas Page
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Bogna Klimek
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Margot Piccinno
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Giovanni Di Liberto
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Sylvain Lemeille
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Anna Lena Kastner
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Yusuf I. Ertuna
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ilena Vincenti
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Daniel D. Pinschewer
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| |
Collapse
|
2
|
Luo H, Liu L, Liu X, Xie Y, Huang X, Yang M, Shao C, Li D. Interleukin-33 (IL-33) promotes DNA damage-resistance in lung cancer. Cell Death Dis 2025; 16:274. [PMID: 40216748 PMCID: PMC11992107 DOI: 10.1038/s41419-025-07624-x] [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: 11/18/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
Resistance to DNA damage is one of the primary mechanisms by which tumor cells evade the effects of standard chemotherapeutic agents and radiotherapy. Dynamic and complex interactions between the tumor microenvironment (TME) and tumor cells critically influence the DNA damage response. Interleukin-33 (IL-33) is a multifunctional cytokine secreted at high levels in response to cellular damage and stress. Recently, increasing evidence has suggested that IL-33 plays a key role in promoting the therapeutic resistance of tumors. However, the actual source of IL-33 during cancer therapy and how IL-33 contributes to a resistant TME remain incompletely understood. In this study, we found that both cancer-associated fibroblasts (CAFs) and tumor cells treated with DNA damage-inducing agents expressed and secreted high levels of IL-33, subsequently leading to enhanced DNA damage repair efficacy. Mechanistically, nuclear IL-33 primarily functions as a transcriptional co-activator of homologous recombination repair (HRR) genes, whereas the active form of IL-33 can drive the non-homologous end joining (NHEJ) pathway via the canonical IL-33/ST2 axis. Overall, we demonstrated that IL-33 plays a key role in mediating a DNA damage-resistant TME, which could represent a potential therapeutic vulnerability in chemoresistant cancer cells.
Collapse
Affiliation(s)
- Haoge Luo
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Liping Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaoping Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yingdong Xie
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xin Huang
- Department of Microbiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Ming Yang
- Department of Biochemistry & Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
- Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Chen Shao
- Department of Biochemistry & Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China.
- Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China.
| | - Dong Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China.
- Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China.
| |
Collapse
|
3
|
Radhouani M, Farhat A, Hakobyan A, Zahalka S, Pimenov L, Fokina A, Hladik A, Lakovits K, Brösamlen J, Dvorak V, Nunes N, Zech A, Idzko M, Krausgruber T, Köhl J, Uluckan O, Kovarik J, Hoehlig K, Vater A, Eckhard M, Sombke A, Fortelny N, Menche J, Knapp S, Starkl P. Eosinophil innate immune memory after bacterial skin infection promotes allergic lung inflammation. Sci Immunol 2025; 10:eadp6231. [PMID: 40184438 DOI: 10.1126/sciimmunol.adp6231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 11/22/2024] [Accepted: 02/27/2025] [Indexed: 04/06/2025]
Abstract
Microbial exposure at barrier interfaces drives development and balance of the immune system, but the consequences of local infections for systemic immunity and secondary inflammation are unclear. Here, we show that skin exposure to the bacterium Staphylococcus aureus persistently shapes the immune system of mice with specific impact on progenitor and mature bone marrow neutrophil and eosinophil populations. The infection-imposed changes in eosinophils were long-lasting and associated with functional as well as imprinted epigenetic and metabolic changes. Bacterial exposure enhanced cutaneous allergic sensitization and resulted in exacerbated allergen-induced lung inflammation. Functional bone marrow eosinophil reprogramming and pulmonary allergen responses were driven by the alarmin interleukin-33 and the complement cleavage fragment C5a. Our study highlights the systemic impact of skin inflammation and reveals mechanisms of eosinophil innate immune memory and organ cross-talk that modulate systemic responses to allergens.
Collapse
Affiliation(s)
- Mariem Radhouani
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Asma Farhat
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anna Hakobyan
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, Vienna, Austria
| | - Sophie Zahalka
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lisabeth Pimenov
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Alina Fokina
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Anastasiya Hladik
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Karin Lakovits
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Jessica Brösamlen
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | | | - Natalia Nunes
- Center for Tumor Biology and Immunology, Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Andreas Zech
- Department of Medicine II, Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Department of Medicine II, Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Thomas Krausgruber
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ozge Uluckan
- Novartis Biomedical Research, Basel, Switzerland
| | - Jiri Kovarik
- Novartis Biomedical Research, Basel, Switzerland
| | | | | | - Margret Eckhard
- Center for Anatomy and Cell Biology, Cell and Developmental Biology, Medical University of Vienna, Vienna, Austria
| | - Andy Sombke
- Center for Anatomy and Cell Biology, Cell and Developmental Biology, Medical University of Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- Center for Tumor Biology and Immunology, Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Jörg Menche
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, Vienna, Austria
- Faculty of Mathematics, University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Network Medicine at the University of Vienna, Vienna, Austria
| | - Sylvia Knapp
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Vienna, Austria
| | - Philipp Starkl
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Akenroye A, Boyce JA, Kita H. Targeting alarmins in asthma: From bench to clinic. J Allergy Clin Immunol 2025; 155:1133-1148. [PMID: 39855362 DOI: 10.1016/j.jaci.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/24/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Over the past 2 decades, mechanistic studies of allergic and type 2 (T2)-mediated airway inflammation have led to multiple approved therapies for the treatment of moderate-to-severe asthma. The approval and availability of these monoclonal antibodies targeting IgE, a T2 cytokine (IL-5) and/or cytokine receptors (IL-5Rα, IL-4Rα) has been central to the progresses made in the management of moderate-to-severe asthma over this period. However, there are persistent gaps in clinician's ability to provide precise care, given that many patients with T2-high asthma do not respond to IgE- or T2 cytokine-targeting therapies and that patients with T2-low asthma have few therapeutic options. The new frontier of precision medicine in asthma, as well as in other allergic diseases, includes the targeting of epithelium-derived cytokines known as alarmins, including thymic stromal lymphopoietin, IL-25, IL-33, and their receptors. The effects of these alarmins, which can act upstream of immune cells, involve both the innate and adaptive systems and hold potential for the treatment of both T2-high and -low disease. Tezepelumab, an anti-thymic stromal lymphopoietin antibody, has already been approved for the treatment of severe asthma. In this review, we discuss our current understanding of alarmin biology with a primary focus on allergic airway diseases. We link the mechanistic corollaries to the clinical implications and advances in drug development targeting alarmins, with a particular focus on currently approved treatments, those under study, and future potential targets in alarmin signaling pathways.
Collapse
Affiliation(s)
- Ayobami Akenroye
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| | - Joshua A Boyce
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, the Department of Medicine, and the Department of Immunology, Mayo Clinic Arizona, Scottsdale, Ariz; Department of Immunology, Mayo Clinic Rochester, Rochester, Minn
| |
Collapse
|
5
|
Ojha S, Sinsinwar S, Chatterjee P, Biswal S, Pradhan P, Beuria TK. Efflux pump modulation by Montelukast and its roles in restoring antibiotic susceptibility in multidrug-resistant Staphylococcus aureus. EBioMedicine 2025; 114:105658. [PMID: 40157128 PMCID: PMC11994358 DOI: 10.1016/j.ebiom.2025.105658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/22/2025] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Staphylococcus aureus and its drug-resistant mutants are mentioned among the WHO's high-priority list of pathogens. Antibiotics like fluoroquinolones and cephalosporins are used to treat multidrug-resistant S. aureus infections. However, a higher expression of efflux pumps (NorA, NorB, and AbcA) induces multidrug resistance. The master regulator, MgrA, regulates the expression of most of these efflux pumps in S. aureus. The phosphorylation status of MgrA is determined by the cellular PknB/RsbU ratio, where PknB, a serine-threonine kinase, and RsbU, a serine-threonine phosphatase, are critical for MgrA functioning. METHODS An FDA-approved drug library was screened using an EtBr-accumulation assay to identify efflux pump inhibitors (EPIs). The synergy of EPIs with antibiotics was studied in vitro and in vivo in the murine skin infection model of female BALB/c mice. The effect of EPIs on mgrA, norB, pknB, and rsbU gene expression, interaction with MgrA, and effects on MgrA phosphorylation were studied. FINDINGS We identified Montelukast as an effective EPI, which showed synergy with moxifloxacin, a substrate of the NorB efflux pump, both in vitro and in the murine skin infection model. Further, Montelukast decreased norB expression and increased the pknB/rsbU expression ratio. Our in vitro results demonstrated that Montelukast strongly interacted with MgrA, facilitated MgrA phosphorylation, and enhanced its affinity for the norB promoter. INTERPRETATION Our study showed that Montelukast repressed MgrA expression and promoted MgrA phosphorylation to suppress norB expression and efflux pump activity, leading to the restoration of antibiotic susceptibility in multidrug-resistant S. aureus. FUNDING The study was supported by SERB-DST, India (CRG/2021/005069), and the BRIC-ILS core.
Collapse
Affiliation(s)
- Suvendu Ojha
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India; Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad Rd, Faridabad, Haryana, 121001, India
| | - Simran Sinsinwar
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - Puja Chatterjee
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India; Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad Rd, Faridabad, Haryana, 121001, India
| | - Sarmistha Biswal
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - Pinkilata Pradhan
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India; Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad Rd, Faridabad, Haryana, 121001, India
| | - Tushar Kant Beuria
- Department of Infectious Disease Biology, Institute of Life Sciences, NALCO Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
6
|
Jean EE, Rossi HL, Hung LY, Inclan-Rico JM, Herbert DR. Myeloid-derived IL-33 drives γδ T cell-dependent resistance against cutaneous infection by Strongyloides ratti. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae038. [PMID: 40073150 PMCID: PMC11952876 DOI: 10.1093/jimmun/vkae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/05/2024] [Indexed: 03/14/2025]
Abstract
Interleukin 33 (IL-33) is a pleiotropic cytokine released from diverse cell types that regulate both pro- and anti-inflammatory responses during pathogen infection. However, it remains unclear whether IL-33 controls key aspects of cutaneous immunity against skin-penetrating parasites. In this study, mice percutaneously infected with the parasitic helminth Strongyloides ratti were investigated to understand mechanisms of anamnestic immunity at the skin barrier. Surprisingly, mice lacking the Type 2 transcription factor STAT6 (signal transducer and activator of transcription 6) had no defects in secondary resistance to infection, whereas IL-33 gene deficiency or local blockade of IL-33 receptor (ST2) signaling abrogated host resistance. Depletion of CD4+ T cells or type 2 innate lymphoid cells had only a moderate impact on protection, but the loss of γδ T cells completely ablated cutaneous immunity against rechallenge. We identified a CD62Lhi IL-33 receptor (ST2)-expressing γδ T cell population that accumulated in the skin of protected mice that was dependent upon IL-33 expression in myeloid lineage antigen-presenting cells. This work suggests a previously unrecognized mechanism wherein noncanonical type 2 immunity operates through myeloid antigen-presenting cells and skin γδ T cells to adaptively repel skin-penetrating helminth larvae.
Collapse
Affiliation(s)
- Erin Evonne Jean
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Heather Lynn Rossi
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Li Yin Hung
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Juan M Inclan-Rico
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - De’Broski R Herbert
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| |
Collapse
|
7
|
Cook DP, Peebles RS. The Cystic Fibrosis Transmembrane Conductance Receptor Brakes Allergic Airway Inflammation. Immunol Rev 2025; 330:e70009. [PMID: 39981881 PMCID: PMC11843850 DOI: 10.1111/imr.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
Cystic fibrosis (CF) is a common autosomal recessive disease resulting from mutations of the gene that encodes the cystic fibrosis transmembrane conductance regulator (CFTR). Although severe pulmonary neutrophilic inflammation is a primary pathologic feature of CF, more recent studies reveal a role for type 2 inflammation that is characterized by eosinophilia directed by both the innate and adaptive immune systems through ILC2 and CD4+ Th2 cells, respectively. We have published that a clear type endotype exists within CF subjects stratified by Th2 inflammation, defined by increased obstructive pulmonary disease and a distinct phenotypic signature of increased allergic disease, infections, and burden of CF complications. Further, we showed an increased risk of death among CF subjects with type 2 inflammatory signatures compared to CF subjects lacking significant type 2 inflammation. The mechanisms of this heightened type 2 inflammatory signature in CF are still being defined, but it is clear that airway epithelial cells from CFTR-deficient mice have increased expression and release of IL-33, a key activator of ILC2 and Th2 cells, compared to persons with normal CFTR function. Further, there is strong evidence that CF regulates CD4+ Th2 function in a cell-intrinsic fashion. These concepts are explored in this review article.
