1
|
Inclan-Rico JM, Napuri CM, Lin C, Hung LY, Ferguson AA, Liu X, Wu Q, Pastore CF, Stephenson A, Femoe UM, Musaigwa F, Rossi HL, Freedman BD, Reed DR, Macháček T, Horák P, Abdus-Saboor I, Luo W, Herbert DR. MrgprA3 neurons drive cutaneous immunity against helminths through selective control of myeloid-derived IL-33. Nat Immunol 2024; 25:2068-2084. [PMID: 39354200 DOI: 10.1038/s41590-024-01982-y] [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: 11/21/2023] [Accepted: 09/11/2024] [Indexed: 10/03/2024]
Abstract
Skin uses interdependent cellular networks for barrier integrity and host immunity, but most underlying mechanisms remain obscure. Herein, we demonstrate that the human parasitic helminth Schistosoma mansoni inhibited pruritus evoked by itch-sensing afferents bearing the Mas-related G-protein-coupled receptor A3 (MrgprA3) in mice. MrgprA3 neurons controlled interleukin (IL)-17+ γδ T cell expansion, epidermal hyperplasia and host resistance against S. mansoni through shaping cytokine expression in cutaneous antigen-presenting cells. MrgprA3 neuron activation downregulated IL-33 but induced IL-1β and tumor necrosis factor in macrophages and type 2 conventional dendritic cells partially through the neuropeptide calcitonin gene-related peptide. Macrophages exposed to MrgprA3-derived secretions or bearing cell-intrinsic IL-33 deletion showed increased chromatin accessibility at multiple inflammatory cytokine loci, promoting IL-17/IL-23-dependent changes to the epidermis and anti-helminth resistance. This study reveals a previously unrecognized intercellular communication mechanism wherein itch-inducing MrgprA3 neurons initiate host immunity against skin-invasive parasites by directing cytokine expression patterns in myeloid antigen-presenting cell subsets.
Collapse
Affiliation(s)
- Juan M Inclan-Rico
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Camila M Napuri
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cailu Lin
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Li-Yin Hung
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Annabel A Ferguson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaohong Liu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qinxue Wu
- Department of Neuroscience, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher F Pastore
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adriana Stephenson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ulrich M Femoe
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fungai Musaigwa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather L Rossi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Tomáš Macháček
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Horák
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Ishmail Abdus-Saboor
- Department of Biological Sciences, Zuckerman Mind, Brain, Behavior Institute, Columbia University, New York, NY, USA
| | - Wenqin Luo
- Department of Neuroscience, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Diaz-Barreiro A, Cereghetti G, Ortega Sánchez FG, Tonacini J, Talabot-Ayer D, Kieffer-Jaquinod S, Kissling VM, Huard A, Swale C, Knowles TPJ, Couté Y, Peter M, Francés-Monerris A, Palmer G. Oxidation-sensitive cysteines drive IL-38 amyloid formation. Cell Rep 2024; 43:114940. [PMID: 39488827 DOI: 10.1016/j.celrep.2024.114940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 07/16/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024] Open
Abstract
Interleukin (IL)-1 family cytokines are essential for host defense at epithelial barriers. The IL-1 family member IL-33 was recently linked to stress granules (SGs). Formation of SGs and other biomolecular condensates is promoted by proteins containing low-complexity regions (LCRs). Computational analysis predicts LCRs in six of the 11 IL-1 family members. Among these, IL-38 contains a long LCR including two amyloid cores. IL-38 localizes to intracellular granules in keratinocytes under oxidative stress (OS) and forms OS-induced amyloid aggregates in cells and in vitro. Interestingly, soluble and aggregated IL-38 are released from keratinocytes in an exosome-enriched extracellular vesicle fraction. Disulfide-bond mapping, in silico modeling, and mutational analysis suggest that oxidation-sensitive cysteines act as redox switches to alter IL-38 conformation and promote its aggregation. Finally, the presence of IL-38 granules in human epidermis facing environmental OS suggests that oxidation-induced amyloidogenesis, as an intrinsic property of IL-38, supports barrier function.
Collapse
Affiliation(s)
- Alejandro Diaz-Barreiro
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland.
| | - Gea Cereghetti
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Francisco Gabriel Ortega Sánchez
- IBS Granada, Institute of Biomedical Research, Granada, Spain; Pulmonology Unit, Hospital Universitario Virgen de las Nieves, Granada, Spain; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - Jenna Tonacini
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Dominique Talabot-Ayer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| | | | - Vera Maria Kissling
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Empa (Swiss Federal Laboratories for Materials Science and Technology), St. Gallen, Switzerland
| | - Arnaud Huard
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Christopher Swale
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Grenoble, France
| | | | - Yohann Couté
- Université Grenoble Alpes, CEA, INSERM, UA13 BGE, CNRS, CEA, FR2048 Grenoble, France
| | - Matthias Peter
- Institute of Biochemistry, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| |
Collapse
|
3
|
Feng Z, Kuang Y, Qi Y, Wang X, Xu P, Chen X. Exogenous IL-33 promotes tumor immunity via macroscopic regulation of ILC2s. Sci Rep 2024; 14:26140. [PMID: 39478174 PMCID: PMC11525627 DOI: 10.1038/s41598-024-77751-6] [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: 07/09/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Interleukin-33 (IL-33) is a pleiotropic molecule that plays various roles in the body. However, how exogenous IL-33 changes the tumor immune microenvironment remains unclear. Our study revealed that exogenous IL-33 exerts anti-tumor effects and effectively suppresses the progression of subcutaneous melanoma. scRNA-seq analysis revealed that exogenous IL-33 reduced neutrophils accumulation, thereby improving the inhibitory immune environment. Flow cytometry analysis revealed that exogenous IL-33 significantly increased the proportion of eosinophils and group 2 innate lymphoid cells (ILC2s). In addition, we identified genes encoding major histocompatibility complex (MHC) class II molecules in this group of ILC2s, suggesting that ILC2s may play a role in antigen presentation. In Il7rCreArg1flox/flox mice, the decrease of ILC2s led to a reduction in the proportion of eosinophils. Furthermore, we found that exogenous IL-33 effectively promoted the differentiation of ILC2s and their accumulation in tumors, thereby enhancing the anti-tumor immune response. These findings may pave the way for developing new cancer immunotherapies that use IL-33 as an activator to enhance anti-tumor immune responses.
Collapse
Affiliation(s)
- Zhenchu Feng
- Department of Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, China
| | - Ye Kuang
- Department of Gynecology and Obstetrics, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yuan Qi
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Wang
- Department of Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, China
| | - Peng Xu
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, 157 Baojian Road, Harbin, 150086, China.
| | - Xi Chen
- Department of Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, China.
| |
Collapse
|
4
|
Qian F, He R, Du X, Wei Y, Zhou Z, Fan J, He Y. Microglia and Astrocytes Responses Contribute to Alleviating Inflammatory Damage by Repetitive Transcranial Magnetic Stimulation in Rats with Traumatic Brain Injury. Neurochem Res 2024; 49:2636-2651. [PMID: 38909329 DOI: 10.1007/s11064-024-04197-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/30/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a therapeutic strategy that shows promise in ameliorating the clinical sequelae following traumatic brain injury (TBI). These improvements are associated with neuroplastic changes in neurons and their synaptic connections. However, it has been hypothesized that rTMS may also modulate microglia and astrocytes, potentially potentiating their neuroprotective capabilities. This study aims to investigate the effects of high-frequency rTMS on microglia and astrocytes that may contribute to its neuroprotective effects. Feeney's weight-dropping method was used to establish rat models of moderate TBI. To evaluate the neuroprotective effect of high frequency rTMS on rats by observing the synaptic ultrastructure and the level of neuron apoptosis. The levels of several important inflammation-related proteins within microglia and astrocytes were assessed through immunofluorescence staining and western blot. Our findings demonstrate that injured neurons can be rescued through the modulation of microglia and astrocytes by rTMS. This modulation plays a key role in preserving the synaptic ultrastructure and inhibiting neuronal apoptosis. Among microglia, we observed that rTMS inhibited the levels of proinflammatory factors (CD16, IL-6 and TNF-α) and promoted the levels of anti-inflammatory factors (CD206, IL-10 and TNF-β). rTMS also reduced the levels of pyroptosis within microglia and pyroptosis-related proteins (NLRP3, Caspase-1, GSDMD, IL-1β and IL-18). Moreover, rTMS downregulated P75NTR expression and up-regulated IL33 expression in astrocytes. These findings suggest that regulation of microglia and astrocytes is the mechanism through which rTMS attenuates neuronal inflammatory damage after moderate TBI.
Collapse
Affiliation(s)
- FangFang Qian
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - RenHong He
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - XiaoHui Du
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Yi Wei
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Zhou Zhou
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - JianZhong Fan
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| | - YouHua He
- Department of Comprehensive Medical Treatment Ward, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| |
Collapse
|
5
|
Erickson HL, Taniguchi S, Raman A, Leitenberger JJ, Malhotra SV, Oshimori N. Cancer stem cells release interleukin-33 within large oncosomes to promote immunosuppressive differentiation of macrophage precursors. Immunity 2024; 57:1908-1922.e6. [PMID: 39079535 PMCID: PMC11324407 DOI: 10.1016/j.immuni.2024.07.004] [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: 09/11/2023] [Revised: 03/12/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024]
Abstract
In squamous cell carcinoma (SCC), macrophages responding to interleukin (IL)-33 create a TGF-β-rich stromal niche that maintains cancer stem cells (CSCs), which evade chemotherapy-induced apoptosis in part via activation of the NRF2 antioxidant program. Here, we examined how IL-33 derived from CSCs facilitates the development of an immunosuppressive microenvironment. CSCs with high NRF2 activity redistributed nuclear IL-33 to the cytoplasm and released IL-33 as cargo of large oncosomes (LOs). Mechanistically, NRF2 increased the expression of the lipid scramblase ATG9B, which exposed an "eat me" signal on the LO surface, leading to annexin A1 (ANXA1) loading. These LOs promoted the differentiation of AXNA1 receptor+ myeloid precursors into immunosuppressive macrophages. Blocking ATG9B's scramblase activity or depleting ANXA1 decreased niche macrophages and hindered tumor progression. Thus, IL-33 is released from live CSCs via LOs to promote the differentiation of alternatively activated macrophage, with potential relevance to other settings of inflammation and tissue repair.
Collapse
Affiliation(s)
- Hannah L Erickson
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Sachiko Taniguchi
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Anish Raman
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Justin J Leitenberger
- Department of Dermatology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Sanjay V Malhotra
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA; Center for Experimental Therapeutics, Oregon Health and Science University, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Naoki Oshimori
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA; Department of Dermatology, Oregon Health and Science University, Portland, OR 97239, USA; Department of Otolaryngology, Head & Neck Surgery, Oregon Health and Science University, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
6
|
Bisen S, Verma SK, Mukhopadhyay CS, Singh NK. A neutrophil elastase-generated mature form of IL-33 is a potent regulator of endothelial cell activation and proliferative retinopathy. Exp Mol Med 2024; 56:1703-1716. [PMID: 39085349 PMCID: PMC11372157 DOI: 10.1038/s12276-024-01279-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: 02/03/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 08/02/2024] Open
Abstract
Human interleukin-33 (IL-33) is a 270 amino acid protein that belongs to the IL-1 cytokine family and plays an important role in various inflammatory disorders. Neutrophil proteases (Cathepsin G and Elastase) and mast cell proteases (tryptase and chymase) regulate the activity of IL-33 by processing full-length IL-33 into its mature form. There is little evidence on the role of these mature forms of IL-33 in retinal endothelial cell signaling and pathological retinal angiogenesis. Here, we cloned, expressed, and purified the various mature forms of human IL-33 and then evaluated the effects of IL-3395-270, IL-3399-270, IL-33109-270, and IL-33112-270 on angiogenesis in human retinal microvascular endothelial cells (HRMVECs). We observed that IL-3395-270, IL-3399-270, IL-33109-270, and IL-33112-270 significantly induced HRMVEC migration, tube formation and sprouting angiogenesis. However, only IL-3399-270 could induce HRMVEC proliferation. We used a murine model of oxygen-induced retinopathy (OIR) to assess the role of these mature forms of IL-33 in pathological retinal neovascularization. Our 3'-mRNA sequencing and signaling studies indicated that IL-3399-270 and IL-33109-270 were more potent at inducing endothelial cell activation and angiogenesis than the other mature forms. We found that genetic deletion of IL-33 significantly reduced OIR-induced retinal neovascularization in the mouse retina and that intraperitoneal administration of mature forms of IL-33, mainly IL-3399-270 and IL-33109-270, significantly restored ischemia-induced angiogenic sprouting and tuft formation in the hypoxic retinas of IL-33-/- mice. Thus, our study results suggest that blockade or inhibition of IL-33 cleavage by neutrophil proteases could help mitigate pathological angiogenesis in proliferative retinopathies.
