1
|
Gu S, Wang R, Zhang W, Wen C, Chen C, Liu S, Lei Q, Zhang P, Zeng S. The production, function, and clinical applications of IL-33 in type 2 inflammation-related respiratory diseases. Front Immunol 2024; 15:1436437. [PMID: 39301028 PMCID: PMC11410612 DOI: 10.3389/fimmu.2024.1436437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
Epithelial-derived IL-33 (Interleukin-33), as a member of alarm signals, is a chemical substance produced under harmful stimuli that can promote innate immunity and activate adaptive immune responses. Type 2 inflammation refers to inflammation primarily mediated by Type 2 helper T cells (Th2), Type 2 innate lymphoid cells (ILC2), and related cytokines. Type 2 inflammation manifests in various forms in the lungs, with diseases such as asthma and chronic obstructive pulmonary disease chronic obstructive pulmonary disease (COPD) closely associated with Type 2 inflammation. Recent research suggests that IL-33 has a promoting effect on Type 2 inflammation in the lungs and can be regarded as an alarm signal for Type 2 inflammation. This article provides an overview of the mechanisms and related targets of IL-33 in the development of lung diseases caused by Type 2 inflammation, and summarizes the associated treatment methods. Analyzing lung diseases from a new perspective through the alarm of Type 2 inflammation helps to gain a deeper understanding of the pathogenesis of these related lung diseases. This, in turn, facilitates a better understanding of the latest treatment methods and potential therapeutic targets for diseases, with the expectation that targeting lL-33 can propose new strategies for disease prevention.
Collapse
Affiliation(s)
- Shiyao Gu
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruixuan Wang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wantian Zhang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Cen Wen
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunhua Chen
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Su Liu
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qian Lei
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Zhang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Si Zeng
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
2
|
Wang L, Cheng J, Huang J, Xiao T, Tang Z. The mechanism of IL-13 targeting IL-13Rα2 in regulating oral mucosal FBs through PI3K/AKT/mTOR. Oral Dis 2024; 30:3142-3154. [PMID: 37897109 DOI: 10.1111/odi.14760] [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: 06/22/2022] [Revised: 08/30/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023]
Abstract
OBJECTIVE The objective of this investigation was to examine the presence of interleukin (IL)-13 and its receptor IL-13Rα2 in the tissues of oral submucous fibrosis (OSF), investigate their biological functions, and explore the underlying mechanisms involved in the development of OSF. MATERIALS AND METHODS The expression of IL-13 and IL-13Rα2 in the oral mucosa of patients with OSF and normal individuals was determined through immunohistochemistry and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Primary fibroblasts (FBs) were extracted through enzymatic digestion and then cultured. Immunofluorescence was employed to identify the FB cultures and the location of IL-13Rα2. The effects of IL-13/IL-13Rα2/PI3K/AKT/mTOR on the migration, proliferation, and secretion of fiber-related proteins of FBs were explored via the wound healing assay, CCK-8 assay, EDU assay, and RT-qPCR. The impact of IL-13Rα2 silencing and PI3K/AKT inhibition on the effect of IL-13 on FBs was analyzed by RT-qPCR and Western blotting. RESULTS IL-13 and IL-13Rα2 were highly expressed in OSF. Primary FBs were successfully extracted and cultured. IL-13Rα2 was found to be localized in myofibroblasts. IL-13 promoted the proliferation, migration, and secretion of fibril-associated proteins in FBs. The proliferation, migration, and secretion of fibril-associated proteins of FBs were decreased following IL-13Rα2 silencing and inhibition of the PI3K/AKT/mTOR pathway. CONCLUSION IL-13 may promote the proliferation, migration, and secretion of fiber-related proteins of FBs through the PI3K/AKT/mTOR pathway by targeting IL-13Rα2.
