1
|
Takahashi M, Mizuno-Kamiya M, Rahman S, Tsuruta H, Ikeno K, Kawaki H, Nakamura G, Muramatsu Y, Nikaido T, Fujita H, Kondoh N. Productions of Th2 cytokines, IL-4 and IL-10, were enhanced via the function of IL-2 from anti-CD3 antibody-stimulated mouse spleen cells treated with caffeic acid phenethyl ester. J Oral Biosci 2024:S1349-0079(24)00196-8. [PMID: 39241928 DOI: 10.1016/j.job.2024.09.001] [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: 05/23/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
OBJECTIVES Interleukin (IL)-2 production by mouse spleen cells stimulated with an anti-CD3 antibody is significantly enhanced by caffeic acid phenethyl ester (CAPE), a major constituent of Chinese propolis (CP). In this study, we evaluated the functional significance of IL-2 in CAPE-treated activated spleen cells. METHODS Mouse spleen cells were stimulated with an anti-CD3 monoclonal antibody in the presence of CAPE. Cytokine production was examined using an enzyme-linked immunosorbent assay (ELISA). Messenger RNA level expression was examined via reverse transcription quantitative polymerase chain reaction (RT-PCR). IL-2 function was assessed using IL-2 and a neutralizing antibody. Spleen cell subsets were identified and characterized using flow cytometry. RESULTS CAPE treatment of anti-CD3 antibody-stimulated spleen cells reduced IFN-γ production, then enhanced IL-2 production, followed by enhancement of IL-4 and IL-10 production. The Th2 cytokine production enhancing effects of CAPE were completely abolished by addition of an anti-IL-2 neutralizing antibody. In the absence of CAPE, exogenously added IL-2 could enhance IL-4 production to a lesser degree, but did not stimulate IL-10 production, in stimulated spleen cells. Interestingly, CAPE significantly reduced the proportions of CD4+ and CD8+ cells, and increased those of CD4-CD8- cells among anti-CD3 stimulated spleen cells, in the presence or absence of anti-IL-2 neutralizing antibody treatment. CONCLUSIONS CAPE reduced IFN-γ production, then enhanced IL-4 and IL-10 production via the activity of specifically elevated IL-2 in stimulated spleen cells. CAPE exerted these effects in a CD4- CD8- cell specific manner.
Collapse
Affiliation(s)
- Moe Takahashi
- Department of Oral and Maxillofacial Surgery, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Japan.
| | - Masako Mizuno-Kamiya
- Chemistry Laboratory, Department of Business Administration, Asahi University School of Business Administration, Mizuho, Gifu 501-0296, Japan.
| | - Shifa Rahman
- Department of Operative Dentistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Gifu 500-8471, Japan.
| | - Hanemi Tsuruta
- Department of Operative Dentistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Gifu 500-8471, Japan.
| | - Kumiko Ikeno
- AKITAYAHONTEN CO., LTD.R&D, Gifu 500-8471, Japan.
| | - Harumi Kawaki
- Department of Oral Biochemistry, Asahi University School of Dentistry Mizuho, Gifu 501-0296, Japan.
| | | | - Yasunori Muramatsu
- Department of Oral and Maxillofacial Surgery, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Japan.
| | - Toru Nikaido
- Department of Operative Dentistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Gifu 500-8471, Japan.
| | - Hisakazu Fujita
- Department of Scientific and Linguistic Fundamentals for Nursing, Osaka Metropolitan University Graduate School of Nursing, Osaka 558-8585, Japan.
| | - Nobuo Kondoh
- Chemistry Laboratory, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Gifu 500-8471, Japan.