Collapse
Affiliation(s)
- Shivantika Bisen
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, 48202, USA
| | - Shailendra Kumar Verma
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, 48202, USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, San Diego, CA, 92037, USA
| | - Chandra Sekhar Mukhopadhyay
- Department of Bioinformatics, School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, 141004, India
| | - Nikhlesh K Singh
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA.
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
7
|
Hai S, Li X, Xie E, Wu W, Gao Q, Yu B, Hu J, Xu F, Zheng X, Zhang BH, Wu D, Yan W, Ning Q, Wang X. Intestinal IL-33 promotes microbiota-derived trimethylamine N -oxide synthesis and drives metabolic dysfunction-associated steatotic liver disease progression by exerting dual regulation on HIF-1α. Hepatology 2024:01515467-990000000-00950. [PMID: 38985971 DOI: 10.1097/hep.0000000000000985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND AND AIMS Gut microbiota plays a prominent role in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD). IL-33 is highly expressed at mucosal barrier sites and regulates intestinal homeostasis. Herein, we aimed to investigate the role and mechanism of intestinal IL-33 in MASLD. APPROACH AND RESULTS In both humans and mice with MASLD, hepatic expression of IL-33 and its receptor suppression of tumorigenicity 2 (ST2) showed no significant change compared to controls, while serum soluble ST2 levels in humans, as well as intestinal IL-33 and ST2 expression in mice were significantly increased in MASLD. Deletion of global or intestinal IL-33 in mice alleviated metabolic disorders, inflammation, and fibrosis associated with MASLD by reducing intestinal barrier permeability and rectifying gut microbiota dysbiosis. Transplantation of gut microbiota from IL-33 deficiency mice prevented MASLD progression in wild-type mice. Moreover, IL-33 deficiency resulted in a decrease in the abundance of trimethylamine N -oxide-producing bacteria. Inhibition of trimethylamine N -oxide synthesis by 3,3-dimethyl-1-butanol mitigated hepatic oxidative stress in mice with MASLD. Nuclear IL-33 bound to hypoxia-inducible factor-1α and suppressed its activation, directly damaging the integrity of the intestinal barrier. Extracellular IL-33 destroyed the balance of intestinal Th1/Th17 and facilitated Th1 differentiation through the ST2- Hif1a - Tbx21 axis. Knockout of ST2 resulted in a diminished MASLD phenotype resembling that observed in IL-33 deficiency mice. CONCLUSIONS Intestinal IL-33 enhanced gut microbiota-derived trimethylamine N -oxide synthesis and aggravated MASLD progression through dual regulation on hypoxia-inducible factor-1α. Targeting IL-33 and its associated microbiota may provide a potential therapeutic strategy for managing MASLD.
Collapse
Affiliation(s)
- Suping Hai
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xitang Li
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Erliang Xie
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhui Wu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Gao
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Binghui Yu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Junjian Hu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Feiyang Xu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xizhe Zheng
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Bin-Hao Zhang
- Department of Surgery, Hepatic Surgery Center, Institute of Hepato-Pancreato-Biliary Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Weiming Yan
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Jiao M, Wang C, Tang X, Dai C, Zhang N, Fan A, Qian Z, Liu S, Zhang F, Li B, Xu Y, Tan Z, Gong F, Lu Y, Zheng F. Active secretion of IL-33 from astrocytes is dependent on TMED10 and promotes central nervous system homeostasis. Brain Behav Immun 2024; 119:539-553. [PMID: 38663774 DOI: 10.1016/j.bbi.2024.04.028] [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: 10/10/2023] [Revised: 04/07/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
Interleukin-33 (IL-33), secreted by astrocytes, regulates the synapse development in the spinal cord and hippocampus and suppresses autoimmune disease in the central nervous system (CNS). However, the mechanism of unconventional protein secretion of this cytokine remains unclear. In this study, we found that IFN-γ promotes the active secretion of IL-33 from astrocytes, and the active secretion of IL-33 from cytoplasm to extracellular space was dependent on interaction with transmembrane emp24 domain 10 (TMED10) via the IL-1 like cytokine domain in astrocytes. Knockout of Il-33 or its receptor St2 induced hippocampal astrocyte activation and depressive-like disorder in naive mice, as well as increased spinal cord astrocyte activation and polarization to a neurotoxic reactive subtype and aggravated passive experimental autoimmune encephalomyelitis (EAE). Our results have identified that IL-33 is actively secreted by astrocytes through the unconventional protein secretion pathway facilitated by TMED10 channels. This process helps maintain CNS homeostasis by inhibiting astrocyte activation.
Collapse
Affiliation(s)
- Mengya Jiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chenchen Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xuhuan Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chan Dai
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Na Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anqi Fan
- College of Life Science, Yangtze University, Jingzhou 434025, China
| | - Zhigang Qian
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiwang Liu
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feng Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yong Xu
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng Tan
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Feili Gong
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yisheng Lu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China.
| |
Collapse
|
9
|
Alhallak K, Nagai J, Zaleski K, Marshall S, Salloum T, Derakhshan T, Hayashi H, Feng C, Kratchmarov R, Lai J, Kuchibhotla V, Nishida A, Balestrieri B, Laidlaw T, Dwyer DF, Boyce JA. Mast cells control lung type 2 inflammation via prostaglandin E 2-driven soluble ST2. Immunity 2024; 57:1274-1288.e6. [PMID: 38821053 PMCID: PMC11168874 DOI: 10.1016/j.immuni.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 01/26/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024]
Abstract
Severe asthma and sinus disease are consequences of type 2 inflammation (T2I), mediated by interleukin (IL)-33 signaling through its membrane-bound receptor, ST2. Soluble (s)ST2 reduces available IL-33 and limits T2I, but little is known about its regulation. We demonstrate that prostaglandin E2 (PGE2) drives production of sST2 to limit features of lung T2I. PGE2-deficient mice display diminished sST2. In humans with severe respiratory T2I, urinary PGE2 metabolites correlate with serum sST2. In mice, PGE2 enhanced sST2 secretion by mast cells (MCs). Mice lacking MCs, ST2 expression by MCs, or E prostanoid (EP)2 receptors by MCs showed reduced sST2 lung concentrations and strong T2I. Recombinant sST2 reduced T2I in mice lacking PGE2 or ST2 expression by MCs back to control levels. PGE2 deficiency also reversed the hyperinflammatory phenotype in mice lacking ST2 expression by MCs. PGE2 thus suppresses T2I through MC-derived sST2, explaining the severe T2I observed in low PGE2 states.
Collapse
Affiliation(s)
- Kinan Alhallak
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jun Nagai
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Kendall Zaleski
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sofia Marshall
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tamara Salloum
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tahereh Derakhshan
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Hiroaki Hayashi
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Chunli Feng
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Radomir Kratchmarov
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Juying Lai
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Virinchi Kuchibhotla
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Airi Nishida
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Barbara Balestrieri
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tanya Laidlaw
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel F Dwyer
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Joshua A Boyce
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
10
|
Reis RA, Stolf CS, de Carvalho Sampaio HA, da Costa Silva BY, Özlü T, Kenger EB, Miguel MMV, Santamaria MP, Monteiro MDF, Casati MZ, Casarin RCV. Impact of dietary inflammatory index on gingival health. J Periodontol 2024; 95:550-562. [PMID: 38152036 DOI: 10.1002/jper.23-0292] [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: 05/10/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Periodontal disease is a biofilm-dependent chronic inflammatory condition triggered by a host response. Several factors impact systemic inflammation and could lead to changes in disease pathogenesis. Recently, studies have assessed the influence of nutritional patterns on the development of periodontitis. In the present cross-sectional study, we evaluated the dietary inflammatory profile on periodontal conditions, focusing on clinical, subgingival microbial, and cytokine assessment of individuals with periodontal health or gingivitis. METHODS One hundred patients with periodontal health or gingivitis were included. Plaque index (PI), Bleeding on probing (BoP), the probing depth (PD), and the clinical attachment level (CAL) for each patient were assessed. Nutritional data and the Dietary Inflammatory Index (DII) were recorded by two 24-h food recalls on non-consecutive days. Biofilm and gingival crevicular fluid (GCF) to assess the microbiome profile and inflammatory biomarkers were collected. Multiple regressions focused on the DII, age, and sex as predictors of periodontal conditions were done. RESULTS Age and moderate DII scores increased the risk of gingivitis by 1.64 and 3.94 times, respectively. Males with an elevated DII score had 27.15 times higher odds of being diagnosed with gingivitis and BoP (β = 6.54; p = 0.03). Elderly patients with a moderate or high DII score were less prone to gingivitis and increased BoP (p < 0.04) compared with younger subjects. Considering the DII, there were no differences in microbial alpha and beta diversity; however, distinct species abundance and a higher concentration of monocyte-chemoattractant protein-1 and interleukin 33 were seen in patients with a higher DII. CONCLUSION A pro-inflammatory diet significantly contributes to periodontal inflammation, modulating inflammatory biomarkers and affecting the subgingival microbial community in healthy individuals.
Collapse
Affiliation(s)
- Roberta Andrade Reis
- Periodontics Division, Department of Prosthodontics and Periodontics, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - Camila Schmidt Stolf
- Periodontics Division, Department of Prosthodontics and Periodontics, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | | | - Bruna Yhang da Costa Silva
- Federal Institute of Education, Science and Technology of Ceará, Limoeiro do Norte, Fortaleza, CE, Brazil
| | - Tuğçe Özlü
- Nutrition and Dietetics, Faculty of Health Sciences, Bahcesehir University, Istanbul, Turkey
| | - Emre Batuhan Kenger
- Nutrition and Dietetics, Faculty of Health Sciences, Istanbul Bilgi University, Istanbul, Turkey
| | - Manuela Maria Vianal Miguel
- Division of Periodontics, Department of Diagnosis and Surgery, Institute of Science and Technology - São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Mauro Pedrine Santamaria
- Division of Periodontics, Department of Diagnosis and Surgery, Institute of Science and Technology - São Paulo State University (Unesp), São José dos Campos, SP, Brazil
- Division of Periodontology, Center of Oral Health Research, College of Dentistry - University of Kentucky (UK), Lexington, Kentucky, USA
| | - Mabelle de Freitas Monteiro
- Periodontics Division, Department of Prosthodontics and Periodontics, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - Marcio Zaffalon Casati
- Periodontics Division, Department of Prosthodontics and Periodontics, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - Renato Corrêa Viana Casarin
- Periodontics Division, Department of Prosthodontics and Periodontics, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| |
Collapse
|
11
|
Hofherr A, Liarte Marin E, Musial B, Seth A, Slidel T, Conway J, Baker D, Hansen PB, Challis B, Bartesaghi S, Bhat M, Pecoits-Filho R, Tu X, Selvarajah V, Woollard K, Heerspink HJ. Inhibition of Interleukin-33 to Reduce Glomerular Endothelial Inflammation in Diabetic Kidney Disease. Kidney Int Rep 2024; 9:1876-1891. [PMID: 38899206 PMCID: PMC11184260 DOI: 10.1016/j.ekir.2024.03.009] [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: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction Inflammation is a significant contributor to cardiorenal morbidity and mortality in diabetic kidney disease (DKD). The pathophysiological mechanisms linking systemic, subacute inflammation and local, kidney injury-initiated immune maladaptation is partially understood. Methods Here, we explored the expression of proinflammatory cytokines in patients with DKD; investigated mouse models of type 1 and type 2 diabetes (T2D); evaluated glomerular signaling in vitro; performed post hoc analyses of systemic and urinary markers of inflammation; and initiated a phase 2b clinical study (FRONTIER-1; NCT04170543). Results Transcriptomic profiling of kidney biopsies from patients with DKD revealed significant glomerular upregulation of interleukin-33 (IL-33). Inhibition of IL-33 signaling reduced glomerular damage and albuminuria in the uninephrectomized db/db mouse model (T2D/DKD). On a cellular level, inhibiting IL-33 improved glomerular endothelial health by decreasing cellular inflammation and reducing release of proinflammatory cytokines. Therefore, FRONTIER-1 was designed to test the safety and efficacy of the IL-33-targeted monoclonal antibody tozorakimab in patients with DKD. So far, 578 patients are enrolled in FRONTIER-1. The baseline inflammation status of participants (N > 146) was assessed in blood and urine. Comparison to independent reference cohorts (N > 200) validated the distribution of urinary tumor necrosis factor receptor 1 (TNFR1) and C-C motif chemokine ligand 2 (CCL2). Treatment with dapagliflozin for 6 weeks did not alter these biomarkers significantly. Conclusion We show that blocking the IL-33 pathway may mitigate glomerular endothelial inflammation in DKD. The findings from the FRONTIER-1 study will provide valuable insights into the therapeutic potential of IL-33 inhibition in DKD.