Collapse
Affiliation(s)
- Liping Wang
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Academician Workstation for Oral-maxilofacial and Regenerative Medicine & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Jingyi Cheng
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Academician Workstation for Oral-maxilofacial and Regenerative Medicine & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Junhui Huang
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Academician Workstation for Oral-maxilofacial and Regenerative Medicine & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Ting Xiao
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Academician Workstation for Oral-maxilofacial and Regenerative Medicine & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Zhangui Tang
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Academician Workstation for Oral-maxilofacial and Regenerative Medicine & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
3
|
Joerns EK, Karp D, Zhang S, Sparks JA, Adams TN, Makris UE, Newton CA. High Interleukin-13 level is associated with disease stability in interstitial Lung disease. Heliyon 2024; 10:e32118. [PMID: 38882341 PMCID: PMC11176841 DOI: 10.1016/j.heliyon.2024.e32118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Purpose Cytokines can help predict prognosis in interstitial lung disease (ILD) and to differentiate between ILD subtypes. The objectives of our study were to evaluate association of baseline cytokine levels with time to ILD progression and to compare baseline cytokine levels between ILD subtypes. Methods We quantified 27 cytokines using a multiplex assay in peripheral blood samples from 77 patients. Cox proportional hazards regression analysis was performed to evaluate cytokine impact on the time to progression in the total cohort and within each ILD type. We evaluated for significant differences in cytokine levels between ILD types using ANOVA, Wilcoxon signed-rank test and Tukey method. Results Higher IL-13 level was associated with longer time to progression (hazard ratio 0.52 [0.33-0.81], p-value 0.004). FGF-β, GM-CSF, and IL-17 levels differed significantly between fibrotic and inflammatory ILD subgroups. Conclusion IL-13 may be a useful biomarker predicting ILD stability.
Collapse
Affiliation(s)
- Elena K Joerns
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Rheumatic Diseases, Dallas, TX, USA
- Mayo Clinic, Department of Internal Medicine, Division of Rheumatology, Rochester, MN, USA
| | - David Karp
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Rheumatic Diseases, Dallas, TX, USA
| | - Song Zhang
- University of Texas Southwestern Medical Center, Department of Population and Data Sciences, Division of Biostatistics, Dallas, TX, USA
| | - Jeffrey A Sparks
- Brigham and Women's Hospital, Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Harvard Medical School, Boston, MA, USA
| | - Traci N Adams
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Dallas, TX, USA
| | - Una E Makris
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Rheumatic Diseases, Dallas, TX, USA
- Dallas Veterans Affairs Medical Center, Dallas, TX, USA
| | - Chad A Newton
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Dallas, TX, USA
| |
Collapse
|
4
|
Gong X, Liang Y, Wang J, Pang Y, Wang F, Chen X, Zhang Q, Song C, Wang Y, Zhang C, Fang X, Chen X. Highly pathogenic PRRSV upregulates IL-13 production through nonstructural protein 9-mediated inhibition of N6-methyladenosine demethylase FTO. J Biol Chem 2024; 300:107199. [PMID: 38508309 PMCID: PMC11017062 DOI: 10.1016/j.jbc.2024.107199] [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: 11/09/2023] [Revised: 03/04/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV), a highly infectious virus, causes severe losses in the swine industry by regulating the inflammatory response, inducing tissue damage, suppressing the innate immune response, and promoting persistent infection in hosts. Interleukin-13 (IL-13) is a cytokine that plays a critical role in regulating immune responses and inflammation, particularly in immune-related disorders, certain types of cancer, and numerous bacterial and viral infections; however, the underlying mechanisms of IL-13 regulation during PRRSV infection are not well understood. In this study, we demonstrated that PRRSV infection elevates IL-13 levels in porcine alveolar macrophages. PRRSV enhances m6A-methylated RNA levels while reducing the expression of fat mass and obesity associated protein (FTO, an m6A demethylase), thereby augmenting IL-13 production. PRRSV nonstructural protein 9 (nsp9) was a key factor for this modulation. Furthermore, we found that the residues Asp567, Tyr586, Leu593, and Asp595 were essential for nsp9 to induce IL-13 production via attenuation of FTO expression. These insights delineate PRRSV nsp9's role in FTO-mediated IL-13 release, advancing our understanding of PRRSV's impact on host immune and inflammatory responses.