| |
Collapse
|
2
|
Tomasovic LM, Liu K, VanDyke D, Fabilane CS, Spangler JB. Molecular Engineering of Interleukin-2 for Enhanced Therapeutic Activity in Autoimmune Diseases. BioDrugs 2024; 38:227-248. [PMID: 37999893 PMCID: PMC10947368 DOI: 10.1007/s40259-023-00635-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
The interleukin-2 (IL-2) cytokine plays a crucial role in regulating immune responses and maintaining immune homeostasis. Its immunosuppressive effects have been harnessed therapeutically via administration of low cytokine doses. Low-dose IL-2 has shown promise in the treatment of various autoimmune and inflammatory diseases; however, the clinical use of IL-2 is complicated by its toxicity, its pleiotropic effects on both immunostimulatory and immunosuppressive cell subsets, and its short serum half-life, which collectively limit the therapeutic window. As a result, there remains a considerable need for IL-2-based autoimmune disease therapies that can selectively target regulatory T cells with minimal off-target binding to immune effector cells in order to prevent cytokine-mediated toxicities and optimize therapeutic efficacy. In this review, we discuss exciting advances in IL-2 engineering that are empowering the development of novel therapies to treat autoimmune conditions. We describe the structural mechanisms of IL-2 signaling, explore current applications of IL-2-based compounds as immunoregulatory interventions, and detail the progress and challenges associated with clinical adoption of IL-2 therapies. In particular, we focus on protein engineering approaches that have been employed to optimize the regulatory T-cell bias of IL-2, including structure-guided or computational design of cytokine mutants, conjugation to polyethylene glycol, and the development of IL-2 fusion proteins. We also consider future research directions for enhancing the translational potential of engineered IL-2-based therapies. Overall, this review highlights the immense potential to leverage the immunoregulatory properties of IL-2 for targeted treatment of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Luke M Tomasovic
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathy Liu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Derek VanDyke
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Charina S Fabilane
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD, USA
| | - Jamie B Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Sidney Kimmel Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
3
|
Julien S, Challier I, Malleter M, Jouen F, Drouot L, Boyer O. Immune-Mediated Necrotizing Myopathy (IMNM): A Story of Antibodies. Antibodies (Basel) 2024; 13:12. [PMID: 38390873 PMCID: PMC10885118 DOI: 10.3390/antib13010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/10/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Immune-mediated necrotizing myopathy (IMNM) is a rare and severe disease that corresponds to a specific entity of idiopathic inflammatory myopathy. Patients with IMNM suffer from proximal muscle weakness, and present high levels of creatine kinase and necrotic myofibers. Anti-Signal Recognition Particle (SRP) and anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase autoantibodies (HMGCR) have recently been identified in two thirds of patients with IMNM and are used as a hallmark of the disease. In this review, we provide a detailed description of these antibodies and the tests used to detect them in the serum of patients. Based on in vitro studies and mouse models of IMNM, we discuss the role of autoantibodies in the pathogenesis of the disease. Finally, in the light of the latest knowledge, we conclude with a review of recent therapeutic approaches in IMNM.
Collapse
Affiliation(s)
- Sarah Julien
- INSERM U1234, PAn'THER FOCIS Center of Excellence, Université de Rouen, F-76000 Rouen, France
| | - Inès Challier
- Department of Pharmacy, CHU Rouen, F-76000 Rouen, France
| | - Marine Malleter
- INSERM U1234, PAn'THER FOCIS Center of Excellence, Université de Rouen, F-76000 Rouen, France
| | - Fabienne Jouen
- INSERM U1234, PAn'THER FOCIS Center of Excellence, Université de Rouen, F-76000 Rouen, France
- Department of Immunology and Biotherapy, CHU Rouen, F-76000 Rouen, France
| | - Laurent Drouot
- INSERM U1234, PAn'THER FOCIS Center of Excellence, Université de Rouen, F-76000 Rouen, France
| | - Olivier Boyer
- INSERM U1234, PAn'THER FOCIS Center of Excellence, Université de Rouen, F-76000 Rouen, France
- Department of Immunology and Biotherapy, CHU Rouen, F-76000 Rouen, France
| |
Collapse
|
4
|
Su Q, Wang X, Li Y, Zhang J, Bai C, Wang X, Yang L, Zhang J, Zhang SX. Efficacy, Safety and the Lymphocyte Subset Changes of Low-Dose IL-2 in Patients with Autoimmune Rheumatic Diseases: A Systematic Review and Meta-Analysis. Rheumatol Ther 2024; 11:79-96. [PMID: 37980696 PMCID: PMC10796881 DOI: 10.1007/s40744-023-00620-7] [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: 09/23/2023] [Accepted: 10/23/2023] [Indexed: 11/21/2023] Open