Collapse
Affiliation(s)
- Alexis Hofherr
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elena Liarte Marin
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Barbara Musial
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Asha Seth
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Tim Slidel
- Bioinformatics, Oncology R&D, AstraZeneca, Cambridge, UK
| | - James Conway
- Bioinformatics, Oncology R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - David Baker
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Pernille B.L. Hansen
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Benjamin Challis
- Translational Science and Experimental Medicine, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stefano Bartesaghi
- Translational Science and Experimental Medicine, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Bhat
- Translational Science and Experimental Medicine, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Roberto Pecoits-Filho
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
- School of Medicine, Pontificia Universidade de Catolica do Parana, Curitiba, Brazil
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Xiao Tu
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Viknesh Selvarajah
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kevin Woollard
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Hiddo J.L. Heerspink
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
12
|
Canè L, Poto R, Palestra F, Pirozzi M, Parashuraman S, Iacobucci I, Ferrara AL, La Rocca A, Mercadante E, Pucci P, Marone G, Monti M, Loffredo S, Varricchi G. TSLP is localized in and released from human lung macrophages activated by T2-high and T2-low stimuli: relevance in asthma and COPD. Eur J Intern Med 2024; 124:89-98. [PMID: 38402021 DOI: 10.1016/j.ejim.2024.02.020] [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: 01/08/2024] [Revised: 02/05/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Macrophages are the predominant immune cells in the human lung and play a central role in airway inflammation, including asthma and chronic obstructive pulmonary disease (COPD). Thymic stromal lymphopoietin (TSLP), a pleiotropic cytokine mainly expressed by bronchial epithelial cells, plays a key role in asthma and COPD pathobiology. TSLP exists in two variants: the long form (lfTSLP) and a shorter TSLP isoform (sfTSLP). We aimed to localize TSLP in human lung macrophages (HLMs) and investigate the mechanisms of its release from these cells. We also evaluated the effects of the two variants of TSLP on the release of angiogenic factor from HLMs. METHODS We employed immunofluorescence and Western blot to localize intracellular TSLP in HLMs purified from human lung parenchyma. HLMs were activated by T2-high (IL-4, IL-13) and T2-low (lipopolysaccharide: LPS) immunological stimuli. RESULTS TSLP was detected in HLMs and subcellularly localized in the cytoplasm. IL-4 and LPS induced TSLP release from HLMs. Preincubation of macrophages with brefeldin A, known to disrupt the Golgi apparatus, inhibited TSLP release induced by LPS and IL-4. lfTSLP concentration-dependently induced the release of vascular endothelial growth factor-A (VEGF-A), the most potent angiogenic factor, from HLMs. sfTSLP neither activated nor interfered with the activating property of lfTSLP on macrophages. CONCLUSIONS Our results highlight a novel immunologic circuit between HLMs and TSLP. Given the central role of macrophages in airway inflammation, this autocrine loop holds potential translational relevance in understanding innovative aspects of the pathobiology of asthma and chronic inflammatory lung disorders.
Collapse
Affiliation(s)
- Luisa Canè
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Francesco Palestra
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Marinella Pirozzi
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Seetharaman Parashuraman
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Ilaria Iacobucci
- CEINGE Advanced Biotechnologies, Naples, Italy; Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Antonello La Rocca
- Thoracic Surgery Unit - Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Edoardo Mercadante
- Thoracic Surgery Unit - Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Piero Pucci
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Maria Monti
- CEINGE Advanced Biotechnologies, Naples, Italy; Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
| |
Collapse
|
13
|
Chen Z, Balachandran YL, Chong WP, Chan KWY. Roles of Cytokines in Alzheimer's Disease. Int J Mol Sci 2024; 25:5803. [PMID: 38891990 PMCID: PMC11171747 DOI: 10.3390/ijms25115803] [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: 04/16/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
The neuroimmune system is a collection of immune cells, cytokines, and the glymphatic system that plays a pivotal role in the pathogenesis and progression of Alzheimer's disease (AD). Of particular focus are cytokines, a group of immune signaling molecules that facilitate communication among immune cells and contribute to inflammation in AD. Extensive research has shown that the dysregulated secretion of certain cytokines (IL-1β, IL-17, IL-12, IL-23, IL-6, and TNF-α) promotes neuroinflammation and exacerbates neuronal damage in AD. However, anti-inflammatory cytokines (IL-2, IL-3, IL-33, and IL-35) are also secreted during AD onset and progression, thereby preventing neuroinflammation. This review summarizes the involvement of pro- and anti-inflammatory cytokines in AD pathology and discusses their therapeutic potential.
Collapse
Affiliation(s)
- Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
| | - Yekkuni L. Balachandran
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
| | - Wai Po Chong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Kannie W. Y. Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
- Shenzhen Research Institute, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
14
|
Bai Y, Zhou R, Xie X, Zhu A, Nan Y, Wu T, Hu X, Cao Z, Ju D, Fan J. A Novel Bifunctional Fusion Protein (Anti-IL-17A-sST2) Protects against Acute Liver Failure, Modulating the TLR4/MyD88 Pathway and NLRP3 Inflammasome Activation. Biomedicines 2024; 12:1118. [PMID: 38791080 PMCID: PMC11117730 DOI: 10.3390/biomedicines12051118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Acute liver failure (ALF) is a serious inflammatory disorder with high mortality rates, which poses a significant threat to human health. The IL-33/ST2 signal is a crucial regulator in inflammation responses associated with lipopolysaccharide (LPS)-induced macrophages. The IL-17A signaling pathway promotes the release of chemokines and inflammatory cytokines, recruiting neutrophils and T cells under LPS stimulation, thus facilitating inflammatory responses. Here, the potential therapeutic benefits of neutralizing the IL-17A signal and modulating the IL-33/ST2 signal in ALF were investigated. A novel dual-functional fusion protein, anti-IL-17A-sST2, was constructed, which displayed high purity and biological activities. The administration of anti-IL-17A-sST2 resulted in significant anti-inflammatory benefits in ALF mice, amelioration of hepatocyte necrosis and interstitial congestion, and reduction in TNF-α and IL-6. Furthermore, anti-IL-17A-sST2 injection downregulated the expression of TLR4 and NLRP3 as well as important molecules such as MyD88, caspase-1, and IL-1β. The results suggest that anti-IL-17A-sST2 reduced the secretion of inflammatory factors, attenuated the inflammatory response, and protected hepatic function by regulating the TLR4/MyD88 pathway and inhibiting the NLRP3 inflammasome, providing a new therapeutic approach for ALF.
Collapse
Affiliation(s)
- Yu Bai
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Rongrui Zhou
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xinlei Xie
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - An Zhu
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yanyang Nan
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tao Wu
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiaozhi Hu
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zhonglian Cao
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dianwen Ju
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Fudan Zhangjiang Institute, Shanghai 201203, China
| | - Jiajun Fan
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Fudan Zhangjiang Institute, Shanghai 201203, China
- Shanghai Hailu Biological Technology Co., Ltd., Shanghai 201200, China
| |
Collapse
|
15
|
Li J, Wang F, Meng C, Zhu D. Role of TRPV1 and TRPA1 in TSLP production in nasal epithelial cells. Int Immunopharmacol 2024; 131:111916. [PMID: 38522138 DOI: 10.1016/j.intimp.2024.111916] [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: 10/16/2023] [Revised: 02/06/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND TRP protein is sensitive to external temperature changes, but its pathogenic mechanism in the upper airway mucosa is still unclear. OBJECTIVE To investigate the mechanism of TRPV1and TRPA1 in regulating the secretion of inflammatory factors in nasal epithelial cells. METHODS The expression of TRPV1 and TRPA1 in nasal mucosal epithelial cells was investigated using immunofluorescence assays. Epithelial cells were stimulated with TRPV1 and TRPA1 agonists and antagonists, and changes in Ca2+ release and inflammatory factor secretion in epithelial cells were detected. TSLP secretion stimulated with the calcium chelating agent EGTA was evaluated. The transcription factor NFAT was observed by immunofluorescence staining. RESULTS TRPV1 and TRPA1 expression was detected in nasal epithelial cells, and Ca2+ influx was increased after stimulation with agonists. After the activation of TRPV1 and TRPA1, the gene expression of TSLP, IL-25, and IL-33 and the protein expression levels of TSLP and IL-33 were increased, and only TSLP could be inhibited by antagonists and siRNAs. After administration of EGTA, the secretion of TSLP was inhibited significantly, and the expression of the transcription factor NFAT in the nucleus was observed after activation of the TRPV1 and TRPA1 proteins in epithelial cells. CONCLUSION Activation of TRPV1 and TRPA1 on nasal epithelial cells stimulates the generation of TSLP through the Ca2+/NFAT pathway. It also induces upregulation of IL-25 and IL-33 gene expression levels and increased levels of IL-33 protein, leading to the development of airway inflammation.
Collapse
Affiliation(s)
- Jiani Li
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Fang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Cuida Meng
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases, China
| | - Dongdong Zhu
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases, China.
| |
Collapse
|
16
|
Jou E. Clinical and basic science aspects of innate lymphoid cells as novel immunotherapeutic targets in cancer treatment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 209:1-60. [PMID: 39461748 DOI: 10.1016/bs.pmbts.2024.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Immunotherapy has revolutionised cancer treatment over the past decade, demonstrating remarkable efficacy across a broad range of cancer types. However, not all patients or cancer types respond to contemporary clinically-utilised immunotherapeutic strategies, which largely focus on harnessing adaptive immune T cells for cancer treatment. Accordingly, it is increasingly recognised that upstream innate immune pathways, which govern and orchestrate the downstream adaptive immune response, may prove critical in overcoming cancer immunotherapeutic resistance. Innate lymphoid cells (ILCs) are the most recently discovered major innate immune cell population. They have overarching roles in homeostasis and orchestrating protective immunity against pathogens. As innate immune counterparts of adaptive immune T cells, ILCs exert effector functions through the secretion of cytokines and direct cell-to-cell contact, with broad influence on the overall immune response. Importantly, dysregulation of ILC subsets have been associated with a range of diseases, including immunodeficiency disorders, allergy, autoimmunity, and more recently, cancer. ILCs may either promote or inhibit cancer initiation and progression depending on the cancer type and the specific ILC subsets involved. Critically, therapeutic targeting of ILCs and their associated cytokines shows promise against a wide range of cancer types in both preclinical models and early phase oncology clinical trials. This chapter provides a comprehensive overview of the current understanding of ILC subsets and the associated cytokines they produce in cancer pathogenesis, with specific focus on how these innate pathways are, or can be targeted, therapeutically to overcome therapeutic resistance and ultimately improve patient care.
Collapse
Affiliation(s)
- Eric Jou
- Department of Oncology, Oxford University Hospitals, University of Oxford, Oxford, United Kingdom; Kellogg College, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
17
|
He PY, Wu MY, Zheng LY, Duan Y, Fan Q, Zhu XM, Yao YM. Interleukin-33/serum stimulation-2 pathway: Regulatory mechanisms and emerging implications in immune and inflammatory diseases. Cytokine Growth Factor Rev 2024; 76:112-126. [PMID: 38155038 DOI: 10.1016/j.cytogfr.2023.12.001] [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: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 12/30/2023]
Abstract
Interleukin (IL)- 33, a nuclear factor and pleiotropic cytokine of the IL-1 family, is gaining attention owing to its important role in chronic inflammatory and autoimmune diseases. This review extends our knowledge of the effects exerted by IL-33 on target cells by binding to its specific receptor serum stimulation-2 (ST2). Depending on the tissue context, IL-33 performs multiple functions encompassing host defence, immune response, initiation and amplification of inflammation, tissue repair, and homeostasis. The levels and activity of IL-33 in the body are controlled by complex IL-33-targeting regulatory pathways. The unique temporal and spatial expression patterns of IL-33 are associated with host homeostasis and the development of immune and inflammatory disorders. Therefore, understanding the origin, function, and processes of IL-33 under various conditions is crucial. This review summarises the regulatory mechanisms underlying the IL-33/ST2 signalling axis and its potential role and clinical significance in immune and inflammatory diseases, and discusses the current complex and conflicting findings related to IL-33 in host responses.