Collapse
Affiliation(s)
- Xingyu Gong
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China
| | - Yuan Liang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China
| | - Jingjing Wang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China
| | - Yipeng Pang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China
| | - Fang Wang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China
| | - Xiaohan Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China
| | - Qiaoya Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Chengchuang Song
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China; Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Yanhong Wang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China; Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Chunlei Zhang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China; Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Xingtang Fang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China; Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.
| | - Xi Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, China; Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.
| |
Collapse
|
5
|
Heggli I, Mengis T, Laux C, Opitz L, Herger N, Menghini D, Schuepbach R, Farshad-Amacker N, Brunner F, Fields A, Farshad M, Distler O, Dudli S. Low back pain patients with Modic type 1 changes exhibit distinct bacterial and non-bacterial subtypes. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100434. [PMID: 38322145 PMCID: PMC10844677 DOI: 10.1016/j.ocarto.2024.100434] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Objectives Modic type 1 changes (MC1) are vertebral endplate bone marrow (BM) lesions observed on magnetic resonance images in sub-populations of chronic low back pain (CLBP) patients. The etiopathogenesis remains unknown and treatments that modify the underlying pathomechanisms do not exist. We hypothesized that two biological MC1 subtypes exist: a bacterial and a non-bacterial. This would have important implications for developing treatments targeting the underlying pathomechanisms. Methods Intervertebral disc (IVD) samples adjacent to MC1 (n = 34) and control (n = 11) vertebrae were collected from patients undergoing spinal fusion. Cutibacterium acnes (C.acnes) genome copy numbers (GCNs) were quantified in IVD tissues with 16S qPCR, transcriptomic signatures and cytokine profiles were determined in MC1 and control BM by RNA sequencing and immunoassay. Finally, we assessed if C.acnes GCNs are associated with blood plasma cytokines. Results IVD tissues from control levels had <870 C.acnes GCNs/gram IVD. MC1-adjacent IVDs had either "low" (<870) or "high" (>870) C.acnes GCNs. MC1 patients with "high" C.acnes GCNs had upregulated innate immune cell signatures (neutrophil, macrophage/monocyte) and pro-inflammatory cytokines related to neutrophil and macrophage/monocyte function in the BM, consistent with a host defense against bacterium. MC1 patients with "low" C.acnes GCNs had increased adaptive immune cell signatures (T-and B-cell) in the BM and elevated IL-13 blood plasma levels. Conclusion Our study provides the first evidence for the existence of bacterial (C.acnes "high") and non-bacterial (C.acnes "low") subtypes in MC1 patients with CLBP. This supports the need for different treatment strategies.
Collapse
Affiliation(s)
- I. Heggli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - T. Mengis
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - C.J. Laux
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - L. Opitz
- Functional Genomics Center Zurich, University and ETH Zurich, Zurich, Zurich, Switzerland
| | - N. Herger
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - D. Menghini
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - R. Schuepbach
- Unit of Clinical and Applied Research, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - N.A. Farshad-Amacker
- Department of Radiology, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - F. Brunner
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - A.J. Fields
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - M. Farshad
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - O. Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - S. Dudli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Amati F, Bongiovanni G, Tonutti A, Motta F, Stainer A, Mangiameli G, Aliberti S, Selmi C, De Santis M. Treatable Traits in Systemic Sclerosis. Clin Rev Allergy Immunol 2023; 65:251-276. [PMID: 37603199 DOI: 10.1007/s12016-023-08969-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/22/2023]
Abstract
Systemic sclerosis (SSc) is a chronic systemic disease within the spectrum of connective tissue diseases, specifically characterized by vascular abnormalities and inflammatory and fibrotic involvement of the skin and internal organs resulting in high morbidity and mortality. The clinical phenotype of SSc is heterogeneous, and serum autoantibodies together with the extent of skin involvement have a predictive value in the risk stratification. Current recommendations include an organ-based management according to the predominant involvement with only limited individual factors included in the treatment algorithm. Similar to what has been proposed for other chronic diseases, we hypothesize that a "treatable trait" approach based on relevant phenotypes and endotypes could address the unmet needs in SSc stratification and treatment to maximize the outcomes. We provide herein a comprehensive review and a critical discussion of the literature regarding potential treatable traits in SSc, focusing on established and candidate biomarkers, with the purpose of setting the bases for a precision medicine-based approach. The discussion, structured based on the organ involvement, allows to conjugate the pathogenetic mechanisms of tissue injury with the proposed predictors, particularly autoantibodies and other serum biomarkers. Ultimately, we are convinced that precision medicine is the ideal guide to manage a complex condition such as SSc for which available treatments are largely unsatisfactory.