Abstract
INTRODUCTION Current therapies for autoimmune rheumatic diseases (ARDs) have limited efficacy in certain patients, highlighting the need for the development of novel treatment approaches. This meta-analysis aims to assess the efficacy and safety of low-dose interleukin-2 (LD-IL-2) and evaluate the alterations in lymphocyte subsets in various rheumatic diseases following administration of different dosages of LD-IL-2. METHODS A comprehensive search was conducted in PubMed, Web of Science, the Cochrane Library, Embase databases and CNKI to identify relevant studies. A total of 31 trials were included in this meta-analysis. The review protocols were registered on PROSPERO (CRD42022318916), and the study followed the PRISMA guidelines. RESULTS Following LD-IL-2 treatment, patients with ARDs exhibited a significant increase in the number of Th17 cells and Tregs compared to their pre-treatment levels [standardized mean difference (SMD) = 0.50, 95% confidence interval (CI) (0.33, 0.67), P < 0.001; SMD = 1.13, 95% CI (0.97, 1.29), P < 0.001]. Moreover, the Th17/Tregs ratio showed a significant decrease [SMD = - 0.54, 95% CI (- 0.64, - 0.45), P < 0.001]. In patients with rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), LD-IL-2 injection led to a significant increase in Treg numbers, and the Th17/Tregs ratio and disease activity scores, including Disease Activity Score-28 joints (DAS28), Systemic Lupus Erythematosus Disease Activity Index (SELENA-SLEDAI) and Cutaneous Dermatomyositis Disease Area and Severity Index (CDASI), were all significantly reduced. No serious adverse events were reported in any of the included studies. Additionally, 54.8% of patients with lupus nephritis achieved distinct clinical remission following LD-IL-2 treatment. Injection site reactions and fever were the most common side effects of LD-IL-2, occurring in 33.1% and 14.4% of patients, respectively. CONCLUSION LD-IL-2 treatment showed promise and was well tolerated in the management of ARDs, as it effectively promoted the proliferation and functional recovery of Tregs. TRIAL REGISTRATION Retrospectively registered (CRD42022318916, 21/04/2022).
Collapse
Affiliation(s)
- Qinyi Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, 382. Wuyi Road, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Xinmiao Wang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Yongzhi Li
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Jiexiang Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Cairui Bai
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Xuechun Wang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Liu Yang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Jingting Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, 382. Wuyi Road, Taiyuan, Shanxi, China.
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China.
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China.
| |
Collapse
|
5
|
Zhao J, Guo XJ, Shi L. Inflammatory biomarkers in polymyositis/dermatomyositis patients with interstitial lung disease: a retrospective study. Curr Med Res Opin 2024; 40:113-122. [PMID: 37938089 DOI: 10.1080/03007995.2023.2281501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/06/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Dermatomyositis (DM)/polymyositis (PM) is a systemic autoimmune disease characterized by proximal limb muscle with high morbidity and mortality and poor prognosis mediated by immune dysfunction; its etiology is unknown. DM/PM patients are at excessive risk of interstitial lung disease (ILD) and a higher risk of death. However, the role of circulating lymphocyte subsets, which play a pivotal role in occurrence and progression of DM/PM and ILD, respectively, remains unclear in DM/PM patients with ILD. METHODS Demographic characteristics, general data, and peripheral lymphocyte levels measured by flow cytometry were collected and analyzed in 47 DM/PM patients with ILD, 65 patients without ILD, and 105 healthy controls (HCs). RESULTS The most important first symptom of DM/PM patients is rash. Compared with non-ILD patients, the levels of neutrophil/lymphocyte ratio (NLR), systemic inflammatory response index (SIRI) were significantly higher and the levels of C reactive protein (CRP) were significantly lower in patients with ILD. Compared with HCs, DM/PM patients, with or without ILD, had decreased absolute counts of T, CD4 + T, CD8 + T, natural killer (NK), helper T (Th) 1, Th2, Th17, and regulatory T (Treg)cells. The fewest Th1 and Treg cells and the the lowest CD8 + T and Th1 cells percentages were seen in peripheral blood of patients with ILD. Longer duration, decreased lymphocyte/monocyte ratio (LMR)levels and CD8 + T and Th1 cells proportions, and fewer circulating Treg cells were independent risk factors for DM/PM with ILD. CONCLUSIONS The identification of peripheral blood T lymphocyte subsets, especially Treg cells, and blood count in DM/PM appears to be useful in the comprehensive assessment of clinical lung involvement.