Collapse
Affiliation(s)
- Peng-Yi He
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300071, China
| | - Meng-Yao Wu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Yu Duan
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Qi Fan
- Emergency Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Xiao-Mei Zhu
- Tissue Repair and Regeneration Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
18
|
Bao T, Liu X, Hu J, Ma M, Li J, Cao L, Yu B, Cheng H, Zhao S, Tian Z. Recruitment of PVT1 Enhances YTHDC1-Mediated m6A Modification of IL-33 in Hyperoxia-Induced Lung Injury During Bronchopulmonary Dysplasia. Inflammation 2024; 47:469-482. [PMID: 37917328 PMCID: PMC11074042 DOI: 10.1007/s10753-023-01923-1] [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: 07/06/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease that specifically affects preterm infants. Oxygen therapy administered to treat BPD can lead to hyperoxia-induced lung injury, characterized by apoptosis of lung alveolar epithelial cells. Our epitranscriptomic microarray analysis of normal mice lungs and hyperoxia-stimulated mice lungs revealed elevated RNA expression levels of IL-33, as well as increased m6A RNA methylation levels of IL-33 and PVT1 in the hyperoxia-stimulated lungs. This study aimed to investigate the role of the PVT1/IL-33 axis in BPD. A mouse model of BPD was established through hyperoxia induction, and lung histological changes were assessed by hematoxylin-eosin staining. Parameters such as radial alveolar count and mean chord length were measured to assess lung function. Mouse and human lung alveolar epithelial cells (MLE12 and A549, respectively) were stimulated with hyperoxia to create an in vitro BPD model. Cell apoptosis was detected using Western blotting and flow cytometry analysis. Our results demonstrated that silencing PVT1 suppressed apoptosis in MLE12 and A549 cells and improved lung function in hyperoxia-stimulated lungs. Additionally, IL-33 reversed the effects of PVT1 both in vivo and in vitro. Through online bioinformatics analysis and RNA-binding protein immunoprecipitation assays, YTHDC1 was identified as a RNA-binding protein (RBP) for both PVT1 and IL-33. We found that PVT1 positively regulated IL-33 expression by recruiting YTHDC1 to mediate m6A modification of IL-33. In conclusion, silencing PVT1 demonstrated beneficial effects in alleviating BPD by facilitating YTHDC1-mediated m6A modification of IL-33. Inhibition of the PVT1/IL-33 axis to suppress apoptosis in lung alveolar epithelial cells may hold promise as a therapeutic approach for managing hyperoxia-induced lung injury in BPD.
Collapse
Affiliation(s)
- Tianping Bao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Xiangye Liu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Jian Hu
- Department of Pediatrics, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Mengmeng Ma
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Jingyan Li
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Linxia Cao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Bingrui Yu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Huaiping Cheng
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Sai Zhao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China.
| | - Zhaofang Tian
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China.
| |
Collapse
|
19
|
Casciaro M, Gangemi S, Caramori G, Nucera F, Tuccari G, Ieni A. IL-33 immunohistochemical pattern of expression in neoplastic and nonneoplastic peripheral lung tissues of stage 1 o 2 lung adenocarcinoma. Pathol Res Pract 2024; 255:155208. [PMID: 38359512 DOI: 10.1016/j.prp.2024.155208] [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: 11/20/2023] [Revised: 02/01/2024] [Accepted: 02/11/2024] [Indexed: 02/17/2024]
Abstract
IL-33 is a multifaceted cytokine, plays a pivotal role in various biological processes, making it a subject of extensive research and intrigue in the field of immunology. This cytokine acts as a key regulator, effectively putting the brakes on proinflammatory nuclear factor-kappa B (NF-κB), thereby modulating chromatin compaction by promoting nucleosome-to-nucleosome interactions. IL-33's influence extends to the realm of innate and acquired immunity through its binding to the membrane-bound ST2 molecule (ST2L) of the IL-33R complex, which is expressed on various immune cells, such as Th2 cells, mast cells, natural killer cells, myeloid cells, and dendritic cells. IL-33's role in inflammation is far from one-dimensional, as it has been found to have a dual role in inflammatory disorders. In the quest to understand the origins of IL-33, immunohistochemical examination of lung tissue samples from patients with adenocarcinoma could shed light on its presence in bronchial epithelial and vascular endothelial cells, in lung tissue cancerous lesions. For this reason, we conducted a pilot study about the immunohistochemical expression of IL-33 in surgical specimens of stage 1 o 2 lung adenocarcinoma received after lung resection surgery.Our results demonstrated that patients had nuclear IL-33 immunopositivity in the alveolar pneumocytes of the normal lung tissue at the periphery of lung adenocarcinoma specimen. Note the evident negativity of the neoplastic adenocarcinoma cells. Other data showed IL-33 nuclear immunoexpression in endothelial cells of intratumoral vascular structures.This finding could indicate that IL-33 might be involved in regulating blood vessel formation and maintenance within the tumor, which is a critical factor in tumor growth and progression.The presence of IL-33 in normal lung tissue and intratumoral vascular structures may be related to its physiological functions in these contexts, while its absence in neoplastic adenocarcinoma cells could indicate a potential loss of regulatory control, which might have implications for the development and progression of the tumor.
Collapse
Affiliation(s)
- Marco Casciaro
- Unit of Allergy and Clinical Immunology, Department of Medical Sciences, University Hospital of Messina, 9125 Messina, Italy
| | - Sebastiano Gangemi
- School and Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital of Messina, University of Messina, 98125 Messina, Italy
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Medicina e Chirurgia, 43126 Parma, Italy
| | - Francesco Nucera
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, 98125 Messina, Italy
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Section of Pathology, University of Messina, 98125 Messina, Italy
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Section of Pathology, University of Messina, 98125 Messina, Italy.
| |
Collapse
|
20
|
Hayashi R, Srisomboon Y, Iijima K, Maniak PJ, Tei R, Kobayashi T, Matsunaga M, Luo H, Masuda MY, O'Grady SM, Kita H. Cholinergic sensing of allergen exposure by airway epithelium promotes type 2 immunity in the lungs. J Allergy Clin Immunol 2024; 153:793-808.e2. [PMID: 38000698 PMCID: PMC10939907 DOI: 10.1016/j.jaci.2023.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/26/2023] [Accepted: 10/09/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND Nonneuronal cells, including epithelial cells, can produce acetylcholine (ACh). Muscarinic ACh receptor antagonists are used clinically to treat asthma and other medical conditions; however, knowledge regarding the roles of ACh in type 2 immunity is limited. OBJECTIVE Our aim was to investigate the roles of epithelial ACh in allergic immune responses. METHODS Human bronchial epithelial (HBE) cells were cultured with allergen extracts, and their ACh production and IL-33 secretion were studied in vitro. To investigate immune responses in vivo, naive BALB/c mice were treated intranasally with different muscarinic ACh receptor antagonists and then exposed intranasally to allergens. RESULTS At steady state, HBE cells expressed cellular components necessary for ACh production, including choline acetyltransferase and organic cation transporters. Exposure to allergens caused HBE cells to rapidly release ACh into the extracellular medium. Pharmacologic or small-interfering RNA-based blocking of ACh production or autocrine action through the M3 muscarinic ACh receptors in HBE cells suppressed allergen-induced ATP release, calcium mobilization, and extracellular secretion of IL-33. When naive mice were exposed to allergens, ACh was quickly released into the airway lumen. A series of clinical M3 muscarinic ACh receptor antagonists inhibited allergen-induced IL-33 secretion and innate type 2 immune response in the mouse airways. In a preclinical murine model of asthma, an ACh receptor antagonist suppressed allergen-induced airway inflammation and airway hyperreactivity. CONCLUSIONS ACh is released quickly by airway epithelial cells on allergen exposure, and it plays an important role in type 2 immunity. The epithelial ACh system can be considered a therapeutic target in allergic airway diseases.
Collapse
Affiliation(s)
- Ryusuke Hayashi
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Yotesawee Srisomboon
- Department of Animal Science, University of Minnesota, St Paul, Minn; Department of Integrative Biology and Physiology, University of Minnesota, St Paul, Minn
| | - Koji Iijima
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Peter J Maniak
- Department of Animal Science, University of Minnesota, St Paul, Minn; Department of Integrative Biology and Physiology, University of Minnesota, St Paul, Minn
| | - Rinna Tei
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Takao Kobayashi
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Mayumi Matsunaga
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Huijun Luo
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Mia Y Masuda
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minn; Mayo Clinic Graduate School of Biomedical Sciences, Scottsdale, Ariz
| | - Scott M O'Grady
- Department of Animal Science, University of Minnesota, St Paul, Minn; Department of Integrative Biology and Physiology, University of Minnesota, St Paul, Minn
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Ariz; Department of Medicine, Mayo Clinic, Scottsdale, Ariz; Department of Immunology, Mayo Clinic Rochester, Rochester, Minn; Department of Immunology, Mayo Clinic Arizona, Scottsdale, Ariz.
| |
Collapse
|
21
|
Qin Z, Xie L, Li W, Wang C, Li Y. New Insights into Mechanisms Traditional Chinese Medicine for Allergic Rhinitis by Regulating Inflammatory and Oxidative Stress Pathways. J Asthma Allergy 2024; 17:97-112. [PMID: 38405022 PMCID: PMC10888064 DOI: 10.2147/jaa.s444923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
Allergy rhinitis (AR) is becoming more common and has serious medical and societal consequences. Sneezing, paroxysmal nasal blockage, nasal itching, mucosal edema, coughing, and rhinorrhea are symptoms of this type I allergic immunological illness. Immunoglobulin E-mediated inflammation is the cause of it. Because AR is prone to recurrent attacks, extended medication therapy may impair its effectiveness. In addition to negatively affecting the patients' physical health, this can also negatively impact their mental health. During AR development, there are inflammatory and oxidative stress responses that are linked to problems in a number of signal transduction pathways. By using the terms "allergic rhinitis", "traditional Chinese medicine", "inflammation", and "oxidative stress", we screened for pertinent research published over the previous five years in databases like PubMed. We saw that NF-KB, TLR, IL-33/ST2, PI3K/AKT, MAPK, and Nrf2 are some of the most important inflammatory and oxidative stress pathways in AR. Studies have revealed that antioxidant and anti-inflammatory therapy reduced the risk of AR and was therapeutic; however, the impact of the therapy varies widely. The Chinese medical system places a high value on traditional Chinese medicine (TCM), which has been there for virtually all of China's 5000-year history. By influencing signaling pathways related to inflammation and oxidative stress, Chinese herbal medicine and its constituent compounds have been shown to prevent allergic rhinitis. This review will focus on this evidence and provide references for clinical treatment and scientific research applications.
Collapse
Affiliation(s)
- Zhu Qin
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Liangzhen Xie
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Wentao Li
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Chao Wang
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Yan Li
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
22
|
Janjetovic Z, Qayyum S, Reddy SB, Podgorska E, Scott SG, Szpotan J, Mobley AA, Li W, Boda VK, Ravichandran S, Tuckey RC, Jetten AM, Slominski AT. Novel Vitamin D3 Hydroxymetabolites Require Involvement of the Vitamin D Receptor or Retinoic Acid-Related Orphan Receptors for Their Antifibrogenic Activities in Human Fibroblasts. Cells 2024; 13:239. [PMID: 38334631 PMCID: PMC10854953 DOI: 10.3390/cells13030239] [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: 11/27/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
We investigated multiple signaling pathways activated by CYP11A1-derived vitamin D3 hydroxymetabolites in human skin fibroblasts by assessing the actions of these molecules on their cognate receptors and by investigating the role of CYP27B1 in their biological activities. The actions of 20(OH)D3, 20,23(OH)2D3, 1,20(OH)2D3 and 1,20,23(OH)3D3 were compared to those of classical 1,25(OH)2D3. This was undertaken using wild type (WT) fibroblasts, as well as cells with VDR, RORs, or CYP27B1 genes knocked down with siRNA. Vitamin D3 hydroxymetabolites had an inhibitory effect on the proliferation of WT cells, but this effect was abrogated in cells with silenced VDR or RORs. The collagen expression by WT cells was reduced upon secosteroid treatment. This effect was reversed in cells where VDR or RORs were knocked down where the inhibition of collagen production and the expression of anti-fibrotic genes in response to the hydroxymetabolites was abrogated, along with ablation of their anti-inflammatory action. The knockdown of CYP27B1 did not change the effect of either 20(OH)D3 or 20,23(OH)2D3, indicating that their actions are independent of 1α-hydroxylation. In conclusion, the expression of the VDR and/or RORα/γ receptors in fibroblasts is necessary for the inhibition of both the proliferation and fibrogenic activity of hydroxymetabolites of vitamin D3, while CYP27B1 is not required.