Collapse
Affiliation(s)
- Francesco Amati
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Gabriele Bongiovanni
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Antonio Tonutti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesca Motta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Anna Stainer
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Giuseppe Mangiameli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Thoracic Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | - Maria De Santis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
7
|
Steadman T, O'Reilly S. Elevated interleukin-11 in systemic sclerosis and role in disease pathogenesis. J Dermatol 2023; 50:1255-1261. [PMID: 37291792 DOI: 10.1111/1346-8138.16854] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/13/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023]
Abstract
Systemic sclerosis (SSc) is an autoimmune connective tissue disease in which there is elevated inflammation, aberrant cytokine expression, and subsequent fibrosis. Interleukin-11 (IL-11) is a recently described profibrotic cytokine that can mediate fibrosis in the heart, lungs, and skin and is upregulated by transforming Growth Factor-β (TGF-β1). The objective of this study was to quantify the serum levels of IL-11 in early diffuse SSc patients. Also, if IL-11 could regulate the alarmin IL-33 in dermal fibroblasts was quantified. Early diffuse SSc patient sera was isolated and IL-11 was quantified by specific commercial ELISA compared to healthy control (n = 17). Healthy dermal fibroblasts were cultured in vitro and then serum starved and incubated with or without recombinant IL-11. At specific early and late time points the supernatant was quantified for the alarmin IL-33 by specific ELISA. In early diffuse SSc patients it was demonstrated that they have elevated IL-11 in their sera. In a subgroup of SSc patients with interstitial lung disease (ILD) this elevation was particularly pronounced compared to those devoid of fibrotic lung disease. In vitro incubation of healthy dermal fibroblasts led to a significant induction of IL-33 cytokine release into the cell media. IL-11 is a profibrotic cytokine that is elevated in early diffuse SSc and is particularly elevated in those with ILD. This suggests that IL-11 could be a possible biomarker of ILD in SSc. It was also found that IL-11 led to release of the cytokine alarmin IL-33 in fibroblasts at earlier time points but not late time points, suggesting early stimulation elicits an inflammatory response in the local microenvironment but prolonged stimulation leads to fibrosis.
Collapse
|
8
|
Kazakov AS, Deryusheva EI, Rastrygina VA, Sokolov AS, Permyakova ME, Litus EA, Uversky VN, Permyakov EA, Permyakov SE. Interaction of S100A6 Protein with the Four-Helical Cytokines. Biomolecules 2023; 13:1345. [PMID: 37759746 PMCID: PMC10526228 DOI: 10.3390/biom13091345] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/19/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
S100 is a family of over 20 structurally homologous, but functionally diverse regulatory (calcium/zinc)-binding proteins of vertebrates. The involvement of S100 proteins in numerous vital (patho)physiological processes is mediated by their interaction with various (intra/extra)cellular protein partners, including cell surface receptors. Furthermore, recent studies have revealed the ability of specific S100 proteins to modulate cell signaling via direct interaction with cytokines. Previously, we revealed the binding of ca. 71% of the four-helical cytokines via the S100P protein, due to the presence in its molecule of a cytokine-binding site overlapping with the binding site for the S100P receptor. Here, we show that another S100 protein, S100A6 (that has a pairwise sequence identity with S100P of 35%), specifically binds numerous four-helical cytokines. We have studied the affinity of the recombinant forms of 35 human four-helical cytokines from all structural families of this fold to Ca2+-loaded recombinant human S100A6, using surface plasmon resonance spectroscopy. S100A6 recognizes 26 of the cytokines from all families of this fold, with equilibrium dissociation constants from 0.3 nM to 12 µM. Overall, S100A6 interacts with ca. 73% of the four-helical cytokines studied to date, with a selectivity equivalent to that for the S100P protein, with the differences limited to the binding of interleukin-2 and oncostatin M. The molecular docking study evidences the presence in the S100A6 molecule of a cytokine-binding site, analogous to that found in S100P. The findings argue the presence in some of the promiscuous members of the S100 family of a site specific to a wide range of four-helical cytokines. This unique feature of the S100 proteins potentially allows them to modulate the activity of the numerous four-helical cytokines in the disorders accompanied by an excessive release of the cytokines.