Collapse
Affiliation(s)
- Jin Zhao
- Department of Hematology, Shanxi Province Cancer Hospital, Taiyuan, Shanxi, China
- Department of Hematology, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, Shanxi, China
- Department of Hematology, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiao-Jing Guo
- Department of Hematology, Shanxi Province Cancer Hospital, Taiyuan, Shanxi, China
- Department of Hematology, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, Shanxi, China
- Department of Hematology, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lei Shi
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
6
|
Chen S, Zhang P, Duan H, Wang J, Qiu Y, Cui Z, Yin Y, Wan D, Xie L. Gut microbiota in muscular atrophy development, progression, and treatment: New therapeutic targets and opportunities. Innovation (N Y) 2023; 4:100479. [PMID: 37539440 PMCID: PMC10394038 DOI: 10.1016/j.xinn.2023.100479] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023] Open
Abstract
Skeletal muscle atrophy is a debilitating condition that significantly affects quality of life and often lacks effective treatment options. Muscle atrophy can have various causes, including myogenic, neurogenic, and other factors. Recent investigation has underscored a compelling link between the gut microbiota and skeletal muscle. Discerning the potential differences in the gut microbiota associated with muscle atrophy-related diseases, understanding their influence on disease development, and recognizing their potential as intervention targets are of paramount importance. This review aims to provide a comprehensive overview of the role of the gut microbiota in muscle atrophy-related diseases. We summarize clinical and pre-clinical studies that investigate the potential for gut microbiota modulation to enhance muscle performance and promote disease recovery. Furthermore, we delve into the intricate interplay between the gut microbiota and muscle atrophy-related diseases, drawing from an array of studies. Emerging evidence suggests significant differences in gut microbiota composition in individuals with muscle atrophy-related diseases compared with healthy individuals. It is conceivable that these alterations in the microbiota contribute to the pathogenesis of these disorders through bacterium-related metabolites or inflammatory signals. Additionally, interventions targeting the gut microbiota have demonstrated promising results for mitigating disease progression in animal models, underscoring the therapeutic potential of modulating the gut microbiota in these conditions. By analyzing the available literature, this review sheds light on the involvement of the gut microbiota in muscle atrophy-related diseases. The findings contribute to our understanding of the underlying mechanisms and open avenues for development of novel therapeutic strategies targeting the gut-muscle axis.
Collapse
Affiliation(s)
- Shujie Chen
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Puxuan Zhang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Huimin Duan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Jie Wang
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Yuyueyang Qiu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- Department of Biology, Grinnell College, Grinnell, IA 501122, USA
| | - Zongbin Cui
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of the Chinese Academy of Sciences, Beijing 101408, China
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of the Chinese Academy of Sciences, Beijing 101408, China
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
- Department of Stomatology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde, Foshan), Foshan 528308, China
| |
Collapse
|
7
|
Yan H, Yan H, Liu L, Su R, Gao C, Li X, Wang C. Low-dose interleukin-2 treatment increases the proportion of regulatory T cells in patients with rheumatic diseases: A meta-analysis. Autoimmun Rev 2023; 22:103270. [PMID: 36627065 DOI: 10.1016/j.autrev.2023.103270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
BACKGROUND It is now accepted that immune tolerance disorders caused by inadequate Treg cell function or number are important factors in the development and progression of rheumatic diseases. There is increasing evidence that ld IL-2 treatment increases the proportion of Treg cells in patients' peripheral blood, but this conclusion is still controversial. Here, we performed a meta-analysis of reports documenting the proportion of Treg cells and the rate of adverse events in patients with rheumatic disease before and after the administration of ld IL-2 to better understand its effect and safety on Treg cells in the field of rheumatic diseases. METHODS We systematically searched PubMed, Embase, Scopus, Cochrane Library, and Web of science databases up to 15th November 2022 and identified studies that reported the proportion of peripheral blood Treg cells before and after ld IL-2 treatment in patients with rheumatic disease. Random-effects model was used to perform a meta-analysis of Treg cell proportions before and after ld IL-2 administration, and a meta-regression analysis was performed to explore heterogeneity. Inconsistency was evaluated using the I-squared index (I2), and publication bias was assessed by examining funnel plot asymmetry using the Egger tests. RESULTS Eighteen studies involving 1608 patients were included in the meta-analysis. The proportion of Treg cells in peripheral blood of these patients increased significantly after receiving ld IL-2 treatment [1.07 (95% CI 0.86,1.27), p < 0.001, I2 = 67.3%]. Next, Meta-regression was performed for 5 variables including publish year, disease type, trail type and dosage and duration of the medication. The results suggest that these variables do not lead to high heterogeneity. (p = 0.698, 0.267, 0.502, 0.843, 0.560, respectively). And finally, statistical analysis showed no difference in adverse reactions between ld IL-2 group and control group in treatment [1.06 (95% CI 0.86,1.31), p = 0.586, I2 = 53.8%], which is unreliable because the data is so small. CONCLUSIONS Ld IL-2 does increase the proportion of peripheral blood Treg cells in patients with rheumatism, and single and cumulative doses must be considered when using ld IL-2. In addition, more studies on the safety of ld IL-2 are urgently needed.