Collapse
Affiliation(s)
- Zorica Janjetovic
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Shariq Qayyum
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
- Brigham’s Women’s Hospital, Harvard University, Boston, MA 02115, USA
| | - Sivani B. Reddy
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Ewa Podgorska
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - S. Gates Scott
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Justyna Szpotan
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Alisa A. Mobley
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Wei Li
- College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA; (W.L.); (V.K.B.)
| | - Vijay K. Boda
- College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA; (W.L.); (V.K.B.)
| | - Senthilkumar Ravichandran
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Robert C. Tuckey
- School of Molecular Science, The University of Western Australia, Perth 6009, Australia;
| | - Anton M. Jetten
- Cell Biology Section, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA;
| | - Andrzej T. Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
- Cancer Chemoprevention Program, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- VA Medical Center, Birmingham, AL 35294, USA
| |
Collapse
|
23
|
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: 3] [Impact Index Per Article: 3.0] [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
|
24
|
Pathak MP, Patowary P, Chattopadhyay P, Barbhuiyan PA, Islam J, Gogoi J, Wankhar W. Obesity-associated Airway Hyperresponsiveness: Mechanisms Underlying Inflammatory Markers and Possible Pharmacological Interventions. Endocr Metab Immune Disord Drug Targets 2024; 24:1053-1068. [PMID: 37957906 DOI: 10.2174/0118715303256440231028072049] [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: 04/08/2023] [Revised: 08/14/2023] [Accepted: 09/15/2023] [Indexed: 11/15/2023]
Abstract
Obesity is rapidly becoming a global health problem affecting about 13% of the world's population affecting women and children the most. Recent studies have stated that obese asthmatic subjects suffer from an increased risk of asthma, encounter severe symptoms, respond poorly to anti-asthmatic drugs, and ultimately their quality-of-life decreases. Although, the association between airway hyperresponsiveness (AHR) and obesity is a growing concern among the public due to lifestyle and environmental etiologies, however, the precise mechanism underlying this association is yet to establish. Apart from aiming at the conventional antiasthmatic targets, treatment should be directed towards ameliorating obesity pathogenesis too. Understanding the pathogenesis underlying the association between obesity and AHR is limited, however, a plethora of obesity pathologies have been reported viz., increased pro-inflammatory and decreased anti-inflammatory adipokines, depletion of ROS controller Nrf2/HO-1 axis, NLRP3 associated macrophage polarization, hypertrophy of WAT, and down-regulation of UCP1 in BAT following down-regulated AMPKα and melanocortin pathway that may be correlated with AHR. Increased waist circumference (WC) or central obesity was thought to be related to severe AHR, however, some recent reports suggest body mass index (BMI), not WC tends to exaggerate airway closure in AHR due to some unknown mechanisms. This review aims to co-relate the above-mentioned mechanisms that may explain the copious relation underlying obesity and AHR with the help of published reports. A proper understanding of these mechanisms discussed in this review will ensure an appropriate treatment plan for patients through advanced pharmacological interventions.
Collapse
Affiliation(s)
| | - Pompy Patowary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, India
| | | | | | - Johirul Islam
- Department of Pharmaceutical Sciences, School of Health Sciences, Assam Kaziranga University, Jorhat, India
| | - Jyotchna Gogoi
- Department of Biochemistry, Faculty of Science, Assam Down Town University, Guwahati, India
| | - Wankupar Wankhar
- Department of Dialysis, Faculty of Paramedical Science, Assam Down Town University, Guwahati, India
| |
Collapse
|
25
|
Zhou Y, Xu Z, Liu Z. Role of IL-33-ST2 pathway in regulating inflammation: current evidence and future perspectives. J Transl Med 2023; 21:902. [PMID: 38082335 PMCID: PMC10714644 DOI: 10.1186/s12967-023-04782-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Interleukin (IL)-33 is an alarmin of the IL-1 superfamily localized to the nucleus of expressing cells, such as endothelial cells, epithelial cells, and fibroblasts. In response to cellular damage or stress, IL-33 is released and activates innate immune responses in some immune and structural cells via its receptor interleukin-1 receptor like-1 (IL-1RL1 or ST2). Recently, IL-33 has become a hot topic of research because of its role in pulmonary inflammation. The IL-33-ST2 signaling pathway plays a pro-inflammatory role by activating the type 2 inflammatory response, producing type 2 cytokines and chemokines. Elevated levels of IL-33 and ST2 have been observed in chronic pulmonary obstructive disease (COPD). Notably, IL-33 is present in COPD induced by cigarette smoke or acute inflammations. The role of IL-33 in sepsis is becoming increasingly prominent, and understanding its significance in the treatment of sepsis associated with high mortality is critical. In addition to its pro-inflammatory effects, the IL-33-ST2 axis appears to play a role in bacterial clearance and tissue repair. In this review, we focused on the role of the IL-33-ST2 axis in sepsis, asthma, and COPD and summarized the therapeutic targets associated with this axis, providing a basis for future treatment.
Collapse
Affiliation(s)
- Yilu Zhou
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhendong Xu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zhiqiang Liu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
26
|
Saki N, Javan M, Moghimian-Boroujeni B, Kast RE. Interesting effects of interleukins and immune cells on acute respiratory distress syndrome. Clin Exp Med 2023; 23:2979-2996. [PMID: 37330918 DOI: 10.1007/s10238-023-01118-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/10/2023] [Indexed: 06/20/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a medical condition characterized by widespread inflammation in the lungs with consequent proportional loss of gas exchange function. ARDS is linked with severe pulmonary or systemic infection. Several factors, including secretory cytokines, immune cells, and lung epithelial and endothelial cells, play a role in the development and progression of this disease. The present study is based on Pubmed database information (1987-2022) using the words "Acute respiratory distress syndrome", "Interleukin", "Cytokines" and "Immune cells". Cytokines and immune cells play an important role in this disease, with particular emphasis on the balance between pro-inflammatory and anti-inflammatory factors. Neutrophils are one of several important mediators of Inflammation, lung tissue destruction, and malfunction during ARDS. Some immune cells, such as macrophages and eosinophils, play a dual role in releasing inflammatory mediators, recruitment inflammatory cells and the progression of ARDS, or releasing anti-inflammatory mediators, clearing the lung of inflammatory cells, and helping to improve the disease. Different interleukins play a role in the development or inhibition of ARDS by helping to activate various signaling pathways, helping to secrete other inflammatory or anti-inflammatory interleukins, and playing a role in the production and balance between immune cells involved in ARDS. As a result, immune cells and, inflammatory cytokines, especially interleukins play an important role in the pathogenesis of this disease Therefore, understanding the relevant mechanisms will help in the proper diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammadreza Javan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Bahareh Moghimian-Boroujeni
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, 61357-15794, Iran.
| | | |
Collapse
|
27
|
Jiang C, Jin X, Li C, Wen L, Wang Y, Li X, Zhang Z, Tan R. Roles of IL-33 in the Pathogenesis of Cardiac Disorders. Exp Biol Med (Maywood) 2023; 248:2167-2174. [PMID: 37828753 PMCID: PMC10800126 DOI: 10.1177/15353702231198075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023] Open
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 cytokine family and is believed to play important roles in different diseases by binding to its specific receptor suppression of tumorigenicity 2 (ST2). In the heart, IL-33 is expressed in different cells including cardiomyocytes, fibroblasts, endothelium, and epithelium. Although many studies have been devoted to investigating the effects of IL-33 on heart diseases, its roles in myocardial injuries remain obscure, and thus further studies are mandatory to unravel the underlying molecular mechanisms. We highlighted the current knowledge of the molecular and cellular characteristics of IL-33 and then summarized its major roles in different myocardial injuries, mainly focusing on infection, heart transplantation, coronary atherosclerosis, myocardial infarction, and diabetic cardiomyopathy. This narrative review will summarize current understanding and insights regarding the implications of IL-33 in cardiac diseases and its diagnostic and therapeutic potential for cardiac disease management.
Collapse
Affiliation(s)
- Chunjie Jiang
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Xuemei Jin
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
- Department of Preventive Medicine, School of Medicine, Yanbian University, Yanji 133002, China
| | - Chunlei Li
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Luona Wen
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Yuqi Wang
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Xiaojian Li
- Department of Burns, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220 China
| | - Zhi Zhang
- Department of Burns, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220 China
| | - Rongshao Tan
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| |
Collapse
|
28
|
Wang Z, Tang N. Unpacking the complexity of nuclear IL-33 (nIL-33): a crucial regulator of transcription and signal transduction. J Cell Commun Signal 2023:10.1007/s12079-023-00788-1. [PMID: 37878185 DOI: 10.1007/s12079-023-00788-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023] Open
Abstract
Interleukin-33 (IL-33) (NF-HEV), a chromatin-associated nuclear cytokine, is a member of the IL-1 family. IL-33 possesses a nuclear localization signal and a homeodomain (a structure resembling a helix-turn-helix) that can bind to nuclear chromatin. Research has revealed that IL-33 can function as a nuclear factor to regulate various biological processes. This review discusses the cellular localization, functional effects, and immune regulation of full length IL-33 (FLIL-33), cytokine IL-33 (sIL-33) and nuclear IL-33 (nIL-33). In addition, the post-translational modifications of nIL-33 and the hypothesis of using nIL-33 as a treatment method were also summarized. A multidisciplinary approach is required which integrates methods and techniques from genomics, proteomics, cell biology and immunology to provide comprehensive insights into the function and therapeutic potential of nIL-33.
Collapse
Affiliation(s)
- Zengbin Wang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Nanhong Tang
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
29
|
Gao TC, Wang CH, Wang YQ, Mi WL. IL-33/ST2 Signaling in the Pathogenesis of Chronic Pain and Itch. Neuroscience 2023; 529:16-22. [PMID: 37574108 DOI: 10.1016/j.neuroscience.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/15/2023]
Abstract
Interleukin-33 (IL-33) is an inflammatory factor with an extensive range of biological effects and pleiotropic roles in diseases. Evidence suggests that IL-33 and its receptor ST2 play a pivotal role in chronic pain and itch at the level of primary sensory neurons, the spinal cord, and the brain. In this review, we outline an evolving understanding of the roles and mechanisms of IL-33 in chronic pathological pain, including inflammatory, neuropathic, and cancer, and chronic pruritus, such as allergic contact dermatitis, atopic dermatitis, and dry skin. Understanding the key roles of IL-33/ST2 signaling may provide exciting insights into the mechanisms of chronic pain and itch and lead to new clues for therapeutic approaches to the resolution of chronic pain and itch.
Collapse
Affiliation(s)
- Tian-Chi Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Cheng-Hao Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wen-Li Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
30
|
Tsuji G, Yamamura K, Kawamura K, Kido-Nakahara M, Ito T, Nakahara T. Regulatory Mechanism of the IL-33-IL-37 Axis via Aryl Hydrocarbon Receptor in Atopic Dermatitis and Psoriasis. Int J Mol Sci 2023; 24:14633. [PMID: 37834081 PMCID: PMC10572928 DOI: 10.3390/ijms241914633] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Interleukin (IL)-33 and IL-37 have been identified as novel cytokines involved in various inflammatory diseases. However, their specific roles remain largely unknown. Recent studies have shown that IL-33, which triggers inflammation, and IL-37, which suppresses it, cooperatively regulate the balance between inflammation and anti-inflammation. IL-33 and IL-37 are also deeply involved in the pathogenesis of inflammatory skin diseases such as atopic dermatitis (AD) and psoriasis. Furthermore, a signaling pathway by which aryl hydrocarbon receptor (AHR), a receptor for dioxins, regulates the expression of IL-33 and IL-37 has been revealed. Here, we outline recent findings on the mechanisms regulating IL-33 and IL-37 expression in AD and psoriasis. IL-33 expression is partially dependent on mitogen-activated protein kinase (MAPK) activation, and IL-37 has a role in suppressing MAPK in human keratinocytes. Furthermore, IL-33 downregulates skin barrier function proteins including filaggrin and loricrin, thereby downregulating the expression of IL-37, which colocalizes with these proteins. This leads to an imbalance of the IL-33-IL-37 axis, involving increased IL-33 and decreased IL-37, which may be associated with the pathogenesis of AD and psoriasis. Therefore, AHR-mediated regulation of the IL-33-IL-37 axis may lead to new therapeutic strategies for the treatment of AD and psoriasis.