Collapse
Affiliation(s)
- Alexey S. Kazakov
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia; (A.S.K.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Evgenia I. Deryusheva
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia; (A.S.K.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Victoria A. Rastrygina
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia; (A.S.K.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Andrey S. Sokolov
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia; (A.S.K.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Maria E. Permyakova
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia; (A.S.K.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Ekaterina A. Litus
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia; (A.S.K.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Vladimir N. Uversky
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia; (A.S.K.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
- Department of Molecular, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Eugene A. Permyakov
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia; (A.S.K.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Sergei E. Permyakov
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia; (A.S.K.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| |
Collapse
|
9
|
Xie L, Long X, Mo M, Jiang J, Zhang Q, Long M, Li M. Bone marrow mesenchymal stem cell-derived exosomes alleviate skin fibrosis in systemic sclerosis by inhibiting the IL-33/ST2 axis via the delivery of microRNA-214. Mol Immunol 2023; 157:146-157. [PMID: 37028129 DOI: 10.1016/j.molimm.2023.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/24/2023] [Accepted: 03/23/2023] [Indexed: 04/09/2023]
Abstract
Interleukin (IL)- 33 is a tissue-derive proinflammatory cytokine that promotes fibrosis in systemic sclerosis (SSc). microRNA (miR)- 214 expression has been elaborated to be downregulated in SSc patients and exert anti-fibrotic and anti-inflammatory effects. This study elucidates the role of bone marrow mesenchymal stem cell-derived exosome (BMSC-Exos)-delivered miR-214 in SSc and the relationship between this miR and IL-33/ST2 axis. SSc clinical samples were obtained to evaluate levels of miR-214, IL-33, and ST2. Primary fibroblasts and BMSC-Exos were extracted, followed by the co-culture of PKH6-labeled BMSC-Exos and fibroblasts. Subsequently, Exos extracted from miR-214 inhibitor-transfected BMSCs were co-cultured with TGF-β1-stimulated fibroblasts, after which the expression of fibrotic markers, miR-214, IL-33, and ST2, as well as fibroblast proliferation and migration, was determined. A skin fibrosis mouse model was induced with bleomycin (BLM) and treated with BMSC-Exos. Collagen fiber accumulation, collagen content, α-SMA expression, and IL-33 and ST2 levels were examined in BLM-treated or IL-33-knockout mice. IL-33 and ST2 were upregulated and miR-214 was downregulated in SSc patients. Mechanistically, miR-214 targeted IL-33 and blocked the IL-33/ST2 axis. BMSC-Exos delivering miR-214 inhibitor augmented proliferation, migration, and fibrotic gene expression in TGF-β1-stimulated fibroblasts. Similarly, IL-33 induced migration, proliferation, and fibrotic gene expression in fibroblasts via ST2. In BLM-treated mice, IL-33 knockout suppressed skin fibrosis, and BMSC-Exos delivered miR-214 to suppress the IL-33/ST2 axis, thus mitigating skin fibrosis. Conclusively, BMSC-Exos alleviate skin fibrosis through the blockade of the IL-33/ST2 axis by delivering miR-214.