Collapse
Affiliation(s)
- Daniel P. Cook
- Division of Pulmonary, Critical Care and Occupational MedicineUniversity of Iowa Carver School of MedicineIowa CityIowaUSA
| | - R. Stokes Peebles
- Division of Allergy, Pulmonary, and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Pathology, Microbiology, and ImmunologyVanderbilt University School of MedicineNashvilleTennesseeUSA
- United States Department of Veterans AffairsNashvilleTennesseeUSA
| |
Collapse
|
8
|
Rihar M, Bahri R, Forstnerič V, Bulfone‐Paus S, Korošec P. CCL2/C-C chemokine receptor type 2-mediated interactions among mast cells, basophils, and endothelial cells. Clin Transl Allergy 2025; 15:e70044. [PMID: 39988712 PMCID: PMC11847651 DOI: 10.1002/clt2.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND IL-33 is involved in allergic processes by promoting the release of various mast cell (MC) chemokines, including CCL2. However, it is yet unclear which specific cell type is primarily responsible for producing CCL2 during acute allergic reactions. This study aims to investigate the role of IL-33 in promoting CCL2 production in mast cells and assess the effect of MC-derived CCL2 on basophil migration and endothelial permeability. METHODS Human blood-derived MCs (hMCs) were generated from peripheral blood precursors, passively sensitized with IgE, treated with IL-33, and stimulated with anti-IgE. The concentrations of nine cytokines known to influence immune cell chemotaxis (CCL2, CCL5, CCL11, MIP-1α, IL-8, IL-10, IL-13, granulocyte-macrophage colony-stimulating factor (GM-CSF), and vascular endothelial growth factor (VEGF) were assessed in the supernatants of hMCs. Subsequently, we investigated the impact of MC-derived CCL2 on basophil migration in vitro, as well as its effect on endothelial monolayer permeability using human umbilical vein endothelial cells (HUVECs). RESULTS Stimulation with anti-IgE induced a significant release of CCL2, GM-CSF, IL-8 and VEGF from hMCs. Additionally, incubation with IL-33 overnight increased the production of several cytokines. Mast cell-derived CCL2 not only enhanced basophil migration in vitro but also increased endothelial monolayer permeability in HUVECs. The effect was reversed by a C-C chemokine receptor type 2 (CCR2) antagonist, indicating the involvement of CCL2 signaling through the CCR2 receptor. CONCLUSIONS IL-33 induces the production of chemotactic cytokines in hMCs. Mast cell-derived CCL2 plays an important role in basophil chemotaxis in vitro and affects endothelial monolayer permeability in the HUVEC model.
Collapse
Affiliation(s)
- Maruša Rihar
- University Clinic of Respiratory and Allergic Diseases GolnikGolnikSlovenia
- Biotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Rajia Bahri
- Lydia Becker Institute of Immunology and InflammationDivision of Musculoskeletal and Dermatological SciencesSchool of Biological SciencesUniversity of ManchesterManchesterUK
| | - Vida Forstnerič
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
| | - Silvia Bulfone‐Paus
- Lydia Becker Institute of Immunology and InflammationDivision of Musculoskeletal and Dermatological SciencesSchool of Biological SciencesUniversity of ManchesterManchesterUK
| | - Peter Korošec
- University Clinic of Respiratory and Allergic Diseases GolnikGolnikSlovenia
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| |
Collapse
|
9
|
Hassen HY, Govarts E, Remy S, Cox B, Iszatt N, Portengen L, Covaci A, Schoeters G, Den Hond E, Henauw SD, Bruckers L, Koppen G, Verheyen VJ. Association of environmental pollutants with asthma and allergy, and the mediating role of oxidative stress and immune markers in adolescents. ENVIRONMENTAL RESEARCH 2025; 265:120445. [PMID: 39586518 PMCID: PMC11672208 DOI: 10.1016/j.envres.2024.120445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Asthma and allergic diseases are among the common causes of morbidity and mortality globally. Various environmental pollutants are linked to the development of asthma and allergic diseases. Evidence on the role of oxidative stress and immune markers in the association of environmental pollutants with asthma and allergy is scant. We examined cross-sectional associations between environmental pollutants and asthma and allergy, investigated mixture effects and possible mediation by oxidative stress or immune markers. METHODS We used data from the Flemish Environment and Health Study 2016-2020 (FLEHS IV), including 409 adolescents aged 13-16 years. Fifty-four pollutants, including metals, phthalates, Di(isononyl) cyclohexane-1,2-dicarboxylate (DINCH), bisphenols, currently used and legacy pesticides, flame retardants, per- and polyfluoroalkyl substances (PFAS), polyaromatic hydrocarbons (PAHs), and polychlorinated biphenyls (PCBs) were analyzed. Outcomes were self-reported asthma, rhinitis, eczema, allergies, respiratory infection, and airway inflammation, measured through fractional exhaled nitric oxide (FeNO). Single pollutant models using multiple regression analysis and multipollutant models using Bayesian Kernel Machine Regression (BKMR) were fitted. As sensitivity analysis, Bayesian model averaging (BMA) and elastic net (ENET) models were also performed. For Bayesian models, posterior inclusion probabilities (PIP) were used to identify the most important chemicals. Mediation analysis was performed to investigate the role of oxidative stress, measured by urinary 8-hydroxy-2' -deoxyguanosine (8-OHdG), and immune markers (eosinophils, basophils, InterLeukin 8, InterLeukin 6, and Interferon-ᵧ in blood). RESULTS In single pollutant models, FeNO was significantly higher by 20% (95% CI: 6, 36%) and 13% (95% CI: 2, 25%) per interquartile range (IQR) fold in mono-n-butyl phthalate (MnBP) and mono-benzyl phthalate (MBzP), respectively. In BKMR analysis, the group PIPs indicated phthalates and DINCH as the most important group (group PIP = 0.509), with MnBP being the most important pollutant within that group (conditional PIP = 0.564; %change = 28%; 95%CI: 6, 54%). Similar patterns were observed in all multipollutant models. Eosinophil count mediated 37.8% (p = 0.018) and 27.9% (p = 0.045) of the association between MBzP and FeNO, and the association between MnBP and FeNO, respectively. 8-OHdG plays a significant mediating role in the association of 2,4-Dichlorophenoxyacetic acid (2,4-D) (55.4%), 3,5,6-Trichloro-2-pyridinol (TCPY) (48.1%), and 1-Naphthylamine (1-NAP) (32.7%) with rhinitis, while the total effects of these chemicals on rhinitis were not statistically significant. CONCLUSIONS This study found associations between phthalates, MnBP and MBzP, and elevated FeNO, which appeared to be mediated by eosinophil count. 8-OHdG plays a significant mediating role in the association between 2,4-D, TCPY, and 1-NAP with rhinitis, while their direct effects remain non-significant. Use of inflammatory and oxidative stress markers can enhance the understanding of inflammatory processes in asthma and allergic diseases due to environmental pollutants.
Collapse
Affiliation(s)
- Hamid Y Hassen
- Environmental Intelligence Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400, Mol, Belgium.
| | - Eva Govarts
- Environmental Intelligence Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400, Mol, Belgium
| | - Sylvie Remy
- Environmental Intelligence Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400, Mol, Belgium
| | - Bianca Cox
- Environmental Intelligence Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400, Mol, Belgium
| | - Nina Iszatt
- Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway; Centre for Sustainable Diets, Norwegian Institute of Public Health, Oslo, Norway
| | - Lützen Portengen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Adrian Covaci
- Toxicological Center, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Greet Schoeters
- Toxicological Center, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium; Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Elly Den Hond
- Provincial Institute of Hygiene (PIH), Kronenburgstraa 45, 2000, Antwerpen, Belgium; Family Medicine and Population health, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Stefaan De Henauw
- Department of Public Health and Primary Care, Ghent university, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Liesbeth Bruckers
- BioStat, Data Science Institute, Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Gudrun Koppen
- Environmental Intelligence Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400, Mol, Belgium
| | - Veerle J Verheyen
- Environmental Intelligence Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400, Mol, Belgium
| |
Collapse
|
10
|
Obata‐Ninomiya K, Jayaraman T, Ziegler SF. From the bench to the clinic: basophils and type 2 epithelial cytokines of thymic stromal lymphopoietin and IL-33. Clin Transl Immunology 2024; 13:e70020. [PMID: 39654685 PMCID: PMC11626414 DOI: 10.1002/cti2.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024] Open
Abstract
Type 2 epithelial cytokines, including thymic stromal lymphopoietin and IL-33, play central roles in modulation of type 2 immune cells, such as basophils. Basophils are a small subset of granulocytes within the leukocyte population that predominantly exist in the blood. They have non-redundant roles in allergic inflammation in peripheral tissues such as the lung, skin and gut, where they increase and accumulate at inflammatory lesions and exclusively produce large amounts of IL-4, a type 2 cytokine. These inflammatory reactions are known to be, to some extent, phenocopies of infectious diseases of ticks and helminths. Recently, biologics related to both type 2 epithelial cytokines and basophils have been approved by the US Food and Drug Administration for treatment of allergic diseases. We summarised the roles of Type 2 epithelial cytokines and basophils in basic science to translational medicine, including recent findings.
Collapse
Affiliation(s)
| | | | - Steven F Ziegler
- Center of Fundamental ImmunologyBenaroya Research InstituteSeattleWAUSA
- Department of ImmunologyUniversity of Washington School of MedicineSeattleWAUSA
| |
Collapse
|
11
|
Wang H, Wang T, Yan S, Tang J, Zhang Y, Wang L, Xu H, Tu C. Crosstalk of pyroptosis and cytokine in the tumor microenvironment: from mechanisms to clinical implication. Mol Cancer 2024; 23:268. [PMID: 39614288 PMCID: PMC11607834 DOI: 10.1186/s12943-024-02183-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
In the realm of cancer research, the tumor microenvironment (TME) plays a crucial role in tumor initiation and progression, shaped by complex interactions between cancer cells and surrounding non-cancerous cells. Cytokines, as essential immunomodulatory agents, are secreted by various cellular constituents within the TME, including immune cells, cancer-associated fibroblasts, and cancer cells themselves. These cytokines facilitate intricate communication networks that significantly influence tumor initiation, progression, metastasis, and immune suppression. Pyroptosis contributes to TME remodeling by promoting the release of pro-inflammatory cytokines and sustaining chronic inflammation, impacting processes such as immune escape and angiogenesis. However, challenges remain due to the complex interplay among cytokines, pyroptosis, and the TME, along with the dual effects of pyroptosis on cancer progression and therapy-related complications like cytokine release syndrome. Unraveling these complexities could facilitate strategies that balance inflammatory responses while minimizing tissue damage during therapy. This review delves into the complex crosstalk between cytokines, pyroptosis, and the TME, elucidating their contribution to tumor progression and metastasis. By synthesizing emerging therapeutic targets and innovative technologies concerning TME, this review aims to provide novel insights that could enhance treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Tao Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Shuxiang Yan
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jinxin Tang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yibo Zhang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Liming Wang
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410011, China.
| | - Haodong Xu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Shenzhen Research Institute of Central South University, Guangdong, 518063, China.
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
12
|
Feng H, Qin Y, Li Y, Li S, Zheng Y, Yan J, Xu R, Yu S, Liang R, Wang J, Zou H. Research Trend and Future Perspectives of Traditional Chinese Medicine for Atopic Dermatitis from 2004 to 2023: A Bibliometric Analysis. Clin Cosmet Investig Dermatol 2024; 17:2595-2613. [PMID: 39600528 PMCID: PMC11588668 DOI: 10.2147/ccid.s479632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
Aim The application of bibliometric analysis gives insights into research trend and future perspectives. This paper aims to understand the current status of studies on traditional Chinese medicine (TCM) in the treatment of atopic dermatitis (AD), and to investigate the cooperation network, hotspots and trends in the field. Methods The original data were retrieved from the Web of Science Core Collection by searching for the topic "atopic dermatitis" and "traditional Chinese medicine" with the time span of 2004-2023. Microsoft Excel was used to create statistical tables and charts, whereas CiteSpace and VOSViewer were applied for knowledge mapping. Results In total, 252 publications were identified and downloaded. Current research on TCM treating AD is in the steady growth period. China has the largest number of publications, whereas the USA has the greatest international influence. Notably, the Arab Emirates, Malaysia, India and Bangladesh all have a high research impact but with a low amount of publications. Universities in Hong Kong China have the largest number of publications, but their interaction with other institutions is very poor. The top three most productive authors are all from Hong Kong, having a strong collaboration network with each other. The popular research topics in this field are "children" "cell" "activation" "nf kappa b" "inflammation" "asthma" "extract" "management" and "cytokine". According to the analysis of leading co-cited journals and authors, "J ETHNOPHARMACOL" has the largest amount of citations whilst "AM J CHINESE MED" the strongest academic influence. The largest number of co-cited authors comes from "Hon Karn-Lun Ellis", a well-known scholar from Hong Kong. Conclusion TCM has been widely used in AD treatment, honored for its individualized therapeutic concepts of evidence-based treatment. The academic community must form a continuous and deep-going pattern to achieve higher international influence and a stronger research degree in this field.
