1
|
Yan J, Guo S, He J, Huang H, Xu Y. Myeloid-derived suppressor cells in metabolic and cardiovascular disorders. Trends Endocrinol Metab 2025:S1043-2760(25)00024-4. [PMID: 40024876 DOI: 10.1016/j.tem.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
Dysregulation of immune homeostasis can precipitate chronic inflammation, thus significantly contributing to the onset and progression of metabolic and cardiovascular diseases. Myeloid-derived suppressor cells (MDSCs) constitute a heterogeneous population of immature myeloid cells that are mobilized in response to biological stressors such as tissue damage and inflammation. Although MDSCs have been extensively characterized in the contexts of cancer and infectious diseases, emerging evidence highlights their pivotal roles in the pathophysiology of metabolic and cardiovascular disorders. We discuss growing evidence for the involvement of MDSCs in the progression of metabolic and cardiovascular diseases, with the aim of deepening our understanding of MDSCs in cardiometabolic physiology and identifying the necessary steps for the development of innovative MDSC-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jingwei Yan
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Thoracic Surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Shuai Guo
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jun He
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China
| | - Hanpeng Huang
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Yiming Xu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Zhou Z, Zhan C, Li W, Luo W, Liu Y, He F, Tian Y, Lin Z, Song Z. Monocytic myeloid-derived suppressor cells contribute to the exacerbation of bone destruction in periodontitis. J Transl Med 2025; 23:217. [PMID: 39985072 PMCID: PMC11846281 DOI: 10.1186/s12967-025-06214-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/07/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Periodontitis (PD) is a chronic infectious and inflammatory disease characterized by alveolar bone loss. The distinctive activity of immune cells critically exacerbates bone resorption in PD. Myeloid-derived suppressor cells (MDSCs) are known to contribute to various chronic inflammatory conditions, but their role in the pathogenesis and progression of PD remains poorly understood. METHODS We used single-cell transcriptomic analysis with human gingival samples and animal models of experimental periodontitis to examine the role of M-MDSCs in PD. We also explored the therapeutic effect of depleting MDSCs on PD in vivo. Additionally, the mechanisms of long non-coding RNA Neat1 and the pathway of NF-κB-dependent "canonical NLRP3 inflammasome activation" in MDSCs were investigated in PD. RESULTS In this study, we revealed that monocytic (M)-MDSCs were significantly increased in inflamed gingiva of PD patients compared to healthy individuals. Expansion of M-MDSCs was also observed in the mouse model of ligature-induced periodontitis, and depletion of MDSCs in PD mice could ameliorate alveolar bone loss and reduce periodontal inflammation. Mechanistically, we found that long non-coding RNA Neat1 was significantly upregulated in M-MDSCs, which achieved this proinflammatory effect by activating NF-κB signaling in PD. Furthermore, the pathway of NF-κB-dependent "canonical NLRP3 inflammasome activation" was confirmed in the PD mouse model, accompanied by increased secretion of proinflammatory cytokines that drive alveolar bone loss, including IL-1β, IL-6 and TNF-α. CONCLUSIONS In conclusion, this study highlights the pivotal proinflammatory role of M-MDSCs in PD and suggests that targeting these cells may represent a novel immunotherapeutic approach. Future research could focus on strategies to specifically target MDSCs for the treatment of periodontitis.
Collapse
Affiliation(s)
- Zhaocai Zhou
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Chi Zhan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Wenchuan Li
- Guangzhou First People's Hospital, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Wenji Luo
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Yufeng Liu
- Guangzhou First People's Hospital, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Feng He
- Guangzhou First People's Hospital, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Yaguang Tian
- Department of Stomatology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China.
| | - Zhengmei Lin
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China.
| | - Zhi Song
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China.
| |
Collapse
|
3
|
Wang Q, Xiong X, Chen L, Zhu F, Yang X, Zhao W, He S, Zuo J, Lin Z. Periplosides Extract from Cortex periplocae Improve Collagen Antibody-Induced Arthritis by Regulating Macrophage Polarization. Curr Issues Mol Biol 2024; 46:14095-14105. [PMID: 39727971 DOI: 10.3390/cimb46120843] [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: 10/17/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 12/28/2024] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease characterized primarily by the synovial infiltration of inflammatory cells. Macrophage infiltration in the joint synovium is one of the early hallmarks of RA disease activity. Cortex periplocae, which has been widely employed in traditional Chinese medicine (TCM) to alleviate RA, harbors a bioactive compound known as Periploca sepium periplosides (PePs). In this study, collagen antibody-induced arthritis (CAIA) was established in mice through the administration of collagen antibodies and lipopolysaccharide (LPS), followed by treatment with PePs. The therapeutic effects of PePs were evaluated by measuring paw thickness, clinical arthritis scores, and histological changes in joint tissues. Flow cytometry and qRT-PCR were used to assess macrophage polarization in vivo and in vitro. The findings indicate that PePs effectively attenuated CAIA by suppressing the polarization of RAW264.7 cells towards the M1 phenotype while promoting their polarization towards the M2 phenotype. These results provide valuable insights into the scientific significance of PePs as a potential therapeutic agent for RA.