Collapse
Affiliation(s)
- Gaku Tsuji
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Kazuhiko Yamamura
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Koji Kawamura
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Makiko Kido-Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Takeshi Nakahara
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| |
Collapse
|
31
|
Stojanovic B, Gajovic N, Jurisevic M, Stojanovic MD, Jovanovic M, Jovanovic I, Stojanovic BS, Milosevic B. Decoding the IL-33/ST2 Axis: Its Impact on the Immune Landscape of Breast Cancer. Int J Mol Sci 2023; 24:14026. [PMID: 37762328 PMCID: PMC10531367 DOI: 10.3390/ijms241814026] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Interleukin-33 (IL-33) has emerged as a critical cytokine in the regulation of the immune system, showing a pivotal role in the pathogenesis of various diseases including cancer. This review emphasizes the role of the IL-33/ST2 axis in breast cancer biology, its contribution to cancer progression and metastasis, its influence on the tumor microenvironment and cancer metabolism, and its potential as a therapeutic target. The IL-33/ST2 axis has been shown to have extensive pro-tumorigenic features in breast cancer, starting from tumor tissue proliferation and differentiation to modulating both cancer cells and anti-tumor immune response. It has also been linked to the resistance of cancer cells to conventional therapeutics. However, the role of IL-33 in cancer therapy remains controversial due to the conflicting effects of IL-33 in tumorigenesis and anti-tumor response. The possibility of targeting the IL-33/ST2 axis in tumor immunotherapy, or as an adjuvant in immune checkpoint blockade therapy, is discussed.
Collapse
Affiliation(s)
- Bojan Stojanovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.)
| | - Nevena Gajovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia (I.J.)
| | - Milena Jurisevic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Milica Dimitrijevic Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia (I.J.)
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Marina Jovanovic
- Department of Otorinolaringology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia (I.J.)
| | - Bojana S. Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia (I.J.)
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Bojan Milosevic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.)
| |
Collapse
|
32
|
Jiao M, Sun Y, Shi J, Zhang N, Tang X, Fan A, Liu S, Dai C, Qian Z, Zhang F, Wang C, Chen H, Zheng F. IL-33 and HMGB1 modulate the progression of EAE via oppositely regulating each other. Int Immunopharmacol 2023; 122:110653. [PMID: 37467690 DOI: 10.1016/j.intimp.2023.110653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/19/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
Interleukin-33 (IL-33) and high mobility group box 1 (HMGB1) have been reported to play crucial and distinct roles in experimental autoimmune encephalomyelitis (EAE). However, little is known about their interaction in the progression of EAE. In this study, the dynamic expression and release of IL-33 and HMGB1 in different stages of EAE in vivo, and their interaction in vitro were explored. We found that HMGB1 was dominant in pre-onset stage of EAE, while IL-33 was dominant in peak stage. Moreover, both blockade of extracellular HMGB1 in the central nervous system (CNS) and conditional knockout of HMGB1 in astrocytes decreased IL-33 release. HMGB1 promoted the release of IL-33, while IL-33 reduced the release of HMGB1 from primary astrocytes in vitro. Taken together, IL-33 and HMGB1 in the CNS jointly participate in the EAE progression and the inhibitory effect of IL-33 on HMGB1 may be involved in the self-limiting of EAE.
Collapse
Affiliation(s)
- Mengya Jiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yan Sun
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central Minzu University, Wuhan 430074, China; College of Life Sciences, South-Central Minzu University, Wuhan 430074, China
| | - Junyu Shi
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Na Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xuhuan Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Anqi Fan
- College of Life Science, Yangtze University, Jingzhou 434025, China
| | - Shiwang Liu
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chan Dai
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhigang Qian
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Feng Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chenchen Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Huoying Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin 541199, China; Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin 541199, China.
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China.
| |
Collapse
|
33
|
Chatterjee A, Azevedo-Martins JM, Stachler MD. Interleukin-33 as a Potential Therapeutic Target in Gastric Cancer Patients: Current Insights. Onco Targets Ther 2023; 16:675-687. [PMID: 37583706 PMCID: PMC10424681 DOI: 10.2147/ott.s389120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/06/2023] [Indexed: 08/17/2023] Open
Abstract
Gastric cancer is a significant global health problem as it is the fifth most prevalent cancer worldwide and the fourth leading cause of cancer-related mortality. While cytotoxic chemotherapy remains the primary treatment for advanced GC, response rates are limited. Recent progresses, focused on molecular signalling within gastric cancer, have ignited new hope for potential therapeutic targets that may improve survival and/or reduce the toxic effects of traditional therapies. Carcinomas are generally initiated when critical regulatory genes get mutated, but the progression to malignancy is usually supported by the non-neoplastic cells that create a conducive environment for transformation and progression to occur. Interleukin 33 (IL-33) functions as a dual activity cytokine as it is also a nuclear factor. IL-33 is usually present in the nuclei of the cells. Upon tissue damage, it is released into the extracellular space and binds to its receptor, suppression of tumorigenicity 2 (ST2) L, which is expressed on the membranes of the target cells. IL-33 signalling activates the T Helper 2 (Th2) immune response among other responses. Although the studies on the role of IL-33 in gastric cancer are still in the early stages, they have revealed potentially important (though sometimes conflicting) functions or roles in cancer development and progression. The pro-tumorigenic roles include induction and the recruitment of tumor-associated immune cells, promoting metaplasia progression, and inducing stem cell like and EMT properties in gastric cancer cells. Therapeutic interventions to disrupt these functions may provide a unique strategy for gastric cancer prevention and treatment. This review aims to provide a summary of the role of IL-33 in GC, state its multiple functions in relation to GC, and show potential avenues for promising therapeutic investigation.
Collapse
Affiliation(s)
- Annesha Chatterjee
- University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| | | | - Matthew D Stachler
- University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| |
Collapse
|
34
|
Zhu X, Lu J, Rao J, Ru D, Gao M, Shi D, Cao K, Wen S, Dai C, Wang X, Mi W, Liu L, Zhou H. Crosstalk between Interleukin-1 Receptor-Like 1 and Transforming Growth Factor-β Receptor Signaling Promotes Renal Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1029-1045. [PMID: 37236504 DOI: 10.1016/j.ajpath.2023.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/18/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023]
Abstract
IL-33, a member of the IL-1 family, acts as an alarmin in immune response. Epithelial-mesenchymal transition and transforming growth factor-β (TGF-β)–induced fibroblast activation are key events in the development of renal interstitial fibrosis. The current study found increased expression of IL-33 and interleukin-1 receptor-like 1 (IL1RL1, alias ST2), the receptor for IL-33, in human fibrotic renal tissues. In addition, IL-33– or ST2-deficient mice showed significantly reduced levels of fibronectin, α-smooth muscle actin, and vimentin, and increased E-cadherin levels. In HK-2 cells, IL-33 promotes the phosphorylation of the TGF-β receptor (TGF-βR), Smad2, and Smad3, and the production of extracellular matrix (ECM), with reduced expression of E-cadherin. Blocking TGF-βR signaling or suppressing ST2 expression impeded Smad2 and Smad3 phosphorylation, thereby reducing ECM production, suggesting that IL-33–induced ECM synthesis requires cooperation between the two pathways. Mechanistically, IL-33 treatment induced a proximate interaction between ST2 and TGF-βRs, activating downstream Smad2 and Smad3 for ECM production in renal epithelial cells. Collectively, this study identified a novel and essential role for IL-33 in promoting TGF-β signaling and ECM production in the development of renal fibrosis. Therefore, targeting IL-33/ST2 signaling may be an effective therapeutic strategy for renal fibrosis.
Collapse
Affiliation(s)
- Xingxing Zhu
- Department of Immunology, Nanjing Medical University, Nanjing, China.
| | - Jiahui Lu
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Jia Rao
- Department of Immunology, Anhui Medical University, Hefei, China
| | - Dongqing Ru
- Department of Immunology, Nanjing Medical University, Nanjing, China; Central Laboratory, The Second Affiliated Hospital, Henan University of Science and Technology, Luoyang, China
| | - Mengru Gao
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Dongyan Shi
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Kelei Cao
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Shuang Wen
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Chunsun Dai
- Department of Clinical Pathology, The Fourth Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Xuerong Wang
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Wenli Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lixin Liu
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Hong Zhou
- Department of Immunology, Nanjing Medical University, Nanjing, China; Department of Immunology, Anhui Medical University, Hefei, China.
| |
Collapse
|
35
|
Palumbo A, Atzeni F, Murdaca G, Gangemi S. The Role of Alarmins in Osteoarthritis Pathogenesis: HMGB1, S100B and IL-33. Int J Mol Sci 2023; 24:12143. [PMID: 37569519 PMCID: PMC10418664 DOI: 10.3390/ijms241512143] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Osteoarthritis (OA) is a multifactorial disease in which genetics, aging, obesity, and trauma are well-known risk factors. It is the most prevalent joint disease and the largest disability problem worldwide. Recent findings have described the role of damage-associated molecular patterns (DAMPs) in the course of the disease. In particular, alarmins such as HMGB1, IL-33, and S100B, appear implicated in enhancing articular inflammation and favouring a catabolic switch in OA chondrocytes. The aims of this review are to clarify the molecular signalling of these three molecules in OA pathogenesis, to identify their possible use as staging biomarkers, and, most importantly, to find out whether they could be possible therapeutic targets. Osteoarthritic cartilage expresses increased levels of all three alarmins. HMGB1, in particular, is the most studied alarmin with increased levels in cartilage, synovium, and synovial fluid of OA patients. High levels of HMGB1 in synovial fluid of OA joints are positively correlated with radiological and clinical severity. Counteracting HMGB1 strategies have revealed improving results in articular cells from OA patients and in OA animal models. Therefore, drugs against this alarmin, such as anti-HMGB1 antibodies, could be new treatment possibilities that can modify the disease course since available medications only alleviate symptoms.
Collapse
Affiliation(s)
- Antonino Palumbo
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, 98124 Messina, Italy; (A.P.); (F.A.)
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, 98124 Messina, Italy; (A.P.); (F.A.)
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
36
|
Banks WA, Hansen KM, Erickson MA, Crews FT. High-mobility group box 1 (HMGB1) crosses the BBB bidirectionally. Brain Behav Immun 2023; 111:386-394. [PMID: 37146655 DOI: 10.1016/j.bbi.2023.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a ubiquitous protein that regulates transcription in the nucleus, and is an endogenous damage-associated molecular pattern molecule that activates the innate immune system. HMGB1 activates the TLR4 and RAGE recepto, inducing downstream signals reminiscent of cytokines that have been found to cross the blood-brain barrier (BBB). Blood HMGB1 increases in stroke, sepsis, senescence, alcohol binge drinking and other conditions. Here, we examined the ability of HMGB1 radioactively labeled with iodine (I-HMGB1) to cross the BBB. We found that I-HMGB1 readily entered into mouse brain from the circulation with a unidirectional influx rate of 0.654 μl/g-min. All brain regions tested took up I-HMGB1; uptake was greatest by the olfactory bulb and least in the striatum. Transport was not reliably inhibited by unlabeled HMGB1 nor by inhibitors of TLR4, TLR2, RAGE, or CXCR4. Uptake was enhanced by co-injection of wheatgerm agglutinin, suggestive of involvement of absorptive transcytosis as a mechanism of transport. Induction of inflammation/neuroinflammation with lipopolysaccharide is known to increase blood HMGB1; we report here that brain transport is also increased by LPS-induced inflammation. Finally, we found that I-HMGB1 was also transported in the brain-to-blood direction, with both unlabeled HMGB1 or lipopolysaccharide increasing the transport rate. These results show that HMGB1 can bidirectionally cross the BBB and that those transport rates are enhanced by inflammation. Such transport provides a mechanism by which HMGB1 levels would impact neuroimmune signaling in both the brain and periphery.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State.
| | - Kim M Hansen
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State
| | - Michelle A Erickson
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State
| | - Fulton T Crews
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, US State
| |
Collapse
|
37
|
Park JH, Mortaja M, Azin M, Nazarian RM, Demehri S. Nuclear IL-33 in Fibroblasts Promotes Skin Fibrosis. J Invest Dermatol 2023; 143:1302-1306.e4. [PMID: 36708946 PMCID: PMC10293058 DOI: 10.1016/j.jid.2022.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/27/2023]
Affiliation(s)
- Jong Ho Park
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mahsa Mortaja
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Marjan Azin
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rosalynn M Nazarian
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
38
|
Jiang T, Zheng T, Li W, Liu N, Wang M. IL-33/ST2 signaling pathway and Alzheimer's disease: A systematic review and meta-analysis. Clin Neurol Neurosurg 2023; 230:107773. [PMID: 37172376 DOI: 10.1016/j.clineuro.2023.107773] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/03/2023] [Accepted: 05/07/2023] [Indexed: 05/14/2023]
Abstract
The IL-33/ST2 signaling pathway has potential relevance for clinical identification and treatment of Alzheimer's disease (AD). Here, eight databases (including CNKI, Wanfang, SinoMed, VIP, PubMed, Cochrane library, Embase and Web of Science) were employed to search for studies on IL-33/ST2 signaling pathway and its association with AD. Totally, 15 articles were included, of which 5 studies investigated the connection between IL-33 gene polymorphisms and AD, 4 studies explored the serum IL-33 and sST2 levels in patients with AD and Mild cognitive impairment (MCI), and the exact mechanisms underlying IL-33/ST2 signaling pathway in AD were explored in 6 studies. Then, the RevMan 5.4 software was used for meta-analysis, and the related studies were systematically reviewed. The results of the meta-analysis showed that serum IL-33 levels were higher in patients with AD and MCI than in healthy controls (HC), with serum IL-33 levels in AD patients significantly higher than in MCI patients (SMD = 0.26, 95 % CI: 0.02, 0.51; P = 0.04). Compared with HC, the sST2 level was significantly higher in AD patients (SMD = 1.23, 95 % CI: 0.93, 1.53; P < 0.00001) and tended to elevate in patients with MCI. The systematic review indicated that there is a significant relationship between IL-33 gene polymorphisms and susceptibility to AD; The IL-33/ST2 signaling pathway may be one of the future treatment targets for AD. Our study provides evidence to prove that serum IL-33 and sST2 have potential clinical application value as biomarkers for identifying AD.