Collapse
Affiliation(s)
- Huishang Feng
- Department of Dermatology, Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, People’s Republic of China
- Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Yeping Qin
- Department of Dermatology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, People’s Republic of China
| | - Yuanwen Li
- Department of Dermatology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Shuo Li
- Department of Basic Products, China Mobile Communications Group Co., Ltd. Government and Enterprise Customer Branch in Beijing, Beijing, People’s Republic of China
| | - Yuyi Zheng
- Ministry of Education Science and Technology Inspection Department, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Juntang Yan
- Ministry of Education Science and Technology Inspection Department, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Rongting Xu
- Ministry of Education Science and Technology Inspection Department, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Shulin Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Ru Liang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Jiayu Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Huiqin Zou
- Ministry of Education Science and Technology Inspection Department, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| |
Collapse
|
13
|
Phugat S, Sharma J, Kumar S, Jain V, Dhua AK, Yadav DK, Agrawal V, Kumar N, Reddy RP, Suravajhala PN, Mathur P, Agarwala S, Goel P. Genetic landscape of congenital pouch colon: systematic review and functional enrichment study. Pediatr Surg Int 2024; 40:314. [PMID: 39557707 DOI: 10.1007/s00383-024-05878-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Despite extensive clinical documentation, few studies have explored the genetic basis of congenital pouch colon (CPC) which is crucial for early detection, personalized treatment, and genetic counselling. OBJECTIVE To compile the information on the genetic basis of CPC and the functional enrichment of underlying molecular pathways. MATERIALS AND METHODS The review was conducted in accordance with PRISMA guidelines. The implicated genes were investigated for underlying molecular pathways. A network was subsequently created on String-database followed by gene-ontology analysis. RESULTS The study included 20 CPC cases and 52 controls (across 4 studies). Numerous variants, including 24 missense SNPs, 63 frameshift variants, and stop-gain/stop-loss mutations in 11 genes were identified. Notable genetic markers included MUC5B, FRG1, and TAF1B, with potential roles in mucosal barrier functions, colonic muscular development, and ribosomal RNA transcription, respectively. Copy number variants and lnc-EPB41-1-1 were also implicated. Genetic hotspots were identified on chromosomes 11, 17 and 16. RGPD2 and RGPD4, contributing to GTPase activator activity and known to be associated with bowel/colon, were differentially expressed. Pathway analysis highlighted Wnt and HOX pathways, with JAG1 and MLL relevant to CPC pathogenesis. CONCLUSION The study integrates genetic evidence and pathway analysis, shedding light on the complex genetic architecture of CPC. While the importance of genetic markers in the etiopathogenesis of CPC is underscored, the need for validating the findings on larger cohorts, diverse populations and through functional studies is suggested.
Collapse
Affiliation(s)
- Shivani Phugat
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Jyoti Sharma
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sourabh Kumar
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vishesh Jain
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Anjan Kumar Dhua
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Devendra Kumar Yadav
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vikesh Agrawal
- Department of Paediatric Surgery, Netaji Subhash Chandra Bose Government Medical College, Jabalpur, India
| | - Neeta Kumar
- Indian Council of Medical Research, New Delhi, India
| | - Ravi P Reddy
- Department of Pediatric Surgery, Grant Government Medical College, Mumbai, Maharashtra, India
| | | | - Praveen Mathur
- Department of Paediatric Surgery, S.M.S. Medical College, Jaipur, India
| | - Sandeep Agarwala
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Prabudh Goel
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
14
|
Chen X, Mo X, Zhang Y, He D, Xiao R, Cheng Q, Wang H, Liu L, Li WW, Xie P. A comprehensive analysis of the differential expression in the hippocampus of depression induced by gut microbiota compared to traditional stress. Gene 2024; 927:148633. [PMID: 38838871 DOI: 10.1016/j.gene.2024.148633] [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: 02/17/2024] [Revised: 05/22/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Depression, which is a disease of heterogeneous etiology, is characterized by high disability and mortality rates. Gut microbiota are associated with the development of depression. To further explore any differences in the mechanisms of depression induced by gut microbiota and traditional stresses, as well as facilitate the development of microbiota-based interventions, a fecal microbiota transplantation (FMT) depression model was made. This was achieved by transplanting feces from major depressive disorder (MDD) patients into germ-free mice. Second, the mechanisms of the depression induced by gut microbiota were analyzed in comparison with those of the depression caused by different forms of stress. It turned out that mice exhibited depressive-like behavior after FMT. Then, PCR array analysis was performed on the hippocampus of the depressed mice to identify differentially expressed genes (DEGs). The KEGG analysis revealed that the pathways of depression induced by gut microbes are closely associated with immuno-inflammation. To determine the pathogenic pathways of physiological stress and psychological stress-induced depression, raw data was extracted from several databases and KEGG analysis was performed. The results from the analysis revealed that the mechanisms of depression induced by physiological and psychological stress are closely related to the regulation of neurotransmitters and energy metabolism. Interestingly, the immunoinflammatory response was distinct across different etiologies that induced depression. The findings showed that gut microbiota dysbiosis-induced depression was mainly associated with adaptive immunity, while physiological stress-induced depression was more linked to innate immunity. This study compared the pathogenesis of depression caused by gut microbiota dysbiosis, and physiological and psychological stress. We explored new intervention methods for depression and laid the foundation for precise treatment.
Collapse
Affiliation(s)
- Xueyi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Xiaolong Mo
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yangdong Zhang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dian He
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rui Xiao
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Qisheng Cheng
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Haiyang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lanxiang Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Wen-Wen Li
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China.
| |
Collapse
|
15
|
Wang H, He K, Liu Y, Yang L, Wang Z, Wang H, Bai C, Liu J, Zhao L, Ma D, Liu Y. Expression and immune infiltration studies of IL-33-ST2-NF-κB signaling pathway in prostate cancer. Prostate 2024; 84:1398-1410. [PMID: 39113225 DOI: 10.1002/pros.24778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND To analyze the expression of interleukin-33 (IL-33), growth-stimulated expression gene 2 (ST2), nuclear factor-kappaB (NF-κB) and immune cell infiltration in prostate cancer, this study aims to provide an experimental basis for the clinical prevention and treatment of prostate cancer. METHODS The expression of IL-33 in PCa tissues was analyzed using TCGA, TIMER and HPA databases. Using the UALCAN database, the systematic exploration of the relationship between IL-33 and various clinicopathological parameters was conducted. The correlation between IL-33 expression and immune cell infiltration was investigated using TIMER, CIBERSORT and GEPIA databases. To verify these analyses, 22 cases of normal prostate (NP), 76 cases of benign prostatic hyperplasia (BPH), and 100 cases of PCa were recruited. Immunohistochemical staining was performed to examine the expression of IL-33, ST2, NF-κB, and the infiltration of immune cells. Correlations between these factors were then determined. RESULTS The expression of IL-33, ST2 and NF-κB was significantly lower in PCa tissues compared with NP (p < 0.05). IL-33 was not associated with age in PCa but showed associations with race, molecular characteristics, lymph node metastatic status, TP53 mutation and tumor grade. Furthermore, IL-33 was associated with immune cell infiltration. Positive correlations were observed between IL-33 and ST2 expressions, as well as between IL-33 and CD68+ macrophages in BPH and PCa. CONCLUSIONS IL-33, ST2 and NF-κB are lowly expressed in PCa tissues, their expression decreases with the increasing malignancy of cancer. IL-33, ST2 and NF-κB are factors associated with PCa immune infiltration. IL-33 has an inhibitory effect on prostate cancer through the IL-33/ST2/NF-κB signalling pathway.
Collapse
Affiliation(s)
- Han Wang
- School of Basic Medical College, Beihua University, Jilin, China
| | - Kang He
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Yuqi Liu
- School of Basic Medical College, Beihua University, Jilin, China
| | - Lijuan Yang
- School of Basic Medical College, Beihua University, Jilin, China
| | - Zhenjiang Wang
- School of Basic Medical College, Beihua University, Jilin, China
| | - Helin Wang
- School of Basic Medical College, Beihua University, Jilin, China
| | - Chengxia Bai
- School of Basic Medical College, Beihua University, Jilin, China
| | - Jian Liu
- People's hospital in Yushu city, Yushu, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Dongrui Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanbo Liu
- School of Basic Medical College, Beihua University, Jilin, China
| |
Collapse
|
16
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
17
|
Kaushal SK, Parul, Tripathi A, Singh DP, Paul A, Alka K, Shukla S, Singh D. IL-33 prevents age-related bone loss and memory impairment by suppression of Th17 response: evidence in a d-galactose-induced aging mouse model. JBMR Plus 2024; 8:ziae101. [PMID: 39224568 PMCID: PMC11365962 DOI: 10.1093/jbmrpl/ziae101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Cytokines are the primary mediators of age-related disorders. The IL-17/IL-10 axis plays a crucial role in bone destruction and neuro-inflammation. Additionally, a new Th2 cytokine-IL-33-has gained attention for its potential implications in aging-associated conditions. However, the involvement of IL-33 in aging-mediated bone loss and memory impairment remains unclear and needs further investigation. This study reveals the impact of IL-33 on various aspects of the immune system, bone health, and neural functions. To induce senescence, we used d-galactose for its convenience and fewer side effects. The experimental design involved treating 20-week-old C57BL/6J mice with d-galactose subcutaneously for 10 weeks to induce aging-like effects. Thereafter, IL-33 recombinant protein was administered intraperitoneally for 15 days to evaluate its impact on various immune, skeletal, and neural parameters. The results demonstrated that d-galactose-induced aging led to bone loss and compromised osteogenic parameters, accompanied by increased oxidative stress and neurodegeneration in specific brain regions. Behavioral activities were also affected. However, supplementation with IL-33 mitigated these effects, elevating osteogenic parameters and reducing senescence markers in osteoblast cells in an aging mouse model and exerted neuroprotective potential. Notably d-galactose-induced aging was characterized by high bone turnover, reflected by altered serum levels of CTX, PTH, beta-galactosidase, and P1NP. IL-33 treatment attenuated these effects, suggesting its role in regulating bone metabolism. Furthermore, d-galactose-induced aging was associated with increased differentiation of Th17 cells and upregulation of associated markers, such as STAT-3 and ROR-γt, while downregulating Foxp3, which antagonizes Th17 cell differentiation. IL-33 treatment countered these effects by suppressing Th17 cell differentiation and promoting IL-10-producing T-regulatory cells. Overall, these findings provide insights into the potential therapeutic implications of IL-33 in addressing aging-induced bone loss and memory impairment.
Collapse
Affiliation(s)
- Saurabh Kumar Kaushal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Parul
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Alok Tripathi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Devendra Pratap Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Ankita Paul
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Kumari Alka
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Shubha Shukla
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Divya Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
18
|
Chen B, Wu Y, Wu H, Gao J, Meng X, Chen H. IBD functions as a double-edged sword for food allergy in BALB/c mice model. Immunology 2024; 173:394-407. [PMID: 39005140 DOI: 10.1111/imm.13833] [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: 01/04/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Inflammatory bowel disease (IBD) and food allergy (FA) increase in tandem, but the potential impact of IBD on FA remains unclear. We sought to determine the role of IBD on FA. We first assessed the changes of FA-related risk factors in dextran sulphate sodium salt (DSS) induced colitis mice model. Then, we evaluated the role of IBD on FA in mice. FA responses were determined using a clinical allergy score, body temperature change, serum antibody levels, cytokines level and mouse mast cell protease 1 (MMCP-1) concentration. Accumulation of regulatory T cells was tested using flow cytometry. Intestinal changes were identified by histology, immunohistochemistry, gene expression and gut microbial community structure. In DSS-induced colitis mice model, we found the intestinal damage, colonic neutrophil infiltration, and downregulation of splenic Th2 cytokines and Tregs in mesenteric lymph nodes (MLN). Moreover, we also found that IBD can alleviate the FA symptoms and lead to the significant downregulation of Th2 cytokines, serum IgE and MMCP-1. However, IBD exacerbates intestinal injury and promotes the gene expression levels of IL-33 and IL-5 in the small intestine, damages the intestinal tissue structure and aggravates intestinal dysbiosis in FA. IBD functions as a double-edged sword in FA. From the perspective of clinical symptoms and humoral immune responses, IBD can reduce FA response by downregulating Th2 cytokines. But from the perspective of the intestinal immune system, IBD potentially disrupts intestinal tolerance to food antigens by damaging intestinal tissue structure and causing intestinal dysbiosis.
Collapse
Affiliation(s)
- Bihua Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Yuhong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Huan Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, China
| |
Collapse
|
19
|
Sugiura K, Fujita H, Komine M, Yamanaka K, Akiyama M. The role of interleukin-36 in health and disease states. J Eur Acad Dermatol Venereol 2024; 38:1910-1925. [PMID: 38779986 DOI: 10.1111/jdv.19935] [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: 11/03/2023] [Accepted: 01/29/2024] [Indexed: 05/25/2024]
Abstract
The interleukin (IL)-1 superfamily upregulates immune responses and maintains homeostasis between the innate and adaptive immune systems. Within the IL-1 superfamily, IL-36 plays a pivotal role in both innate and adaptive immune responses. Of the four IL-36 isoforms, three have agonist activity (IL-36α, IL-36β, IL-36γ) and the fourth has antagonist activity (IL-36 receptor antagonist [IL-36Ra]). All IL-36 isoforms bind to the IL-36 receptor (IL-36R). Binding of IL-36α/β/γ to the IL-36R recruits the IL-1 receptor accessory protein (IL-1RAcP) and activates downstream signalling pathways mediated by nuclear transcription factor kappa B and mitogen-activated protein kinase signalling pathways. Antagonist binding of IL-36Ra to IL-36R inhibits recruitment of IL-1RAcP, blocking downstream signalling pathways. Changes in the balance within the IL-36 cytokine family can lead to uncontrolled inflammatory responses throughout the body. As such, IL-36 has been implicated in numerous inflammatory diseases, notably a type of pustular psoriasis called generalized pustular psoriasis (GPP), a chronic, rare, potentially life-threatening, multisystemic skin disease characterised by recurrent fever and extensive sterile pustules. In GPP, IL-36 is central to disease pathogenesis, and the prevention of IL-36-mediated signalling can improve clinical outcomes. In this review, we summarize the literature describing the biological functions of the IL-36 pathway. We also consider the evidence for uncontrolled activation of the IL-36 pathway in a wide range of skin (e.g., plaque psoriasis, pustular psoriasis, hidradenitis suppurativa, acne, Netherton syndrome, atopic dermatitis and pyoderma gangrenosum), lung (e.g., idiopathic pulmonary fibrosis), gut (e.g., intestinal fibrosis, inflammatory bowel disease and Hirschsprung's disease), kidney (e.g., renal tubulointerstitial lesions) and infectious diseases caused by a variety of pathogens (e.g., COVID-19; Mycobacterium tuberculosis, Pseudomonas aeruginosa, Streptococcus pneumoniae infections), as well as in cancer. We also consider how targeting the IL-36 signalling pathway could be used in treating inflammatory disease states.