Collapse
Affiliation(s)
- Lihu Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Xiaoping Long
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Meili Mo
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Jinmei Jiang
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Qingxiu Zhang
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Mei Long
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Mei Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
10
|
Serum-soluble ST2 and systemic sclerosis arthropathy. Clin Rheumatol 2023; 42:871-877. [PMID: 36098853 DOI: 10.1007/s10067-022-06367-w] [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: 06/24/2022] [Revised: 08/30/2022] [Accepted: 09/03/2022] [Indexed: 11/03/2022]
Abstract
Interleukin (IL)33 and its receptor ST2 have been involved in the pathogenesis of several conditions, including arthritis. The aim of the study was to evaluate the association between IL33 or soluble ST2 (sST2) serum levels and systemic sclerosis (SSc) articular involvement. IL33 and sST2 serum levels were measured in 64 SSc patients and 24 HC matched for sex and age. Articular involvement assessed by using Disease Activity Score 28 based on erythrocyte sedimentation rate (DAS28-ESR), presence of tendon friction rubs (TFRs) and finger-to-palm (FTP) distance. sST2 serum levels were significantly higher in SSc patients with DAS28-ESR > 3.2 than in SSc patients with DAS28-ESR⩽3.2 [9726.1 (IQR 7746.5 - 14,953.5) pg/mL vs 7611.7 (IQR 5162.6 -11,036.7) pg/mL; p < 0.05]. sST2 serum levels were significantly higher in SSc patients with TFRs compared to SSc patients without TFRs [9726.1 (IQR 7746.5 - 14,953.5) pg/mL vs 7426.4 (IQR 5145.9 - 10,593.5) pg/mL; p < 0.01] and in SSc patients with FTP ≥ 1 cm compared to SSc patients with FTP < 1 cm [9683.7 (IQR 8067.2 - 16,387.6) pg/mL vs 7679.1 (IQR 5246.1 - 11,472.2) pg/mL; p < 0.05]. No significant association was observed between IL33 and DAS28-ESR, TFRs and FTP. A slightly positive linear correlation was found between sST2 and Disease Activity Index (r = 0.294, p < 0.05) and Disease Severity Scale (r = 0.265, p < 0.05). sST2 serum levels were positively correlated with DAS28-ESR (r = 0.371, p < 0.01). Elevated sST2 serum levels were associated with higher articular disease activity, TFRs and hand dysfunction, suggesting that sST2 might have a role in the pathogenesis of SSc articular involvement. Key Points • In SSc patients elevated serum levels of sST2 were associated with higher articular disease activity • High serum levels of sST2 were reported in SSc patients with TFRs and hand dysfunction • sST2 might have a role in the pathogenesis of SSc articular involvement.
Collapse
|
11
|
The Molecular Mechanisms of Systemic Sclerosis-Associated Lung Fibrosis. Int J Mol Sci 2023; 24:ijms24032963. [PMID: 36769282 PMCID: PMC9917655 DOI: 10.3390/ijms24032963] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Systemic sclerosis (SSc), also known as scleroderma, is an autoimmune disorder that affects the connective tissues and has the highest mortality rate among the rheumatic diseases. One of the hallmarks of SSc is fibrosis, which may develop systemically, affecting the skin and virtually any visceral organ in the body. Fibrosis of the lungs leads to interstitial lung disease (ILD), which is currently the leading cause of death in SSc. The identification of effective treatments to stop or reverse lung fibrosis has been the main challenge in reducing SSc mortality and improving patient outcomes and quality of life. Thus, understanding the molecular mechanisms, altered pathways, and their potential interactions in SSc lung fibrosis is key to developing potential therapies. In this review, we discuss the diverse molecular mechanisms involved in SSc-related lung fibrosis to provide insights into the altered homeostasis state inherent to this fatal disease complication.
Collapse
|
12
|
The association of IL-33 and systemic sclerosis: a systematic review and meta-analysis. Immunol Res 2023; 71:60-69. [PMID: 36306005 DOI: 10.1007/s12026-022-09329-1] [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: 08/16/2022] [Accepted: 10/12/2022] [Indexed: 01/20/2023]
Abstract
A meta-analysis of the association between IL-33 and these diseases is lacking, and we aimed to perform a meta-analysis of the association between IL-33 and systemic sclerosis (SSc). We searched relevant papers through PubMed (via Medline), Embase (via Ovid), and the Cochrane Library through May 18th, 2022. Odds ratios (ORs) and weighted mean differences (WMDs) were estimated using a random effect model. A total of 8 papers were included in our meta-analysis. The pooled results showed that SSc patients had significantly higher serum IL-33 levels than healthy controls (HCs) (SMD = 0.64; 95% CI = 0.34, 0.93; P < 0.001); early SSc patients had significantly higher serum IL-33 levels than late SSc patients (SMD = 1.04; 95% CI = 0.28, 1.80; P = 0.007). However, no significant difference was observed between limited cutaneous SSc and diffuse cutaneous SSc (SMD = - 0.35; 95% CI = - 0.76, 0.06; P = 0.094). Our meta-analysis provided important evidence for the use of IL-33 in therapeutic strategies for fibrotic disorders.
Collapse
|