Collapse
Affiliation(s)
- Taotao Jiang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Ting Zheng
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Wenhao Li
- Department of neurosurgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ning Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou 730030, China.
| |
Collapse
|
39
|
Dai X, Shiraishi K, Muto J, Mori H, Murakami M, Sayama K. Nuclear IL-33 Plays an Important Role in EGFR-Mediated Keratinocyte Migration by Regulating the Activation of Signal Transducer and Activator of Transcription 3 and NF-κB. JID INNOVATIONS 2023; 3:100205. [PMID: 37441125 PMCID: PMC10333683 DOI: 10.1016/j.xjidi.2023.100205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 07/15/2023] Open
Abstract
Nuclear IL-33 levels are high at the epidermal edges of skin wounds and facilitate wound healing. However, IL-33-mediated regulation of keratinocyte (KC) biology during wound healing remains poorly understood. During skin-wound healing, KC migration and re-epithelialization are mediated predominantly by EGFR signaling activation and depend on the function of signal transducer and activator of transcription 3 (STAT3). We found that migrating KCs at the leading edges of mouse skin wounds exhibited concomitant induction and nuclear colocalization of IL-33 and phosphorylated STAT3. In cultured human KCs, activation of EGFR signaling caused rapid elevation of nuclear IL-33, which directly interacts with phosphorylated STAT3, promoting STAT3 activation. In vitro KC migration and wound-healing assays revealed that high nuclear IL-33 levels were required for KC migration and wound closure. KC mobility associated with a lack of suprabasal epidermal keratins and extracellular matrix degradation mediated by matrix metalloproteinases (MMPs) control cell migration at the intracellular and extracellular levels, respectively. In EGFR-activated KCs, nuclear IL-33 mediated keratin 1 and 10 downregulation and MMP9 upregulation by promoting STAT3 activation and limited MMP1, MMP3, and MMP10 induction by suppressing NF-κB transactivation. Thus, epidermal nuclear IL-33 is involved in KC migration and wound closure by regulating the STAT3 and NF-κB pathways.
Collapse
Affiliation(s)
- Xiuju Dai
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ken Shiraishi
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Jun Muto
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hideki Mori
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masamoto Murakami
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koji Sayama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
40
|
Duan M, Liu X, Yang Y, Zhang Y, Wu R, Lv Y, Lei H. Orchestrated regulation of immune inflammation with cell therapy in pediatric acute liver injury. Front Immunol 2023; 14:1194588. [PMID: 37426664 PMCID: PMC10323196 DOI: 10.3389/fimmu.2023.1194588] [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: 03/27/2023] [Accepted: 05/26/2023] [Indexed: 07/11/2023] Open
Abstract
Acute liver injury (ALI) in children, which commonly leads to acute liver failure (ALF) with the need for liver transplantation, is a devastating life-threatening condition. As the orchestrated regulation of immune hemostasis in the liver is essential for resolving excess inflammation and promoting liver repair in a timely manner, in this study we focused on the immune inflammation and regulation with the functional involvement of both innate and adaptive immune cells in acute liver injury progression. In the context of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic, it was also important to incorporate insights from the immunological perspective for the hepatic involvement with SARS-CoV-2 infection, as well as the acute severe hepatitis of unknown origin in children since it was first reported in March 2022. Furthermore, molecular crosstalk between immune cells concerning the roles of damage-associated molecular patterns (DAMPs) in triggering immune responses through different signaling pathways plays an essential role in the process of liver injury. In addition, we also focused on DAMPs such as high mobility group box 1 (HMGB1) and cold-inducible RNA-binding protein (CIRP), as well as on macrophage mitochondrial DNA-cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway in liver injury. Our review also highlighted novel therapeutic approaches targeting molecular and cellular crosstalk and cell-based therapy, providing a future outlook for the treatment of acute liver injury.
Collapse
Affiliation(s)
- Mingyue Duan
- Department of Clinical Laboratory, The Affiliated Children’s Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Shaanxi Institute for Pediatric Diseases, The Affiliated Children’s Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoguai Liu
- Department of Infectious Diseases, The Affiliated Children’s Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ying Yang
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Shaanxi Institute for Pediatric Diseases, The Affiliated Children’s Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yanmin Zhang
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Shaanxi Institute for Pediatric Diseases, The Affiliated Children’s Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rongqian Wu
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hong Lei
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Shaanxi Institute for Pediatric Diseases, The Affiliated Children’s Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
41
|
England E, Rees DG, Scott IC, Carmen S, Chan DTY, Chaillan Huntington CE, Houslay KF, Erngren T, Penney M, Majithiya JB, Rapley L, Sims DA, Hollins C, Hinchy EC, Strain MD, Kemp BP, Corkill DJ, May RD, Vousden KA, Butler RJ, Mustelin T, Vaughan TJ, Lowe DC, Colley C, Cohen ES. Tozorakimab (MEDI3506): an anti-IL-33 antibody that inhibits IL-33 signalling via ST2 and RAGE/EGFR to reduce inflammation and epithelial dysfunction. Sci Rep 2023; 13:9825. [PMID: 37330528 PMCID: PMC10276851 DOI: 10.1038/s41598-023-36642-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/07/2023] [Indexed: 06/19/2023] Open
Abstract
Interleukin (IL)-33 is a broad-acting alarmin cytokine that can drive inflammatory responses following tissue damage or infection and is a promising target for treatment of inflammatory disease. Here, we describe the identification of tozorakimab (MEDI3506), a potent, human anti-IL-33 monoclonal antibody, which can inhibit reduced IL-33 (IL-33red) and oxidized IL-33 (IL-33ox) activities through distinct serum-stimulated 2 (ST2) and receptor for advanced glycation end products/epidermal growth factor receptor (RAGE/EGFR complex) signalling pathways. We hypothesized that a therapeutic antibody would require an affinity higher than that of ST2 for IL-33, with an association rate greater than 107 M-1 s-1, to effectively neutralize IL-33 following rapid release from damaged tissue. An innovative antibody generation campaign identified tozorakimab, an antibody with a femtomolar affinity for IL-33red and a fast association rate (8.5 × 107 M-1 s-1), which was comparable to soluble ST2. Tozorakimab potently inhibited ST2-dependent inflammatory responses driven by IL-33 in primary human cells and in a murine model of lung epithelial injury. Additionally, tozorakimab prevented the oxidation of IL-33 and its activity via the RAGE/EGFR signalling pathway, thus increasing in vitro epithelial cell migration and repair. Tozorakimab is a novel therapeutic agent with a dual mechanism of action that blocks IL-33red and IL-33ox signalling, offering potential to reduce inflammation and epithelial dysfunction in human disease.
Collapse
Affiliation(s)
| | - D Gareth Rees
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Ian Christopher Scott
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Sara Carmen
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK
| | | | | | - Kirsty F Houslay
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Teodor Erngren
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Mark Penney
- Early Oncology DMPK, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Jayesh B Majithiya
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Laura Rapley
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dorothy A Sims
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Claire Hollins
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Elizabeth C Hinchy
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | | | - Dominic J Corkill
- Bioscience In Vivo, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Richard D May
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | | | - Tomas Mustelin
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - David C Lowe
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK
| | | | - E Suzanne Cohen
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
42
|
DeMichele E, Sosnowski O, Buret AG, Allain T. Regulatory Functions of Hypoxia in Host-Parasite Interactions: A Focus on Enteric, Tissue, and Blood Protozoa. Microorganisms 2023; 11:1598. [PMID: 37375100 PMCID: PMC10303274 DOI: 10.3390/microorganisms11061598] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Body tissues are subjected to various oxygenic gradients and fluctuations and hence can become transiently hypoxic. Hypoxia-inducible factor (HIF) is the master transcriptional regulator of the cellular hypoxic response and is capable of modulating cellular metabolism, immune responses, epithelial barrier integrity, and local microbiota. Recent reports have characterized the hypoxic response to various infections. However, little is known about the role of HIF activation in the context of protozoan parasitic infections. Growing evidence suggests that tissue and blood protozoa can activate HIF and subsequent HIF target genes in the host, helping or hindering their pathogenicity. In the gut, enteric protozoa are adapted to steep longitudinal and radial oxygen gradients to complete their life cycle, yet the role of HIF during these protozoan infections remains unclear. This review focuses on the hypoxic response to protozoa and its role in the pathophysiology of parasitic infections. We also discuss how hypoxia modulates host immune responses in the context of protozoan infections.
Collapse
Affiliation(s)
- Emily DeMichele
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Olivia Sosnowski
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Thibault Allain
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
43
|
Brunetti G, Barile B, Nicchia GP, Onorati F, Luciani GB, Galeone A. The ST2/IL-33 Pathway in Adult and Paediatric Heart Disease and Transplantation. Biomedicines 2023; 11:1676. [PMID: 37371771 DOI: 10.3390/biomedicines11061676] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
ST2 is a member of interleukin 1 receptor family with soluble sST2 and transmembrane ST2L isoforms. The ligand of ST2 is IL-33, which determines the activation of numerous intracytoplasmic mediators following the binding with ST2L and IL-1RAcP, leading to nuclear signal and cardiovascular effect. Differently, sST2 is released in the blood and works as a decoy receptor, binding IL-33 and blocking IL-33/ST2L interaction. sST2 is mainly involved in maintaining homeostasis and/or alterations of different tissues, as counterbalance/activation of IL-33/ST2L axis is typically involved in the development of fibrosis, tissue damage, inflammation and remodeling. sST2 has been described in different clinical reports as a fundamental prognostic marker in patients with cardiovascular disease, as well as marker for the treatment monitoring of patients with heart failure; however, further studies are needed to better elucidate its role. In this review we reported the current knowledge about its role in coronary artery disease, heart failure, heart transplantation, heart valve disease, pulmonary arterial hypertension, and cardiovascular interventions.
Collapse
Affiliation(s)
- Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Barbara Barile
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Francesco Onorati
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Giovanni Battista Luciani
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Antonella Galeone
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| |
Collapse
|
44
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [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: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
45
|
Thanikachalam PV, Ramamurthy S, Mallapu P, Varma SR, Narayanan J, Abourehab MA, Kesharwani P. Modulation of IL-33/ST2 signaling as a potential new therapeutic target for cardiovascular diseases. Cytokine Growth Factor Rev 2023; 71-72:94-104. [PMID: 37422366 DOI: 10.1016/j.cytogfr.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023]
Abstract
IL-33 belongs to the IL-1 family of cytokines, which function as inducers of Th2 cytokine production by binding with ST2L and IL-1RAcP. This, in turn, activates various signaling pathways, including the mitogen-activated protein kinase (MAPK), the inhibitor of Kappa-B kinase (IKK) pathway, and the phospholipase D-sphingosine kinase pathway. IL-33 has demonstrated protective effects against various cardiovascular diseases (CVDs) by inducing Th2 cytokines and promoting alternative activating M2 polarization. However, the soluble decoy form of ST2 (sST2) mitigates the biological effects of IL-33, exacerbating CVDs. Furthermore, IL-33 also plays a significant role in the development of asthma, arthritis, atopic dermatitis, and anaphylaxis through the activation of Th2 cells and mast cells. In this review, we aim to demonstrate the protective role of IL-33 against CVDs from 2005 to the present and explore the potential of serum soluble ST2 (sST2) as a diagnostic biomarker for CVDs. Therefore, IL-33 holds promise as a potential therapeutic target for the treatment of CVDs.