Collapse
Affiliation(s)
- Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hideki Fujita
- Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Mayumi Komine
- Department of Dermatology, Faculty of Medicine, Jichi Medical University, Tochigi, Japan
| | - Keiichi Yamanaka
- Department of Dermatology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Masashi Akiyama
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
20
|
Gohal G, Moni SS, Bakkari MA, Elmobark ME. A Review on Asthma and Allergy: Current Understanding on Molecular Perspectives. J Clin Med 2024; 13:5775. [PMID: 39407835 PMCID: PMC11476424 DOI: 10.3390/jcm13195775] [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: 09/03/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Asthma, a complex disease characterized by persistent airway inflammation, remains an urgent global health concern. We explored the critical role of allergic biomarkers and dysregulated immune system in asthma through an extensive literature review in databases such as Web of Science, PubMed, EMBASE, Scopus, and Google Scholar. This review summarizes the growing data on the pivotal role of allergic biomarkers and dysregulated immune system in the development and evolution of asthma. Recent studies have uncovered several biomarkers that elucidate intrinsic allergic mechanisms in individuals with asthma. This article highlights these biomarkers' potential in predicting asthma onset, assessing its intensity, guiding therapeutic interventions, and tracking disease progression. We also explore the innovative therapeutic prospects arising from the convergence of allergy and dysregulated immune system in asthma and emphasize the potential for precision medicine approaches. Understanding allergic biomarkers intertwined with a dysregulated immune system heralds a new era in asthma treatment and points to improved and individualized treatment modalities.
Collapse
Affiliation(s)
- Gassem Gohal
- Department of Pediatrics, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Sivakumar S. Moni
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Mohammed Ali Bakkari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | |
Collapse
|
21
|
Redman E, Fierville M, Cavard A, Plaisant M, Arguel MJ, Ruiz Garcia S, McAndrew EM, Girard-Riboulleau C, Lebrigand K, Magnone V, Ponzio G, Gras D, Chanez P, Abelanet S, Barbry P, Marcet B, Zaragosi LE. Cell Culture Differentiation and Proliferation Conditions Influence the In Vitro Regeneration of the Human Airway Epithelium. Am J Respir Cell Mol Biol 2024; 71:267-281. [PMID: 38843491 PMCID: PMC11376247 DOI: 10.1165/rcmb.2023-0356ma] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 06/06/2024] [Indexed: 07/06/2024] Open
Abstract
The human airway mucociliary epithelium can be recapitulated in vitro using primary cells cultured in an air-liquid interface (ALI), a reliable surrogate to perform pathophysiological studies. As tremendous variations exist among media used for ALI-cultured human airway epithelial cells, the aim of our study was to evaluate the impact of several media (BEGM, PneumaCult, Half & Half, and Clancy) on cell type distribution using single-cell RNA sequencing and imaging. Our work revealed the impact of these media on cell composition, gene expression profile, cell signaling, and epithelial morphology. We found higher proportions of multiciliated cells in PneumaCult-ALI and Half & Half, stronger EGF signaling from basal cells in BEGM-ALI, differential expression of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) entry factor ACE2, and distinct secretome transcripts depending on the media used. We also established that proliferation in PneumaCult-Ex Plus favored secretory cell fate, showing the key influence of proliferation media on late differentiation epithelial characteristics. Altogether, our data offer a comprehensive repertoire for evaluating the effects of culture conditions on airway epithelial differentiation and will aid in choosing the most relevant medium according to the processes to be investigated, such as cilia, mucus biology, or viral infection. We detail useful parameters that should be explored to document airway epithelial cell fate and morphology.
Collapse
Affiliation(s)
- Elisa Redman
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
| | - Morgane Fierville
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
- Interdisciplinary Institute for Artificial Intelligence (3IA Côte d'Azur), Université Côte d'Azur, Sophia Antipolis, France; and
| | - Amélie Cavard
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
| | - Magali Plaisant
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
| | - Marie-Jeanne Arguel
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
| | - Sandra Ruiz Garcia
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
| | - Eamon M McAndrew
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
| | - Cédric Girard-Riboulleau
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
| | - Kevin Lebrigand
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
| | - Virginie Magnone
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
| | - Gilles Ponzio
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
| | - Delphine Gras
- Centre de Recherche en Cardiovasculaire et Nutrition, Institut National de la Santé et de la Recherche Médicale (INSERM), and Institut National de Recherche pour L'agriculture, L'alimentation et L'environnement (INRAE), Université Aix-Marseille, Marseille, France
| | - Pascal Chanez
- Centre de Recherche en Cardiovasculaire et Nutrition, Institut National de la Santé et de la Recherche Médicale (INSERM), and Institut National de Recherche pour L'agriculture, L'alimentation et L'environnement (INRAE), Université Aix-Marseille, Marseille, France
| | - Sophie Abelanet
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
| | - Pascal Barbry
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
- Interdisciplinary Institute for Artificial Intelligence (3IA Côte d'Azur), Université Côte d'Azur, Sophia Antipolis, France; and
| | - Brice Marcet
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
| | - Laure-Emmanuelle Zaragosi
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Côte d'Azur
- IHU RespirERA, and
| |
Collapse
|
22
|
Fadhil SH, Saheb EJ. Relationship between the serum level, polymorphism and gene expression of IL-33 in samples of recurrent miscarriage Iraqi women infected with toxoplasmosis. Exp Parasitol 2024; 263-264:108799. [PMID: 39025462 DOI: 10.1016/j.exppara.2024.108799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 05/20/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
One of the many warm-blooded hosts that toxoplasmosis-causing intracellular protozoan parasite Toxoplasma gondii can infect is humans. Cytokines are crucial to stimulate an effective immune response against T. gondii. Interleukin-33 (IL-33) is a unique anti-inflammatory cytokine that suppresses the immune response. The levels of cytokine gene expression are regulated by genetics, and the genetic polymorphisms of these cytokines play a functional role in this process. Single nucleotide polymorphisms (SNPs) are prognostic indicators of illnesses. This study aimed to determine whether toxoplasmosis interacts with serum levels of IL-33 and its SNP in miscarriage women as well as whether serum levels and IL-33 gene expression are related in toxoplasmosis-positive miscarriage women. Two hundred blood samples from patients and controls were collected from AL-Alawiya Maternity Teaching Hospital and AL-Yarmouk Teaching Hospital in Baghdad, Iraq from 2021 to 2022 in order to evaluate the serum level of IL-33 using ELISA test. For the SNP of IL-33, the allelic high-resolution approach was utilized, and real time-PCR was performed to assess gene expression. The results showed that compared to healthy and pregnant women, recurrent miscarriage with toxoplasmosis and recurrent miscarriage women had lower IL-33 concentrations. Additionally, there were significant differences among healthy women, pregnant women, and women with repeated miscarriage who experienced toxoplasmosis. Furthermore, no differences between patients and controls were revealed by gene expression data. The results revealed that recurrent miscarriage, pregnancy, and healthy women all had a slightly higher amount of the IL-33 gene fold. Additionally, the SNP of IL-33 data demonstrated that there was no significant genetic relationship between patients and controls. Recurrent miscarriage women with toxoplasmosis have showed significant differences from pregnant women in the genotypes GG and AA as well as the alleles A and G. There were notable variations between recurrent miscarriage with and without toxoplasmosis in terms of the genotypes AA and AC. The genotypes GG, AA, and allele A in recurrent miscarriage women with toxoplasmosis and recurrent miscarriage women is a protective factor. Taking together, there was a statistically significant negative correlation between toxoplasmosis and IL-33 gene expression, which calls for more quantitative investigation in order to fully comprehend the interaction of mRNA and protein.
Collapse
Affiliation(s)
- Sabreen Hadi Fadhil
- Department of Biology, Collage of Science, Baghdad University, Baghdad, Iraq.
| | - Entsar Jabbar Saheb
- Department of Biology, Collage of Science, Baghdad University, Baghdad, Iraq
| |
Collapse
|
23
|
Li T, Dou Y, Ji J, Chen H, Zhu S, Wang M, Xiong Y, Wang Z, Shan J, Qian K, An L, Lin L, Wang S, Dai Q. Lipidomics reveals the serum profiles of pediatric allergic rhinitis and its severity. Biomed Chromatogr 2024; 38:e5927. [PMID: 38866427 DOI: 10.1002/bmc.5927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/06/2024] [Accepted: 05/21/2024] [Indexed: 06/14/2024]
Abstract
Allergic rhinitis (AR) is a prevalent upper airway chronic inflammatory disease in children worldwide. The role of bioactive lipids in the regulation of AR has been recognized, but the underlying serum lipidomic basis of its pathology remains unclear. We utilized ultra-performance liquid chromatography (UPLC)-Q-Exactive Orbitrap/mass spectrometry (MS) to investigate the serum lipidomic profiles of children with AR. The lipidomic analysis identified 42 lipids that were differentially expressed (p < 0.05, fold change > 2) between the AR (n = 75) and normal control groups (n = 44). Specifically, the serum levels of diacylglycerol (DG), triacylglycerol (TG), fatty acid (FA), lysophosphatidylcholine (LPC), lysophosphatidylethanolamine, phosphatidyl-ethanolamine, and cardiolipins were significantly higher in the AR group. The diagnostic potential of the identified lipids was further evaluated using receiver operating characteristic curve analysis. The analysis revealed that five lipids, including FA 30:7, LPC O-18:1, LPC 18:0, LPC 16:0, and DG 34:0, had area under the curve values greater than 0.9 (p < 0.05). Furthermore, serum levels of IgE and IL-33, markers of AR severity, were found to have a significant positive correlation (p < 0.05) with DGs, LPCs, TGs, and FAs in AR patients. This study revealed the lipid disorders associated with AR and its severity, providing new insights into the pathological process of AR.
Collapse
Affiliation(s)
- Tao Li
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuzhu Dou
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianjian Ji
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Chen
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaoyun Zhu
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Wang
- Department of Traditional Chinese Medicine, Wuxi Traditional Chinese Medicine Hospital, Wuxi, China
| | - Yingcai Xiong
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhao Wang
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Li An
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lili Lin
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shouchuan Wang
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qigang Dai
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
24
|
Newman M, Connery H, Kannan S, Gautam A, Hammamieh R, Chakraborty N, Boyd J. Fentanyl Overdose Causes Prolonged Cardiopulmonary Dysregulation in Male SKH1 Mice. Pharmaceuticals (Basel) 2024; 17:941. [PMID: 39065791 PMCID: PMC11279777 DOI: 10.3390/ph17070941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Fentanyl overdose is a survivable condition that commonly resolves without chronic overt changes in phenotype. While the acute physiological effects of fentanyl overdose, such as opioid-induced respiratory depression (OIRD) and Wooden Chest Syndrome, represent immediate risks of lethality, little is known about longer-term systemic or organ-level impacts for survivors. In this study, we investigated the effects of a single, bolus fentanyl overdose on components of the cardiopulmonary system up to one week post. SKH1 mice were administered subcutaneous fentanyl at the highest non-lethal dose (62 mg/kg), LD10 (110 mg/kg), or LD50 (135 mg/kg), before euthanasia at 40 min, 6 h, 24 h, or 7 d post-exposure. The cerebral cortex, heart, lungs, and plasma were assayed using an immune monitoring 48-plex panel. The results showed significantly dysregulated cytokine, chemokine, and growth factor concentrations compared to time-matched controls, principally in hearts, then lungs and plasma to a lesser extent, for the length of the study, with the cortex largely unaffected. Major significant analytes contributing to variance included eotaxin-1, IL-33, and betacellulin, which were generally downregulated across time. The results of this study suggest that cardiopulmonary toxicity may persist from a single fentanyl overdose and have wide implications for the endurance of the expanding population of survivors.