Collapse
Affiliation(s)
- Huanhuan Yan
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Huer Yan
- College of Basic Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lu Liu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women' Hospital/Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China.
| |
Collapse
|
8
|
Li ZG, He J, Miao M. Low-dose interleukin-2 therapy in autoimmune and inflammatory diseases: facts and hopes. Sci Bull (Beijing) 2023; 68:10-13. [PMID: 36621432 DOI: 10.1016/j.scib.2022.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Zhan-Guo Li
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism and Immune Diagnosis, Peking University People's Hospital, Beijing 100044, China; Peking-Tsinghua Center for Life Sciences, Beijing 100871, China.
| | - Jing He
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism and Immune Diagnosis, Peking University People's Hospital, Beijing 100044, China
| | - Miao Miao
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism and Immune Diagnosis, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
9
|
Connolly CM, Plomp L, Paik JJ, Allenbach Y. Possible future avenues for myositis therapeutics: DM, IMNM and IBM. Best Pract Res Clin Rheumatol 2022; 36:101762. [PMID: 35778272 DOI: 10.1016/j.berh.2022.101762] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Idiopathic inflammatory myopathies (IIMs) represent a heterogeneous group of systemic autoimmune diseases characterized by immune-mediated muscle injury. As insights into pathogenesis of IIM evolve, novel therapeutic strategies have become available to optimize outcomes. Herein, we summarize novel and emerging strategies in the management of dermatomyositis (DM), immunemediated necrotizing myopathy (IMNM), and inclusion body myositis (IBM).
Collapse
Affiliation(s)
- Caoilfhionn M Connolly
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lotta Plomp
- Department of Internal Medicine and Clinical Immunology, Pitié Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Julie J Paik
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Yves Allenbach
- Department of Internal Medicine and Clinical Immunology, Pitié Salpêtrière Hospital, Sorbonne University, Paris, France.
| |
Collapse
|
10
|
Tsuruta H, Mizuno-Kamiya M, Takahashi M, Ando M, Ikeno K, Ueno K, Takayama E, Kawaki H, Nakamura G, Nikaido T, Fujita H, Kondoh N. Enhanced production of IL-2 from anti-CD3 antibody-stimulated mouse spleen cells by artepillin C, a major component of Brazilian green propolis. J Oral Biosci 2022; 64:366-375. [PMID: 35667586 DOI: 10.1016/j.job.2022.05.007] [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: 05/06/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVES In this report, we attempt to clarify the immune modulatory effects of Brazilian green propolis (BGP) and its major component, artepillin C, on the cytokine production of anti-CD3 antibody-stimulated mouse spleen cells. We also estimate the physiological mechanism affecting artepillin C's upon the cells. METHODS Male C3H/HeN mouse spleen cells stimulated by antiCD3 monoclonal antibody were co-cultured with BGP, artepillin C, and HC030031, a transient receptor potential ankyrin 1 (TRPA1) Ca2+ channel antagonist. The synthesis of interferon (IFN)-γ, interleukin (IL)-6, IL-17, IL-4, IL-10, and IL-2 was assayed by enzyme-linked immunoassay. The expression of IL-2 mRNA and the protein product were examined by reverse transcription-quantitative polymerase chain reaction and Western blot analyses, respectively. RESULTS The production of IL-2 was markedly enhanced, while that of IL-4 and IL-10 was not significantly affected; by contrast, the production of IFN-γ, IL-6, and IL-17 was significantly reduced in the antibody-stimulated spleen cells treated with BGP at a non-cytostatic concentration. These effects were reproduced in the cells treated with artepillin C. The expression of IL-2 mRNA was unaffected; however, that of the protein was significantly enhanced in the artepillin C-treated cells compared to untreated control cells. The enhancement of protein expression and the production of IL-2 by artepillin C was significantly alleviated by adding HC030031. CONCLUSIONS Artepillin C is an important regulator of cytokine synthesis from activated spleen cells. The agent specifically augmented the expression of IL-2 via the Ca2+-permeable cation channel, TRPA1, at least in part, at the translational or secretion levels.