Collapse
Affiliation(s)
- Punniyakoti Veeraveedu Thanikachalam
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India.
| | - Srinivasan Ramamurthy
- College of Pharmacy and Health Sciences, University of Science and Technology of Fujairah, Fujairah, United Arab Emirates
| | - Poojitha Mallapu
- Department of Pharmacology, GRT Institute of Pharmaceutical Education and Research, Tiruttani, India
| | - Sudhir Rama Varma
- Department of Clinical Sciences, Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Jayaraj Narayanan
- Department of Basic Sciences, Center of Medical and Bio-allied Health Sciences Research, Ajman university, Ajman, United Arab Emirates
| | - Mohammed As Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| |
Collapse
|
46
|
Wang X, Hallen NR, Lee M, Samuchiwal S, Ye Q, Buchheit KM, Maxfield AZ, Roditi RE, Bergmark RW, Bhattacharyya N, Ryan T, Gakpo D, Raychaudhuri S, Dwyer D, Laidlaw TM, Boyce JA, Gutierrez-Arcelus M, Barrett NA. Type 2 inflammation drives an airway basal stem cell program through insulin receptor substrate signaling. J Allergy Clin Immunol 2023; 151:1536-1549. [PMID: 36804595 PMCID: PMC10784786 DOI: 10.1016/j.jaci.2023.01.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyposis (CRSwNP) is a type 2 (T2) inflammatory disease associated with an increased number of airway basal cells (BCs). Recent studies have identified transcriptionally distinct BCs, but the molecular pathways that support or inhibit human BC proliferation and differentiation are largely unknown. OBJECTIVE We sought to determine the role of T2 cytokines in regulating airway BCs. METHODS Single-cell and bulk RNA sequencing of sinus and lung airway epithelial cells was analyzed. Human sinus BCs were stimulated with IL-4 and IL-13 in the presence and absence of inhibitors of IL-4R signaling. Confocal analysis of human sinus tissue and murine airway was performed. Murine BC subsets were sorted for RNA sequencing and functional assays. Fate labeling was performed in a murine model of tracheal injury and regeneration. RESULTS Two subsets of BCs were found in human and murine respiratory mucosa distinguished by the expression of basal cell adhesion molecule (BCAM). BCAM expression identifies airway stem cells among P63+KRT5+NGFR+ BCs. In the sinonasal mucosa, BCAMhi BCs expressing TSLP, IL33, CCL26, and the canonical BC transcription factor TP63 are increased in patients with CRSwNP. In cultured BCs, IL-4/IL-13 increases the expression of BCAM and TP63 through an insulin receptor substrate-dependent signaling pathway that is increased in CRSwNP. CONCLUSIONS These findings establish BCAM as a marker of airway stem cells among the BC pool and demonstrate that airway epithelial remodeling in T2 inflammation extends beyond goblet cell metaplasia to the support of a BC stem state poised to perpetuate inflammation.
Collapse
Affiliation(s)
- Xin Wang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Nils R Hallen
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Minkyu Lee
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Sachin Samuchiwal
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Qihua Ye
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Kathleen M Buchheit
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Alice Z Maxfield
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Rachel E Roditi
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Regan W Bergmark
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Neil Bhattacharyya
- Department of Otolaryngology, Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Boston, Mass
| | - Tessa Ryan
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Deb Gakpo
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Boston, Mass; Divisions of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Mass; Versus Arthritis Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Dan Dwyer
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Tanya M Laidlaw
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Joshua A Boyce
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Maria Gutierrez-Arcelus
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Mass; Division of Immunology, Boston Children's Hospital, Boston, Mass
| | - Nora A Barrett
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| |
Collapse
|
47
|
Cook DP, Thomas CM, Wu AY, Rusznak M, Zhang J, Zhou W, Cephus JY, Gibson-Corley KN, Polosukhin VV, Norlander AE, Newcomb DC, Stoltz DA, Peebles RS. Cystic Fibrosis Reprograms Airway Epithelial IL-33 Release and Licenses IL-33-Dependent Inflammation. Am J Respir Crit Care Med 2023; 207:1486-1497. [PMID: 36952660 PMCID: PMC10263140 DOI: 10.1164/rccm.202211-2096oc] [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: 11/14/2022] [Accepted: 03/23/2023] [Indexed: 03/25/2023] Open
Abstract
Rationale: Type 2 inflammation has been described in people with cystic fibrosis (CF). Whether loss of CFTR (cystic fibrosis transmembrane conductance regulator) function contributes directly to a type 2 inflammatory response has not been fully defined. Objectives: The potent alarmin IL-33 has emerged as a critical regulator of type 2 inflammation. We tested the hypothesis that CFTR deficiency increases IL-33 expression and/or release and deletion of IL-33 reduces allergen-induced inflammation in the CF lung. Methods: Human airway epithelial cells (AECs) grown from non-CF and CF cell lines and Cftr+/+ and Cftr-/- mice were used in this study. Pulmonary inflammation in Cftr+/+ and Cftr-/- mice with and without IL-33 or ST2 (IL-1 receptor-like 1) germline deletion was determined by histological analysis, BAL, and cytokine analysis. Measurements and Main Results: After allergen challenge, both CF human AECs and Cftr-/- mice had increased IL-33 expression compared with control AECs and Cftr+/+ mice, respectively. DUOX1 (dual oxidase 1) expression was increased in CF human AECs and Cftr-/- mouse lungs compared with control AECs and lungs from Cftr+/+ mice and was necessary for the increased IL-33 release in Cftr-/- mice compared with Cftr+/+ mice. IL-33 stimulation of Cftr-/- CD4+ T cells resulted in increased type 2 cytokine production compared with Cftr+/+ CD4+ T cells. Deletion of IL-33 or ST2 decreased both type 2 inflammation and neutrophil recruitment in Cftr-/- mice compared with Cftr+/+ mice. Conclusions: Absence of CFTR reprograms airway epithelial IL-33 release and licenses IL-33-dependent inflammation. Modulation of the IL-33/ST2 axis represents a novel therapeutic target in CF type 2-high and neutrophilic inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Katherine N. Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Dawn C. Newcomb
- Department of Internal Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David A. Stoltz
- Department of Internal Medicine and
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; and
| | - R. Stokes Peebles
- Department of Internal Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, Tennessee
| |
Collapse
|
48
|
Kang MH, Hong J, Lee J, Cha MS, Lee S, Kim HY, Ha SJ, Lim YT, Bae YS. Discovery of highly immunogenic spleen-resident FCGR3 +CD103 + cDC1s differentiated by IL-33-primed ST2 + basophils. Cell Mol Immunol 2023:10.1038/s41423-023-01035-8. [PMID: 37246159 DOI: 10.1038/s41423-023-01035-8] [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: 11/08/2022] [Accepted: 04/25/2023] [Indexed: 05/30/2023] Open
Abstract
Recombinant interleukin-33 (IL-33) inhibits tumor growth, but the detailed immunological mechanism is still unknown. IL-33-mediated tumor suppression did not occur in Batf3-/- mice, indicating that conventional type 1 dendritic cells (cDC1s) play a key role in IL-33-mediated antitumor immunity. A population of CD103+ cDC1s, which were barely detectable in the spleens of normal mice, increased significantly in the spleens of IL-33-treated mice. The newly emerged splenic CD103+ cDC1s were distinct from conventional splenic cDC1s based on their spleen residency, robust effector T-cell priming ability, and surface expression of FCGR3. DCs and DC precursors did not express Suppressor of Tumorigenicity 2 (ST2). However, recombinant IL-33 induced spleen-resident FCGR3+CD103+ cDC1s, which were found to be differentiated from DC precursors by bystander ST2+ immune cells. Through immune cell fractionation and depletion assays, we found that IL-33-primed ST2+ basophils play a crucial role in the development of FCGR3+CD103+ cDC1s by secreting IL-33-driven extrinsic factors. Recombinant GM-CSF also induced the population of CD103+ cDC1s, but the population neither expressed FCGR3 nor induced any discernable antitumor immunity. The population of FCGR3+CD103+ cDC1s was also generated in vitro culture of Flt3L-mediated bone marrow-derived DCs (FL-BMDCs) when IL-33 was added in a pre-DC stage of culture. FL-BMDCs generated in the presence of IL-33 (FL-33-DCs) offered more potent tumor immunotherapy than control Flt3L-BMDCs (FL-DCs). Human monocyte-derived DCs were also more immunogenic when exposed to IL-33-induced factors. Our findings suggest that recombinant IL-33 or an IL-33-mediated DC vaccine could be an attractive protocol for better tumor immunotherapy.
Collapse
Affiliation(s)
- Myeong-Ho Kang
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - JungHyub Hong
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - Jinjoo Lee
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - Min-Suk Cha
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - Hye-Young Kim
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Sang-Jun Ha
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yong Taik Lim
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
- Department of Nano Engineering and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea.
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea.
| |
Collapse
|
49
|
Pisani LF, Teani I, Vecchi M, Pastorelli L. Interleukin-33: Friend or Foe in Gastrointestinal Tract Cancers? Cells 2023; 12:1481. [PMID: 37296602 PMCID: PMC10252908 DOI: 10.3390/cells12111481] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Accumulating evidence suggests that Interleukin-33 (IL-33), a member of the IL-1 family, has crucial roles in tissue homeostasis and repair, type 2 immunity, inflammation, and viral infection. IL-33 is a novel contributing factor in tumorigenesis and plays a critical role in regulating angiogenesis and cancer progression in a variety of human cancers. The partially unraveled role of IL-33/ST2 signaling in gastrointestinal tract cancers is being investigated through the analysis of patients' samples and by studies in murine and rat models. In this review, we discuss the basic biology and mechanisms of release of the IL-33 protein and its involvement in gastrointestinal cancer onset and progression.
Collapse
Affiliation(s)
- Laura Francesca Pisani
- Gastroenterology and Endoscopy Unit, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy
| | - Isabella Teani
- Department of Medicine, University of Verona, 37129 Verona, Italy;
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Luca Pastorelli
- Department of Health Sciences, University of Milan, 20122 Milan, Italy
- Gastroenterology and Liver Unit, ASST Santi Paolo e Carlo, 20142 Milan, Italy
| |
Collapse
|
50
|
Jiang Y, Donati V, Peters GJ, Giovannetti E, Deng DM. Fungal mycobiome-mediated immune response: a non-negligible promoter in pancreatic oncogenesis and chemoresistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:284-290. [PMID: 37457135 PMCID: PMC10344719 DOI: 10.20517/cdr.2023.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 07/18/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers in humans due to late diagnosis and poor response to treatments. The tumor microenvironment (TME) of PDAC is characterized by a distinctive, suppressive immune profile, which inhibits the protective functions of anti-tumor immunity and thereby contributes to PDAC progression. Recently, the study of Alam et al. discovered for the first time that the intratumoral fungal mycobiome could contribute to the recruitment and activation of type 2 immune cells in the TME of PDAC via enhancing the secretion of a chemoattractant, interleukin (IL-) 33. In this article, we reviewed the important findings of this study. Together with our findings, we synthetically discussed the role of the fungal mycobiome in orchestrating the immune response and thereby modulating tumor progression.
Collapse
Affiliation(s)
- Yaling Jiang
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam 1081 LA, Netherlands
| | - Valentina Donati
- Department of Medical Oncology, Amsterdam University Medical Centers, location VUMC, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, Netherlands
- Unit of Pathological Anatomy 2, Azienda Ospedaliero-Universitaria Pisana, Pisa 56100, Italy
| | - Godefridus J. Peters
- Department of Medical Oncology, Amsterdam University Medical Centers, location VUMC, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, Netherlands
- Department of Biochemistry, Medical University of Gdansk, Gdansk 80-210, Poland
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Centers, location VUMC, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, Netherlands
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Pisa 56017, Italy
| | - Dong Mei Deng
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam 1081 LA, Netherlands
| |
Collapse
|