Collapse
Affiliation(s)
- Mackenzie Newman
- Department of Orthopaedic Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA;
- Department of Physiology, Pharmacology and Toxicology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA
| | - Heather Connery
- Department of Physiology, Pharmacology and Toxicology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA
| | - Swapna Kannan
- Walter Reed Army Institute of Research, Silver Spring, MD 20907, USA
| | - Aarti Gautam
- Walter Reed Army Institute of Research, Silver Spring, MD 20907, USA
| | - Rasha Hammamieh
- Walter Reed Army Institute of Research, Silver Spring, MD 20907, USA
| | | | - Jonathan Boyd
- Department of Orthopaedic Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA;
- Department of Physiology, Pharmacology and Toxicology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
25
|
Wu L, Zhu X, Luo C, Zhao Y, Pan S, Shi K, Chen Y, Qiu J, Shen Z, Guo J, Jie W. Mechanistic role of RND3-regulated IL33/ST2 signaling on cardiomyocyte senescence. Life Sci 2024; 348:122701. [PMID: 38724005 DOI: 10.1016/j.lfs.2024.122701] [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: 03/20/2024] [Revised: 04/23/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
Hyperinflammatory responses are pivotal in the cardiomyocyte senescence pathophysiology, with IL33 serving as a crucial pro-inflammatory mediator. Our previous findings highlighted RND3's suppressive effect on IL33 expression. This study aims to explore the role of RND3 in IL33/ST2 signaling activation and in cardiomyocyte senescence. Intramyocardial injection of exogenous IL33 reduces the ejection fraction and fractional shortening of rats, inducing the appearance of senescence-associated secretory phenotype (SASP) in myocardial tissues. Recombinant IL33 treatment of AC16 cardiomyocytes significantly upregulated expression of SASP factors like IL1α, IL6, and MCP1, and increased the p-p65/p65 ratio and proportions of SA-β-gal and γH2AX-positive cells. NF-κB inhibitor pyrrolidinedithiocarbamate ammonium (PDTC) and ST2 antibody astegolimab treatments mitigated above effects. RND3 gene knockout H9C2 cardiomyocytes using CRISPR/Cas9 technology upregulated IL33, ST2L, IL1α, IL6, and MCP1 levels, decreased sST2 levels, and increased SA-β-gal and γH2AX-positive cells. A highly possibility of binding between RND3 and IL33 proteins was showed by molecular docking and co-immunoprecipitation, and loss of RND3 attenuated ubiquitination mediated degradation of IL33; what's more, a panel of ubiquitination regulatory genes closely related to RND3 were screened using qPCR array. In contrast, RND3 overexpression in rats by injection of AAV9-CMV-RND3 particles inhibited IL33, ST2L, IL1α, IL6, and MCP1 expression in cardiac tissues, decreased serum IL33 levels, and increased sST2 levels. These results suggest that RND3 expression in cardiomyocytes modulates cell senescence by inhibiting the IL33/ST2/NF-κB signaling pathway, underscoring its potential as a therapeutic target in cardiovascular senescence.
Collapse
Affiliation(s)
- Linxu Wu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China; Public Research Center of Hainan Medical University, Haikou 571199, P.R. China
| | - Xinglin Zhu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Cai Luo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Yangyang Zhao
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Shanshan Pan
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Kaijia Shi
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Yan Chen
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Jianmin Qiu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Zhihua Shen
- Department of Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, P.R. China.
| | - Junli Guo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China.
| | - Wei Jie
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China.
| |
Collapse
|
26
|
Ito K, Ogawa T, Tanigaki T, Kameda K, Hashimoto H, Kawana A, Kimizuka Y. Eosinophilic pleural effusion due to Staphylococcus epidermidis infection: A case report. Respir Med Case Rep 2024; 51:102075. [PMID: 39006194 PMCID: PMC11245978 DOI: 10.1016/j.rmcr.2024.102075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/16/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Eosinophilic pleural effusion is rare, and the cause is often obscure. A 73-year-old man with no relevant medical history presented with exertional dyspnea. Chest imaging revealed left-sided pleural effusion, and pleural fluid examination revealed eosinophilic pleural effusion. Blood tests revealed an increased peripheral blood eosinophil count and elevated Immunoglobulin E levels. Staphylococcus epidermidis was detected in pleural specimens collected via thoracoscopy. Antimicrobial therapy targeting Staphylococcus epidermidis resolved the eosinophilic pleural effusion and elevated peripheral blood eosinophil count. Staphylococcus epidermidis infection may be considered as a cause of eosinophilic pleural effusion when the diagnosis is difficult.
Collapse
Affiliation(s)
- Koki Ito
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takunori Ogawa
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Tomomi Tanigaki
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Koji Kameda
- Division of Thoracic Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hiroshi Hashimoto
- Division of Thoracic Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Akihiko Kawana
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yoshifumi Kimizuka
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| |
Collapse
|
27
|
Jamwal A, Colomb F, McSorley HJ, Higgins MK. Structural basis for IL-33 recognition and its antagonism by the helminth effector protein HpARI2. Nat Commun 2024; 15:5226. [PMID: 38890291 PMCID: PMC11189471 DOI: 10.1038/s41467-024-49550-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
IL-33 plays a significant role in inflammation, allergy, and host defence against parasitic helminths. The model gastrointestinal nematode Heligmosomoides polygyrus bakeri secretes the Alarmin Release Inhibitor HpARI2, an effector protein that suppresses protective immune responses and asthma in its host by inhibiting IL-33 signalling. Here we reveal the structure of HpARI2 bound to mouse IL-33. HpARI2 contains three CCP-like domains, and we show that it contacts IL-33 primarily through the second and third of these. A large loop which emerges from CCP3 directly contacts IL-33 and structural comparison shows that this overlaps with the binding site on IL-33 for its receptor, ST2, preventing formation of a signalling complex. Truncations of HpARI2 which lack the large loop from CCP3 are not able to block IL-33-mediated signalling in a cell-based assay and in an in vivo female mouse model of asthma. This shows that direct competition between HpARI2 and ST2 is responsible for suppression of IL-33-dependent responses.
Collapse
Affiliation(s)
- Abhishek Jamwal
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, South Parks Rd, Oxford, OX1 3QU, UK
| | - Florent Colomb
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK
| | - Henry J McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK.
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, South Parks Rd, Oxford, OX1 3QU, UK.
| |
Collapse
|
28
|
Cheng C, Hsu SK, Chen YC, Liu W, Shu ED, Chien CM, Chiu CC, Chang WT. Burning down the house: Pyroptosis in the tumor microenvironment of hepatocellular carcinoma. Life Sci 2024; 347:122627. [PMID: 38614301 DOI: 10.1016/j.lfs.2024.122627] [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: 11/20/2023] [Revised: 03/20/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
A high mortality rate makes hepatocellular carcinoma (HCC) a difficult cancer to treat. When surgery is not possible, liver cancer patients are treated with chemotherapy. However, HCC management and treatment are difficult. Sorafenib, which is a first-line treatment for hepatocellular carcinoma, initially slows disease progression. However, sorafenib resistance limits patient survival. Recent studies have linked HCC to programmed cell death, which has increased researcher interest in therapies targeting cell death. Pyroptosis, which is an inflammatory mode of programmed cell death, may be targeted to treat HCC. Pyroptosis pathways, executors, and effects are examined in this paper. This review summarizes how pyroptosis affects the tumor microenvironment (TME) in HCC, including the role of cytokines such as IL-1β and IL-18 in regulating immune responses. The use of chemotherapies and their ability to induce cancer cell pyroptosis as alternative treatments and combining them with other drugs to reduce side effects is also discussed. In conclusion, we highlight the potential of inducing pyroptosis to treat HCC and suggest ways to improve patient outcomes. Studies on cancer cell pyroptosis may lead to new HCC treatments.
Collapse
Affiliation(s)
- Chi Cheng
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yen-Chun Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - En-De Shu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ching-Ming Chien
- Department of Medical Sciences Industry, College of Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
29
|
Abdurrahman G, Pospich R, Steil L, Gesell Salazar M, Izquierdo González JJ, Normann N, Mrochen D, Scharf C, Völker U, Werfel T, Bröker BM, Roesner LM, Gómez-Gascón L. The extracellular serine protease from Staphylococcus epidermidis elicits a type 2-biased immune response in atopic dermatitis patients. Front Immunol 2024; 15:1352704. [PMID: 38895118 PMCID: PMC11183529 DOI: 10.3389/fimmu.2024.1352704] [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: 12/08/2023] [Accepted: 05/02/2024] [Indexed: 06/21/2024] Open
Abstract
Background Atopic dermatitis (AD) is a chronic, relapsing inflammatory skin disease with skin barrier defects and a misdirected type 2 immune response against harmless antigens. The skin microbiome in AD is characterized by a reduction in microbial diversity with a dominance of staphylococci, including Staphylococcus epidermidis (S. epidermidis). Objective To assess whether S. epidermidis antigens play a role in AD, we screened for candidate allergens and studied the T cell and humoral immune response against the extracellular serine protease (Esp). Methods To identify candidate allergens, we analyzed the binding of human serum IgG4, as a surrogate of IgE, to S. epidermidis extracellular proteins using 2-dimensional immunoblotting and mass spectrometry. We then measured serum IgE and IgG1 binding to recombinant Esp by ELISA in healthy and AD individuals. We also stimulated T cells from AD patients and control subjects with Esp and measured the secreted cytokines. Finally, we analyzed the proteolytic activity of Esp against IL-33 and determined the cleavage sites by mass spectrometry. Results We identified Esp as the dominant candidate allergen of S. epidermidis. Esp-specific IgE was present in human serum; AD patients had higher concentrations than controls. T cells reacting to Esp were detectable in both AD patients and healthy controls. The T cell response in healthy adults was characterized by IL-17, IL-22, IFN-γ, and IL-10, whereas the AD patients' T cells lacked IL-17 production and released only low amounts of IL-22, IFN-γ, and IL-10. In contrast, Th2 cytokine release was higher in T cells from AD patients than from healthy controls. Mature Esp cleaved and activated the alarmin IL-33. Conclusion The extracellular serine protease Esp of S. epidermidis can activate IL-33. As an antigen, Esp elicits a type 2-biased antibody and T cell response in AD patients. This suggests that S. epidermidis can aggravate AD through the allergenic properties of Esp.
Collapse
Affiliation(s)
- Goran Abdurrahman
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Rebecca Pospich
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Leif Steil
- Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | | | | | - Nicole Normann
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Daniel Mrochen
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Christian Scharf
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Lennart M. Roesner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Lidia Gómez-Gascón
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
30
|
Kim YY, Jeong S, Lee SW, Lee SJ, Rho MC, Kim SH, Lee S. Bakuchicin alleviates ovalbumin-induced allergic asthma by regulating M2 macrophage polarization. Inflamm Res 2024; 73:725-737. [PMID: 38538755 DOI: 10.1007/s00011-024-01859-8] [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: 11/08/2023] [Revised: 12/20/2023] [Accepted: 02/02/2024] [Indexed: 04/30/2024] Open
Abstract
OBJECTIVE Asthma is an airway inflammatory disease caused by activation of numerous immune cells including macrophages. Bakuchicin (BKC) is known to exhibit anti-inflammatory effects and type 2 T helper (Th2) regulation, but has not been investigated for airway inflammation. This study aimed to evaluate the effects of BKC on airway inflammation and demonstrate the mechanisms of macrophage polarization. METHODS The anti-inflammatory effects were determined using lipopolysaccharide (LPS)-stimulated macrophages. The ovalbumin (OVA)-induced asthma mouse model was used to evaluate the effects of BKC on airway inflammation and Th2 responses. Moreover, the effect of BKC on macrophage polarization was confirmed in bone marrow-derived macrophages (BMDMs) differentiation. RESULTS BKC suppressed nitric oxide production and expression of pro-inflammatory cytokines by inhibiting signaling pathway in LPS-stimulated macrophages. In an OVA-induced asthma model, BKC treatment alleviated histological changes and mast cell infiltration and reduced the levels of eosinophil peroxidase, β-hexosaminidase, and immunoglobulin levels. In addition, BKC alleviated Th2 responses and M2 macrophage populations in bronchoalveolar fluid. In BMDMs, BKC suppressed IL-4-induced M2 macrophage polarization and the expression of M2 markers such as arginase-1 and Fizz-1 through inhibiting sirtuin 2 levels. CONCLUSION BKC could be a drug candidate for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Yeon-Yong Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Republic of Korea
- Cell and Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seungwon Jeong
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Republic of Korea
| | - Seung Woong Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Republic of Korea
| | - Seung-Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Republic of Korea
| | - Mun-Chual Rho
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Republic of Korea
| | - Sang-Hyun Kim
- Cell and Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| | - Soyoung Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Republic of Korea.
| |
Collapse
|
31
|
Taruselli MT, Qayum AA, Abebayehu D, Caslin HL, Dailey JM, Kotha A, Burchett JR, Kee SA, Maldonado TD, Ren B, Chao W, Zou L, Haque TT, Straus D, Ryan JJ. IL-33 Induces Cellular and Exosomal miR-146a Expression as a Feedback Inhibitor of Mast Cell Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1277-1286. [PMID: 38381001 PMCID: PMC10984763 DOI: 10.4049/jimmunol.2200916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
IL-33 is an inflammatory cytokine that promotes allergic disease by activating group 2 innate lymphoid cells, Th2 cells, and mast cells. IL-33 is increased in asthmatics, and its blockade suppresses asthma-like inflammation in mouse models. Homeostatic control of IL-33 signaling is poorly understood. Because the IL-33 receptor, ST2, acts via cascades used by the TLR family, similar feedback mechanisms may exist. MicroRNA (miR)-146a is induced by LPS-mediated TLR4 signaling and serves as a feedback inhibitor. Therefore, we explored whether miR-146a has a role in IL-33 signaling. IL-33 induced cellular and exosomal miR-146a expression in mouse bone marrow-derived mast cells (BMMCs). BMMCs transfected with a miR-146a antagonist or derived from miR-146a knockout mice showed enhanced cytokine expression in response to IL-33, suggesting that miR-146a is a negative regulator of IL-33-ST2 signaling. In vivo, miR-146a expression in plasma exosomes was elevated after i.p. injection of IL-33 in wild-type but not mast cell-deficient KitW-sh/W-sh mice. Finally, KitW-sh/W-sh mice acutely reconstituted with miR-146a knockout BMMCs prior to IL-33 challenge had elevated plasma IL-6 levels compared with littermates receiving wild-type BMMCs. These results support the hypothesis that miR-146a is a feedback regulator of IL-33-mediated mast cell functions associated with allergic disease.