Collapse
Affiliation(s)
- Hanemi Tsuruta
- Department of Oral Biochemistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Japan; Department of Operative Dentistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Gifu 500-8471, Japan.
| | - Masako Mizuno-Kamiya
- Chemistry Laboratory, Department of Business Administration, Asahi University School of Business Administration, Mizuho, Gifu 501-0296, Japan.
| | - Moe Takahashi
- Department of Oral and Maxillofacial Surgery, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Japan.
| | - Megumi Ando
- Department of Oral Biochemistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Japan.
| | - Kumiko Ikeno
- AKITAYA HONTEN CO., LTD., R&D, Gifu 500-8471, Japan.
| | - Kyohei Ueno
- Department of Oral Biochemistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Japan.
| | - Eiji Takayama
- Department of Oral Biochemistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Japan.
| | - Harumi Kawaki
- Chemistry Laboratory, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Gifu 500-8471, Japan.
| | | | - Toru Nikaido
- Department of Operative Dentistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Gifu 500-8471, Japan.
| | - Hisakazu Fujita
- Department of Scientific and Linguistic Fundamentals for Nursing, Osaka City University Graduate School of Nursing, Osaka 558-8585, Japan.
| | - Nobuo Kondoh
- Department of Oral Biochemistry, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Japan.
| |
Collapse
|
11
|
Zhufeng Y, Xu J, Miao M, Wang Y, Li Y, Huang B, Guo Y, Tian J, Sun X, Li J, Lu D, Li Z, Li Y, He J. Modification of Intestinal Microbiota Dysbiosis by Low-Dose Interleukin-2 in Dermatomyositis: A Post Hoc Analysis From a Clinical Trial Study. Front Cell Infect Microbiol 2022; 12:757099. [PMID: 35360108 PMCID: PMC8964112 DOI: 10.3389/fcimb.2022.757099] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
The microbiota has been observed altered in autoimmune diseases, including idiopathic inflammatory myopathies (IIMs), and associated with different treatments. Low-dose IL-2 treatment emerges as a new option for active IIMs. This study aims to explore the role of low-dose IL-2 in regulating intestinal dysbiosis involved in the IIMs. In this study, 13 patients with active IIMs were enrolled and received 1 ×106 IU of IL-2 subcutaneously every other day for 12 weeks plus standard care. The clinical response and immune response were assessed. Stool samples were obtained to explore the structural and functional alterations of the fecal microbiota targeting the V3–V4 region of the 16S rRNA gene and analyze their associations with clinical and immunological characteristics. Our study demonstrated that diversity of microbiota decreased remarkably in patients with IIMs, compared to healthy controls. The inflammatory-related bacteria, such as Prevotellaceae increased, while some butyrate-producing bacteria, such as Pseudobutyrivibrio, Lachnospiraceae, Roseburia, and Blautia, decreased significantly. The alteration associated with disease activities in patients with IIMs. After low-dose IL-2 treatment, 92.31% (12/13) of patients achieved IMACS DOI at week 12. Proportion of Treg cells significantly increased at week 12 compared with that in baseline (15.9% [7.73, 19.4%] vs. 9.89% [6.02, 11.8%], P = 0.015). Interestingly, certain butyrate-producing bacteria increase significantly after IL-2 treatment, like Lachnospiraceae, Pseudobutyrivibrio, etc., and are associated with a rise in L-Asparagine and L-Leucine. The effects of low-dose IL-2 on gut microbiota were more apparent in NOD mice. Together, the data presented demonstrated that low-dose IL-2 was effective in active IIMs and highlighted the potential for modifying the intestinal microbiomes of dysbiosis to treat IIMs.
Collapse
Affiliation(s)
- Yunzhi Zhufeng
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
| | - Jun Xu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
| | - Miao Miao
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
| | - Yifan Wang
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
| | - Yimin Li
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
| | - Bo Huang
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
| | - Yixue Guo
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
| | - Jiayi Tian
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
| | - Jing Li
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
| | - Dan Lu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yuhui Li
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- *Correspondence: Yuhui Li, ; Jing He,
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- *Correspondence: Yuhui Li, ; Jing He,
| |
Collapse
|