Collapse
Affiliation(s)
| | - Amina Abdul Qayum
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Daniel Abebayehu
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Heather L. Caslin
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Jordan M. Dailey
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Aditya Kotha
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Jason R. Burchett
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Sydney A. Kee
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Tania D. Maldonado
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Boyang Ren
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, HSF2 G-S003B, 20 Penn Street, Baltimore, 21201
| | - Wei Chao
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, HSF2 G-S003B, 20 Penn Street, Baltimore, 21201
| | - Lin Zou
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, HSF2 G-S003B, 20 Penn Street, Baltimore, 21201
| | - Tamara T. Haque
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - David Straus
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - John J. Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| |
Collapse
|
32
|
Elmetwalli A, Abdel-Monem MO, El-Far AH, Ghaith GS, Albalawi NAN, Hassan J, Ismail NF, El-Sewedy T, Alnamshan MM, ALaqeel NK, Al-Dhuayan IS, Hassan MG. Probiotic-derived silver nanoparticles target mTOR/MMP-9/BCL-2/dependent AMPK activation for hepatic cancer treatment. Med Oncol 2024; 41:106. [PMID: 38575697 PMCID: PMC10995097 DOI: 10.1007/s12032-024-02330-8] [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: 01/07/2024] [Accepted: 02/08/2024] [Indexed: 04/06/2024]
Abstract
Recent advances in nanotechnology have offered novel ways to combat cancer. By utilizing the reducing capabilities of Lactobacillus acidophilus, silver nanoparticles (AgNPs) are synthesized. The anti-cancer properties of AgNPs have been demonstrated in previous studies against several cancer cell lines; it has been hypothesized that these compounds might inhibit AMPK/mTOR signalling and BCL-2 expression. Consequently, the current research used both in vitro and in silico approaches to study whether Lactobacillus acidophilus AgNPs could inhibit cell proliferation autophagy and promote apoptosis in HepG2 cells. The isolated strain was identified as Lactobacillus acidophilus strain RBIM based on 16 s rRNA gene analysis. Based on our research findings, it has been observed that this particular strain can generate increased quantities of AgNPs when subjected to optimal growing conditions. The presence of silanols, carboxylates, phosphonates, and siloxanes on the surface of AgNPs was confirmed using FTIR analysis. AgNPs were configured using UV-visible spectroscopy at 425 nm. In contrast, it was observed that apoptotic cells exhibited orange-coloured bodies due to cellular shrinkage and blebbing initiated by AgNP treatment, compared to non-apoptotic cells. It is worth mentioning that AgNPs exhibited remarkable selectivity in inducing cell death, specifically in HepG2 cells, unlike normal WI-38 cells. The half-maximum inhibitory concentration (IC50) values for HepG2 and WI-38 cells were 4.217 µg/ml and 154.1 µg/ml, respectively. AgNPs induce an upregulation in the synthesis of inflammation-associated cytokines, including (TNF-α and IL-33), within HepG2 cells. AgNPs co-treatment led to higher glutathione levels and activating pro-autophagic genes such as AMPK.Additionally, it resulted in the suppression of mTOR, MMP-9, BCL-2, and α-SMA gene expression. The docking experiments suggest that the binding of AgNPs to the active site of the AMPK enzyme leads to inhibiting its activity. The inhibition of AMPK ultimately results in the suppression of the mechanistic mTOR and triggers apoptosis in HepG2 cells. In conclusion, the results of our study indicate that the utilization of AgNPs may represent a viable strategy for the eradication of liver cancerous cells through the activation of apoptosis and the enhancement of immune system reactions.
Collapse
Affiliation(s)
- Alaa Elmetwalli
- Department of Clinical Trial Research Unit and Drug Discovery, Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt.
- Microbiology Division, Higher Technological Institute of Applied Health Sciences, Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt.
| | - Mohamed O Abdel-Monem
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt
| | - Gehad S Ghaith
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| | | | - Jihan Hassan
- Department of Applied Medical Chemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Nadia F Ismail
- Health Information Management Program, Biochemistry, Faculty of Health Science Technology, Borg El Arab Technological University, Alexandria, Egypt
| | - Tarek El-Sewedy
- Department of Applied Medical Chemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Mashael Mashal Alnamshan
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| | - Nouf K ALaqeel
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| | - Ibtesam S Al-Dhuayan
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| | - Mervat G Hassan
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| |
Collapse
|
33
|
Wang X, Shields C, Tardo G, Peacock G, Hester E, Anderson M, Williams JM, Cornelius DC. IL-33 supplementation improves uterine artery resistance and maternal hypertension in response to placental ischemia. Am J Physiol Heart Circ Physiol 2024; 326:H1006-H1016. [PMID: 38363211 PMCID: PMC11279736 DOI: 10.1152/ajpheart.00045.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/17/2024]
Abstract
Preeclampsia (PE), a leading cause of maternal/fetal morbidity and mortality, is a hypertensive pregnancy disorder with end-organ damage that manifests after 20 wk of gestation. PE is characterized by chronic immune activation and endothelial dysfunction. Clinical studies report reduced IL-33 signaling in PE. We use the Reduced Uterine Perfusion Pressure (RUPP) rat model, which mimics many PE characteristics including reduced IL-33, to identify mechanisms mediating PE pathophysiology. We hypothesized that IL-33 supplementation would improve blood pressure (BP), inflammation, and oxidative stress (ROS) during placental ischemia. We implanted intraperitoneal mini-osmotic pumps infusing recombinant rat IL-33 (1 µg/kg/day) into normal pregnant (NP) and RUPP rats from gestation day 14 to 19. We found that IL-33 supplementation in RUPP rats reduces maternal blood pressure and improves the uterine artery resistance index (UARI). In addition to physiological improvements, we found decreased circulating and placental cytolytic Natural Killer cells (cNKs) and decreased circulating, placental, and renal TH17s in IL-33-treated RUPP rats. cNK cell cytotoxic activity also decreased in IL-33-supplemented RUPP rats. Furthermore, renal ROS and placental preproendothelin-1 (PPET-1) decreased in RUPP rats treated with IL-33. These findings demonstrate a role for IL-33 in controlling vascular function and maternal BP during pregnancy by decreasing inflammation, renal ROS, and PPET-1 expression. These data suggest that IL-33 may have therapeutic potential in managing PE.NEW & NOTEWORTHY Though decreased IL-33 signaling has been clinically associated with PE, the mechanisms linking this signaling pathway to overall disease pathophysiology are not well understood. This study provides compelling evidence that mechanistically links reduced IL-33 with the inflammatory response and vascular dysfunction observed in response to placental ischemia, such as in PE. Data presented in this study submit the IL-33 signaling pathway as a possible therapeutic target for the treatment of PE.
Collapse
Affiliation(s)
- Xi Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Corbin Shields
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Geilda Tardo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Greg Peacock
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Emily Hester
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Marissa Anderson
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
34
|
Shi Z, Liu Z, Wei Y, Zhang R, Deng Y, Li D. The role of dermal fibroblasts in autoimmune skin diseases. Front Immunol 2024; 15:1379490. [PMID: 38545113 PMCID: PMC10965632 DOI: 10.3389/fimmu.2024.1379490] [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: 01/31/2024] [Accepted: 03/01/2024] [Indexed: 04/18/2024] Open
Abstract
Fibroblasts are an important subset of mesenchymal cells in maintaining skin homeostasis and resisting harmful stimuli. Meanwhile, fibroblasts modulate immune cell function by secreting cytokines, thereby implicating their involvement in various dermatological conditions such as psoriasis, vitiligo, and atopic dermatitis. Recently, variations in the subtypes of fibroblasts and their expression profiles have been identified in these prevalent autoimmune skin diseases, implying that fibroblasts may exhibit distinct functionalities across different diseases. In this review, from the perspective of their fundamental functions and remarkable heterogeneity, we have comprehensively collected evidence on the role of fibroblasts and their distinct subpopulations in psoriasis, vitiligo, atopic dermatitis, and scleroderma. Importantly, these findings hold promise for guiding future research directions and identifying novel therapeutic targets for treating these diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Dong Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Chudakov DB, Shustova OA, Kotsareva OD, Generalov AA, Streltsova MS, Vavilova YD, Fattakhova GV. Chemical chaperone TUDCA selectively inhibits production of allergen-specific IgE in a low-dose model of allergy. BIOMEDITSINSKAIA KHIMIIA 2024; 70:5-14. [PMID: 38450676 DOI: 10.18097/pbmc20247001005] [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: 03/08/2024]
Abstract
The cellular response to endoplasmic reticulum (ER) stress accompanies plasma cell maturation and is one of triggers and cofactors of the local inflammatory response. Chemical chaperones, low-molecular substances that eliminate pathological ER stress, are proposed as means of treating pathologies associated with ER stress. The aim of this study was to evaluate the effect and mechanisms of influence of chemical chaperones on the humoral response in a low-dose model of allergy. The allergic immune response was induced in BALB/c mice by repeated administration of ovalbumin at a dose of 100 ng for 6 weeks. Some animals were injected with both the antigen and the chemical chaperones, TUDCA (tauroursodeoxycholic acid) or 4-PBA (4-phenylbutyrate). Administration of TUDCA, but not 4-PBA, suppressed production of allergen-specific IgE (a 2.5-fold decrease in titer). None of the chemical chaperones affected the production of specific IgG1. The effect of TUDCA was associated with suppression of the switch to IgE synthesis in regional lymph nodes. This phenomenon was associated with suppressed expression of genes encoding cytokines involved in type 2 immune response, especially Il4 and Il9, which in turn could be caused by suppression of IL-33 release. In addition, TUDCA significantly suppressed expression of the cytokine APRIL, and to a lesser extent, BAFF. Thus, TUDCA inhibition of the allergy-specific IgE production is due to suppression of the release of IL-33 and a decrease in the production of type 2 immune response cytokines, as well as suppression of the expression of the cytokines APRIL and BAFF.
Collapse
Affiliation(s)
- D B Chudakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - O A Shustova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - O D Kotsareva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - A A Generalov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - M S Streltsova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Yu D Vavilova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - G V Fattakhova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
36
|
Che K, Luo Y, Song X, Yang Z, Wang H, Shi T, Wang Y, Wang X, Wu H, Yu L, Liu B, Wei J. Macrophages reprogramming improves immunotherapy of IL-33 in peritoneal metastasis of gastric cancer. EMBO Mol Med 2024; 16:251-266. [PMID: 38238529 PMCID: PMC10897402 DOI: 10.1038/s44321-023-00012-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 09/24/2023] [Accepted: 11/20/2023] [Indexed: 02/17/2024] Open
Abstract
Peritoneal metastasis (PM) has a suppressive tumor immune microenvironment (TIME) that limits the effects of immunotherapy. This study aimed to investigate the immunomodulatory effects of intraperitoneal administration of IL-33, a cytokine that is reported to potentiate antitumor immunity and inhibit metastasis. We found survival was significantly prolonged in patients with high IL-33 mRNA expression. In immunocompetent mice, intraperitoneal administration of IL-33 could induce a celiac inflammatory environment, activate immunologic effector cells, and reverse the immunosuppressive tumor microenvironment, which effectively delayed tumor progression and PM of gastric cancer. Mechanistically, IL-33 could induce M2 polarization by activating p38-GATA-binding protein 3 signaling. IL-33 combined with anti-CSF1R or p38 inhibitor to regulate tumor-associated macrophages (TAMs) had a synergistic antitumor effect. Inducing a local inflammatory milieu by IL-33 administration provided a novel approach for treating peritoneal metastasis, which, when combined with TAM reprogramming to reshape TIME, can achieve better treatment efficacy.
Collapse
Affiliation(s)
- Keying Che
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuting Luo
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xueru Song
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhe Yang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hanbing Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tao Shi
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yue Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuan Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hongyan Wu
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lixia Yu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jia Wei
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China.
- Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, Nanjing, China.
| |
Collapse
|
37
|
Huang HT, Tzeng SF. Interleukin-33 has the protective effect on oligodendrocytes against impairment induced by cuprizone intoxication. Neurochem Int 2024; 172:105645. [PMID: 38016520 DOI: 10.1016/j.neuint.2023.105645] [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: 08/19/2023] [Revised: 11/09/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023]
Abstract
Our prior investigations have demonstrated the pivotal role of IL-33 in facilitating the maturation of oligodendrocytes (OLs), prompting our interest in exploring its potential therapeutic effects. In this study, our focus was directed towards deciphering the functions of interleukin-33 (IL-33) in established demyelinating mouse model induced by the feeding of cuprizone (CPZ)-containing diet. We observed the reduction in corpus callosal adenomatous polyposis coli (APC)+ OLs with IL-33 expression in mice subjected to CPZ feeding for durations of 6 and 8 weeks. In parallel, the levels of IL-33 in the corpus callosum declined after CPZ-containing diet. Furthermore, we conducted experiments utilizing primary oligodendrocyte precursor cells (OPCs) and mature OLs, which were exposed to CPZ. A decrease in the expression of myelin basic protein (MBP) was evident in the cultures of mature OLs after treatment with CPZ. Additionally, both IL-33 mRNA and protein levels exhibited downregulation. To counteract the diminished IL-33 levels induced by CPZ, we employed a lentiviral vector to overexpress IL-33 in OLs. Intriguingly, the overexpression of IL-33 (IL33OE) in OLs resulted in a more distinct membranous morphology following CPZ treatment when compared to that observed in OL Mock cultures. Moreover, MBP protein levels in the presence of CPZ were higher in IL33OE OLs than that detected in OL Mock cultures. These findings collectively indicate that IL-33 possesses the capability to mitigate CPZ-induced damage and bolster OL homeostasis. In summary, our study underscores the importance of IL-33 in the context of demyelinating diseases, shedding light on its potential therapeutic implications for fostering remyelination and preserving OL function.
Collapse
Affiliation(s)
- Hui-Ting Huang
- Department of Life Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
38
|
Albrecht M, Garn H, Buhl T. Epithelial-immune cell interactions in allergic diseases. Eur J Immunol 2024; 54:e2249982. [PMID: 37804068 DOI: 10.1002/eji.202249982] [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: 05/17/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/08/2023]
Abstract
Epithelial/immune interactions are characterized by the different properties of the various epithelial tissues, the mediators involved, and the varying immune cells that initiate, sustain, or abrogate allergic diseases on the surface. The intestinal mucosa, respiratory mucosa, and regular skin feature structural differences according to their primary function and surroundings. In the context of these specialized functions, the active role of the epithelium in shaping immune responses is increasingly recognizable. Crosstalk between epithelial and immune cells plays an important role in maintaining homeostatic conditions. While cells of the myeloid cell lineage, mainly macrophages, are the dominating immune cell population in the skin and the respiratory tract, lymphocytes comprise most intraepithelial immune cells in the intestine under healthy conditions. Common to all surface epithelia is the fact that innate immune cells represent the first line of immunosurveillance that either directly defeats invading pathogens or initiates and coordinates more effective successive immune responses involving adaptive immune cells and effector cells. Pharmacological approaches for the treatment of allergic and chronic inflammatory diseases involving epithelial barriers target immunological mediators downstream of the epithelium (such as IL-4, IL-5, IL-13, and IgE). The next generation of therapeutics involves upstream events of the inflammatory cascade, such as epithelial-derived alarmins and related mediators.
Collapse
Affiliation(s)
- Melanie Albrecht
- Molecular Allergology, Vice President´s Research Group, Paul-Ehrlich-Institut, Langen, Germany
| | - Holger Garn
- Translational Inflammation Research Division and Core Facility for Single Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University of Marburg, Marburg, Germany
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
39
|
Yasen A, Yang Z, Feng J, Liang R, Dai T, Li K, Cai Y, Wang G. IL-33/ST2 Signaling and its Correlation with Macrophage Heterogeneity and Clinicopathologic Features in Human Intrahepatic Cholangiocarcinoma. Curr Cancer Drug Targets 2024; 24:1144-1156. [PMID: 38299398 DOI: 10.2174/0115680096276605240108112135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND IL-33/ST2 signaling plays crucial roles in the development and progression of various human malignancies. However, its significance in intrahepatic cholangiocarcinoma (ICC) still remains unclear. OBJECTIVE This study aimed to investigate the expression of IL-33/ST2 signaling and its correlations with macrophage heterogeneity and ICC patients' clinicopathologic features. METHODS The expression of different phenotype macrophage markers and IL-33/ST2 signalingrelated markers was detected. The correlation between L-33/ST2 signaling and different phenotype macrophage markers as well as ICC patients' clinicopathologic data was evaluated. RESULTS Massive heterogeneous cancer cells and PAS-positive cells were observed in tumor tissues. CD68-positive cells accumulated in tumor tissues and expression of both M1 phenotype markers and M2 phenotype macrophage markers was higher in tumor samples than para-carcinoma samples. However, M2 phenotype macrophages represented the dominant macrophage population in ICC tissues. Plasma levels of IL-33, ST2, and MIF were evidently enhanced in ICC patients compared to healthy controls. IL-33/ST2 signaling-related markers exhibited a massive increase in tumor samples than para-carcinoma samples. IL-33 and ST2 expression in ICC tissues was positively associated with M1 and M2 phenotype macrophages. Plasma levels of IL-33, ST2, and MIF were correlated with the diameter of tumor lesions, lymph node metastasis, TNM stage, and tumor differentiation degree. Multivariate analysis demonstrated IL-33 expression to exhibit a correlation with the diameter of tumor lesions, lymph node metastasis, and TNM stage. Additionally, there was a relationship observed between ST2, MIF expression, and diameter of tumor lesions plus TNM stage. CONCLUSION IL-33/ST2 signaling exhibited a positive relationship with macrophage heterogeneity in ICC tissues, and upregulated levels of IL-33, ST2, and MIF were associated with aggressive clinicopathologic characteristics. These findings may provide promising diagnostic biomarkers and potential therapeutic strategies for ICC patients targeting IL-33/ST2 signaling.
Collapse
Affiliation(s)
- Aimaiti Yasen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao High Street, Shapingba District, Chongqing, 400037, China
| | - ZhanDong Yang
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, Province, China
| | - Jun Feng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, Guangdong Province, China
| | - RunBin Liang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| | - TianXing Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| | - Kai Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| | - YuHong Cai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| | - GuoYing Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| |
Collapse
|
40
|
Yang Q, Zheng Y, Dong J, Xu Q, Li S, Li A, Xiong X, Zhang Y. Serum interleukin-33 combined with FEF75% z-score and FeNO improves the diagnostic accuracy of asthma in children. J Pediatr (Rio J) 2024; 100:81-87. [PMID: 37741632 PMCID: PMC10751693 DOI: 10.1016/j.jped.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 09/25/2023] Open
Abstract
OBJECTIVE To investigate the diagnostic efficacy of serum IL-33 single indicator and combined indicators for asthma in children. METHODS 132 children were initially diagnosed with asthma during acute exacerbation and 100 healthy children were included. Serum IL-33 concentration differences were compared between asthmatic and normal children. Correlations between IL-33 with pulmonary function parameters, FeNO, peripheral blood EOS counts and serum total IgE were analyzed in asthmatic children. ROC curves were used to assess IL-33 diagnostic efficacy and its combined indicators. To prevent overfitting of the predictive model, the hold-out cross-validation method was used. RESULTS (1) Serum IL-33 concentrations were significantly higher in children with asthma than in normal children (p < 0.001). (2) IL-33 concentration was negatively correlated with FVC z-score, FEV1 z-score and FEF75% z-score in asthmatic children (p < 0.05). (3) The area under the ROC curve of IL-33 was 0.821, which was higher than those of FeNO, FVC z-score, and FEV1 z-score. (4) Cross-validation of the combined indicators showed that IL-33 significantly improved asthma diagnostic efficacy. The combination of IL-33, FEF75% z-score, and FeNO showed the highest diagnostic efficacy, with the AUC, sensitivity, and specificity of the combined indicator being 0.954, 90.1%, and 89. 0%, respectively, and good extrapolation of the predictive model. CONCLUSION Serum IL-33 is higher in children with asthma and increases with the severity of pulmonary ventilation obstruction. A single indicator of serum IL-33 demonstrates moderate diagnostic accuracy, and its combination with FEF75% z-score and FeNO significantly improves the diagnostic accuracy in childhood asthma.
Collapse
Affiliation(s)
- Qiuyan Yang
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China; Henan Pediatric Clinical Research Center, Zhengzhou, China; Henan Key Laboratory of Child Brain Injury, Zhengzhou, China; Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Yuehong Zheng
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Junjun Dong
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Qingrong Xu
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Shufang Li
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Aijun Li
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Xiaoman Xiong
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Yanli Zhang
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China; Henan Pediatric Clinical Research Center, Zhengzhou, China; Henan Key Laboratory of Child Brain Injury, Zhengzhou, China; Institute of Neuroscience of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
41
|
Wang Y, Ge WL, Wang SJ, Liu YY, Zhang ZH, Hua Y, Zhang XF, Zhang JJ. MiR-548t-5p regulates pancreatic ductal adenocarcinoma metastasis through an IL-33-dependent crosstalk between cancer cells and M2 macrophages. Cell Cycle 2024; 23:169-187. [PMID: 38267823 PMCID: PMC11037285 DOI: 10.1080/15384101.2024.2309026] [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: 10/24/2023] [Revised: 12/07/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
IL-33 has been associated with pro- and anticancer functions in cancer. However, its role in pancreatic cancer metastasis remains unknown. This study aimed to explore the role of miR-548t-5p/IL-33 axis in the metastasis of pancreatic cancer. Luciferase activity assay, qRT-PCR, Western blot and ELISA were performed to prove whether IL-33 is the target of miR-548t-5p. In vivo metastasis assay and cellular transwell assay were performed to explore the role of miR-548t-5p/IL-33 axis in the invasion and metastasis of pancreatic cancer. Co-culture experiments and immunohistochemistry were performed to observe whether IL-33 affects cell invasion and metastasis dependent on the involvement of M2 macrophages. THP-1 cell induction experiment and flow cytometry were performed to explore the effect of IL-33 on macrophage polarization. CCK-8, colony formation, cell apoptosis, cell cycle, cell wound healing and transwell assay were performed to investigate the effect of IL-33 induced M2 macrophages on cell malignant biological behavior by coculturing pancreatic cancer cells with the conditioned medium (CM) from macrophages. We found that miR-548t-5p regulated the expression and secretion of IL-33 in pancreatic cancer cells by directly targeting IL-33 mRNA. IL-33 secreted by cancer cells promoted the recruitment and activation of macrophages to a M2-like phenotype. In turn, IL-33 induced M2 macrophages promoted the migration and invasion of cancer cells. Moreover, IL-33 affected pancreatic cancer cell invasion dependent on the involvement of M2 macrophages in the co-culture system. Thus, our study suggested that manipulation of this IL-33-dependent crosstalk has a therapeutic potential for the treatment of pancreatic cancer metastasis.
Collapse
Affiliation(s)
- Yan Wang
- Endoscopy Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Yining, China
| | - Wan-Li Ge
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shao-Jun Wang
- Department of Biochemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu-Yong Liu
- Department of Biochemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhi-Han Zhang
- Department of Biochemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Hua
- Department of Biochemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiong-Fei Zhang
- Department of Biochemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Jing Zhang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute, Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Bolger GB. Therapeutic Targets and Precision Medicine in COPD: Inflammation, Ion Channels, Both, or Neither? Int J Mol Sci 2023; 24:17363. [PMID: 38139192 PMCID: PMC10744217 DOI: 10.3390/ijms242417363] [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: 11/13/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The development of a wider range of therapeutic options is a key objective in drug discovery for chronic obstructive pulmonary disease (COPD). Fundamental advances in lung biology have the potential to greatly expand the number of therapeutic targets in COPD. The recently reported successful Phase 3 clinical trial of the first biologic agent for COPD, the monoclonal antibody dupilumab, adds additional support to the importance of targeting inflammatory pathways in COPD. However, numerous other cellular mechanisms are important targets in COPD therapeutics, including airway remodeling, the CFTR ion channel, and mucociliary function. Some of these emerging targets can be exploited by the expanded use of existing COPD drugs, such as roflumilast, while targeting others will require the development of novel molecular entities. The identification of additional therapeutic targets and agents has the potential to greatly expand the value of using clinical and biomarker data to classify COPD into specific subsets, each of which can be predictive of an enhanced response to specific subset(s) of targeted therapies. The author reviews established and emerging drug targets in COPD and uses this as a framework to define a novel classification of COPD based on therapeutic targets. This novel classification has the potential to enhance precision medicine in COPD patient care and to accelerate clinical trials and pre-clinical drug discovery efforts.
Collapse
Affiliation(s)
- Graeme B Bolger
- BZI Pharma LLC, 1500 1st Ave N., Unit 36, Birmingham, AL 35203-1872, USA
| |
Collapse
|
43
|
Meng Q, Wang Y, Yuan T, Su Y, Li Z, Sun S. Osteoclast: The novel whistleblower in osteonecrosis of the femoral head. GENE REPORTS 2023; 33:101833. [DOI: 10.1016/j.genrep.2023.101833] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
44
|
Li W, Liu M, Chu M. Strategies targeting IL-33/ST2 axis in the treatment of allergic diseases. Biochem Pharmacol 2023; 218:115911. [PMID: 37981174 DOI: 10.1016/j.bcp.2023.115911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023]
Abstract
Interleukin-33 (IL-33) and its receptor Serum Stimulation-2 (ST2, also called Il1rl1) are members of the IL-1 superfamily that plays a crucial role in allergic diseases. The interaction of IL-33 and ST2 mainly activates NF-κB signaling and MAPK signaling via the MyD88/IRAK/TRAF6 module, resulting in the production and secretion of pro-inflammatory cytokines. The IL-33/ST2 axis participates in the pathogenesis of allergic diseases, and therefore serves as a promising strategy for allergy treatment. In recent years, strategies blocking IL-33/ST2 through targeting regulation of IL-33 and ST2 or targeting the molecules involved in the signal transduction have been extensively studied mostly in animal models. These studies provide various potential therapeutic agents other than antibodies, such as small molecules, nucleic acids and traditional Chinese medicines. Herein, we reviewed potential targets and agents targeting IL-33/ST2 axis in the treatment of allergic diseases, providing directions for further investigations on treatments for IL-33 induced allergic diseases.
Collapse
Affiliation(s)
- Wenran Li
- Department of Immunology, School of Basic Medical Sciences, Health Science Centre, Peking University. Beijing, China
| | - Mengqi Liu
- Department of Immunology, School of Basic Medical Sciences, Health Science Centre, Peking University. Beijing, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Sciences, Health Science Centre, Peking University. Beijing, China; Beijing Life Science Academy, Beijing, China.
| |
Collapse
|
45
|
Kattner AA. Colonizing foreign terrain: Insights into bacterial enteropathogens. Biomed J 2023; 46:100681. [PMID: 38042347 PMCID: PMC10774447 DOI: 10.1016/j.bj.2023.100681] [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: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/04/2023] Open
Abstract
In this present issue of the Biomedical Journal insights into pediatric campylobacteriosis are granted, and a potential path to developing a parenteral vaccine against enterotoxigenic E. coli is demonstrated. Additionally, a study shows how the use of extracorporeal shockwave therapy contributes to countering osteonecrosis of the femoral head. Furthermore, the relation between intimate partner violence and a saliva biomarker is explored. Finally, findings concerning the risk of dementia in patients with autonomic nervous system dysregulation are elucidated; and patterns of non-Alzheimer disease pathophysiology in individuals with depressive disorder are revealed.
Collapse
|
46
|
Franke K, Li Z, Bal G, Zuberbier T, Babina M. Synergism between IL-33 and MRGPRX2/FcεRI Is Primarily Due to the Complementation of Signaling Modules, and Only Modestly Supplemented by Prolonged Activation of Selected Kinases. Cells 2023; 12:2700. [PMID: 38067128 PMCID: PMC10705352 DOI: 10.3390/cells12232700] [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: 09/13/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
Skin mast cells (MCs) express high levels of MRGPRX2, FcεRI, and ST2, and vigorously respond to their ligands when triggered individually. IL-33/ST2 also potently synergizes with other receptors, but the molecular underpinnings are poorly understood. Human skin-derived MCs were stimulated via different receptors individually or jointly in the presence/absence of selective inhibitors. TNF was quantified by ELISA. Signaling cascades were studied by immunoblot. TNF was stimulated by FcεRI ≈ ST2 > MRGPRX2. Surprisingly, neither FcεRI nor MRGPRX2 stimulation elicited NF-κB activation (IκB degradation, p65 phosphorylation) in stark contrast to IL-33. Accordingly, TNF production did not depend on NF-κB in FcεRI- or MRGPRX2-stimulated MCs, but did well so downstream of ST2. Conversely, ERK1/2 and PI3K were the crucial modules upon FcεRI/MRGPRX2 stimulation, while p38 was key to the IL-33-elicited route. The different signaling prerequisites were mirrored by their activation patterns with potent pERK/pAKT after FcεRI/MRGPRX2, but preferential induction of pp38/NF-κB downstream of ST2. FcεRI/MRGPRX2 strongly synergized with IL-33, and some synergy was still observed upon inhibition of each module (ERK1/2, JNK, p38, PI3K, NF-κB). IL-33's contribution to synergism was owed to p38 > JNK > NF-κB, while the partner receptor contributed through ERK > PI3K ≈ JNK. Concurrent IL-33 led to slightly prolonged pERK (downstream of MRGPRX2) or pAKT (activated by FcεRI), while the IL-33-elicited modules (pp38/NF-κB) remained unaffected by co-stimulation of FcεRI/MRGPRX2. Collectively, the strong synergistic activity of IL-33 primarily results from the complementation of highly distinct modules following co-activation of the partner receptor rather than by altered signal strength of the same modules.
Collapse
Affiliation(s)
- Kristin Franke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Zhuoran Li
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Gürkan Bal
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Torsten Zuberbier
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
47
|
Salamon D, Ujvari D, Hellberg A, Hirschberg AL. DHT and Insulin Upregulate Secretion of the Soluble Decoy Receptor of IL-33 From Decidualized Endometrial Stromal Cells. Endocrinology 2023; 165:bqad174. [PMID: 37972259 PMCID: PMC10681354 DOI: 10.1210/endocr/bqad174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/17/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Interleukin 33 (IL-33) signaling regulates most of the key processes of pregnancy, including decidualization, trophoblast proliferation and invasion, vascular remodeling, and placental growth. Accordingly, dysregulation of IL-33, its membrane-bound receptor (ST2L, transducer of IL-33 signaling), and its soluble decoy receptor (sST2, inhibitor of IL-33 signaling) has been linked to a wide range of adverse pregnancy outcomes that are common in women with obesity and polycystic ovary syndrome, that is, conditions associated with hyperandrogenism, insulin resistance, and compensatory hyperinsulinemia. To reveal if androgens and insulin might modulate uteroplacental IL-33 signaling, we investigated the effect of dihydrotestosterone (DHT) and/or insulin on the expression of ST2L and sST2 (along with the activity of their promoter regions), IL-33 and sIL1RAP (heterodimerization partner of sST2), during in vitro decidualization of endometrial stromal cells from 9 healthy women. DHT and insulin markedly upregulated sST2 secretion, in addition to the upregulation of its messenger RNA (mRNA) expression, while the proximal ST2 promoter, from which the sST2 transcript originates, was upregulated by insulin, and in a synergistic manner by DHT and insulin combination treatment. On the other hand, sIL1RAP was slightly downregulated by insulin and IL-33 mRNA expression was not affected by any of the hormones, while ST2L mRNA expression and transcription from its promoter region (distal ST2 promoter) could not be detected or showed a negligibly low level. We hypothesize that high levels of androgens and insulin might lead to subfertility and pregnancy complications, at least partially, through the sST2-dependent downregulation of uteroplacental IL-33 signaling.
Collapse
Affiliation(s)
- Daniel Salamon
- Department of Women's and Children's Health, Karolinska Institute, SE-171 64 Stockholm, Sweden
| | - Dorina Ujvari
- Department of Women's and Children's Health, Karolinska Institute, SE-171 64 Stockholm, Sweden
- Department of Microbiology, Tumor and Cell Biology, National Pandemic Centre, Centre for Translational Microbiome Research, Karolinska Institute, SE-171 64 Stockholm, Sweden
| | - Anton Hellberg
- Department of Women's and Children's Health, Karolinska Institute, SE-171 64 Stockholm, Sweden
| | - Angelica Lindén Hirschberg
- Department of Women's and Children's Health, Karolinska Institute, SE-171 64 Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| |
Collapse
|
48
|
Yazici D, Ogulur I, Pat Y, Babayev H, Barletta E, Ardicli S, Bel Imam M, Huang M, Koch J, Li M, Maurer D, Radzikowska U, Satitsuksanoa P, Schneider SR, Sun N, Traidl S, Wallimann A, Wawrocki S, Zhakparov D, Fehr D, Ziadlou R, Mitamura Y, Brüggen MC, van de Veen W, Sokolowska M, Baerenfaller K, Nadeau K, Akdis M, Akdis CA. The epithelial barrier: The gateway to allergic, autoimmune, and metabolic diseases and chronic neuropsychiatric conditions. Semin Immunol 2023; 70:101846. [PMID: 37801907 DOI: 10.1016/j.smim.2023.101846] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/08/2023]
Abstract
Since the 1960 s, our health has been compromised by exposure to over 350,000 newly introduced toxic substances, contributing to the current pandemic in allergic, autoimmune and metabolic diseases. The "Epithelial Barrier Theory" postulates that these diseases are exacerbated by persistent periepithelial inflammation (epithelitis) triggered by exposure to a wide range of epithelial barrier-damaging substances as well as genetic susceptibility. The epithelial barrier serves as the body's primary physical, chemical, and immunological barrier against external stimuli. A leaky epithelial barrier facilitates the translocation of the microbiome from the surface of the afflicted tissues to interepithelial and even deeper subepithelial locations. In turn, opportunistic bacterial colonization, microbiota dysbiosis, local inflammation and impaired tissue regeneration and remodelling follow. Migration of inflammatory cells to susceptible tissues contributes to damage and inflammation, initiating and aggravating many chronic inflammatory diseases. The objective of this review is to highlight and evaluate recent studies on epithelial physiology and its role in the pathogenesis of chronic diseases in light of the epithelial barrier theory.
Collapse
Affiliation(s)
- Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Elena Barletta
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Swiss Institute of Bioinformatics (SIB), Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mengting Huang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Jana Koch
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Swiss Institute of Bioinformatics (SIB), Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Debbie Maurer
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | | | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Na Sun
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China
| | - Stephan Traidl
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Alexandra Wallimann
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sebastian Wawrocki
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Damir Zhakparov
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Danielle Fehr
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Reihane Ziadlou
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marie-Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Katja Baerenfaller
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Swiss Institute of Bioinformatics (SIB), Davos, Switzerland
| | - Kari Nadeau
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland.
| |
Collapse
|
49
|
Lameire S, Hammad H. Lung epithelial cells: Upstream targets in type 2-high asthma. Eur J Immunol 2023; 53:e2250106. [PMID: 36781404 DOI: 10.1002/eji.202250106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
Over the last years, technological advances in the field of asthma have led to the identification of two disease endotypes, namely, type 2-high and type 2-low asthma, characterized by different pathophysiologic mechanisms at a cellular and molecular level. Although specific immune cells are important contributors to each of the recognized asthma endotype, the lung epithelium is now regarded as a crucial player able to orchestrate responses to inhaled environmental triggers such as allergens and microbes. The impact of the epithelium goes beyond its physical barrier. It is nowadays considered as a part of the innate immune system that can actively respond to insults. Activated epithelial cells, by producing a specific set of cytokines, trigger innate and adaptive immune cells to cause pathology. Here, we review how the epithelium contributes to the development of Th2 sensitization to allergens and asthma with a "type 2-high" signature, in both murine models and human studies of this asthma endotype. We also discuss epithelial responses to respiratory viruses, such as rhinovirus, respiratory syncytial virus, and SARS-CoV-2, and how these triggers influence not only asthma development but also asthma exacerbation. Finally, we also summarize the results of promising clinical trials using biologicals targeting epithelial-derived cytokines in asthmatic patients.
Collapse
Affiliation(s)
- Sahine Lameire
- Laboratory of Mucosal Immunology and Immunoregulation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- Laboratory of Mucosal Immunology and Immunoregulation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
50
|
Kanazawa N, Iyoda M, Suzuki T, Tachibana S, Nagashima R, Honda H. Exploring the significance of interleukin-33/ST2 axis in minimal change disease. Sci Rep 2023; 13:18776. [PMID: 37907612 PMCID: PMC10618262 DOI: 10.1038/s41598-023-45678-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023] Open
Abstract
Minimal change disease (MCD), a common cause of idiopathic nephrotic syndrome, has been postulated to exhibit an association with allergic conditions. Recent studies revealed the crucial role of interleukin (IL)-33 in type 2 innate immunity. We hypothesized that development of MCD involves an IL-33-related immune response. We examined 49 patients with biopsy-proven MCD, 6 healthy volunteers, and 29 patients in remission. In addition to clinical features, serum and urinary levels of IL-33 and soluble suppression of tumorigenicity 2 protein (sST2), a secreted form of the receptor of IL-33, were analyzed. Although IL-33 was barely detectable in either MCD or control samples, sST2 levels at diagnosis were elevated in MCD patients. Serum sST2 levels of MCD patients were correlated with serum total protein level (r = - 0.36, p = 0.010) and serum creatinine level (r = 0.34, p = 0.016). Furthermore, the elevated sST2 levels were observed to decrease following remission. Immunofluorescence revealed IL-33 expression in the podocytes among MCD patients, with a significant increase compared with controls. In vitro, mouse podocyte cells incubated with serum from a MCD patient at disease onset showed increased IL-33 secretion. These results suggest an IL-33-related immune response plays a role in MCD.
Collapse
Affiliation(s)
- Nobuhiro Kanazawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masayuki Iyoda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
- Department of Microbiology and Immunology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Taihei Suzuki
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shohei Tachibana
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|