Collapse
Affiliation(s)
- Que Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaoyu Xiong
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Li Chen
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fenghua Zhu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoqian Yang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Weimin Zhao
- Department of Natural Product Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shijun He
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianping Zuo
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zemin Lin
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
4
|
Lin L, Luo J, Cai Y, Wu X, Zhou L, Li T, Wang X, Xu H. Mass cytometry identifies imbalance of multiple immune-cell subsets associated with biologics treatment in ankylosing spondylitis. Int J Rheum Dis 2024; 27:e15378. [PMID: 39420773 DOI: 10.1111/1756-185x.15378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE This study aims to comprehensively investigate immune-cell landscapes in ankylosing spondylitis (AS) patients and explore longitudinal immunophenotyping changes induced by biological agents. METHODS We employed mass cytometry with 35 cellular markers to analyze blood samples from 34 AS patients and 13 healthy controls (HC). Eleven AS patients were re-evaluated 1 month (4 patients) and 3 months (7 patients) after treatment with biological agents. Flow Self-Organizing Maps (FlowSOM) clustering was performed to identify specific cellular metaclusters. We compared cellular abundances across distinct subgroups and validated subset differences using gating strategies in flow cytometry scatter plots, visualized with FlowJo software. The proportions of differential subsets were then used for intercellular and clinical correlation analysis, as well as for constructing diagnostic models based on the random forest algorithm. RESULTS In AS patients, we identified and validated nine different immune-cell subsets compared to HC. Three subsets increased: helper T-cell 17 (Th17), mucosa-associated invariant T-cell (MAIT), and classical monocytes (CM). Six subsets decreased: effector memory T-cell (TEM), naïve B cells, transitional B cells, IL10+ memory B cells, non-classical monocytes (NCM), and neutrophils. Treatments with biological agents could rectify cellular abnormalities, particularly the imbalance of CM/NCM. Furthermore, these subsets may serve as biomarkers for assessing disease activity and constructing effective diagnostic models for AS. CONCLUSION These findings provide novel insights into the specific patterns of immune cell in AS, facilitating the further development of novel biomarkers and potential therapeutic targets for AS patients.
Collapse
Affiliation(s)
- Li Lin
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Jing Luo
- School of Medicine, Tsinghua University, Beijing, China
| | - Yue Cai
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Ling Zhou
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Ting Li
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Xiaobing Wang
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
- School of Medicine, Tsinghua University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
5
|
Nepal MR, Shah S, Kang KT. Dual roles of myeloid-derived suppressor cells in various diseases: a review. Arch Pharm Res 2024; 47:597-616. [PMID: 39008186 DOI: 10.1007/s12272-024-01504-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that originate from bone marrow stem cells. In pathological conditions, such as autoimmune disorders, allergies, infections, and cancer, normal myelopoiesis is altered to facilitate the formation of MDSCs. MDSCs were first shown to promote cancer initiation and progression by immunosuppression with the assistance of various chemokines and cytokines. Recently, various studies have demonstrated that MDSCs play two distinct roles depending on the physiological and pathological conditions. MDSCs have protective roles in autoimmune disorders (such as uveoretinitis, multiple sclerosis, rheumatoid arthritis, ankylosing spondylitis, type 1 diabetes, autoimmune hepatitis, inflammatory bowel disease, alopecia areata, and systemic lupus erythematosus), allergies, and organ transplantation. However, they play negative roles in infections and various cancers. Several immunosuppressive functions and mechanisms of MDSCs have been determined in different disease conditions. This review comprehensively discusses the associations between MDSCs and various pathological conditions and briefly describes therapeutic approaches.
Collapse
Affiliation(s)
- Mahesh Raj Nepal
- College of Pharmacy, Duksung Women's University, Seoul, South Korea
- Duksung Innovative Drug Center, Duksung Women's University, Seoul, South Korea
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Sajita Shah
- College of Pharmacy, Duksung Women's University, Seoul, South Korea
- Duksung Innovative Drug Center, Duksung Women's University, Seoul, South Korea
- The Comprehensive Cancer Center, Department of Radiation Oncology, Ohio State University, Columbus, OH, USA
| | - Kyu-Tae Kang
- College of Pharmacy, Duksung Women's University, Seoul, South Korea.
- Duksung Innovative Drug Center, Duksung Women's University, Seoul, South Korea.
| |
Collapse
|
6
|
Yu B, Wang Y, Bing T, Tang Y, Huang J, Xiao H, Liu C, Yu Y. Platinum Prodrug Nanoparticles with COX-2 Inhibition Amplify Pyroptosis for Enhanced Chemotherapy and Immune Activation of Pancreatic Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310456. [PMID: 38092007 DOI: 10.1002/adma.202310456] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/26/2023] [Indexed: 12/22/2023]
Abstract
Pyroptosis, an emerging mechanism of programmed cell death, holds great potential to trigger a robust antitumor immune response. Platinum-based chemotherapeutic agents can induce pyroptosis via caspase-3 activation. However, these agents also enhance cyclooxygenase-2 (COX-2) expression in tumor tissues, leading to drug resistance and immune evasion in pancreatic cancer and significantly limiting the effectiveness of chemotherapy-induced pyroptosis. Here, an amphiphilic polymer (denoted as PHDT-Pt-In) containing both indomethacin (In, a COX-2 inhibitor) and platinum(IV) prodrug (Pt(IV)) is developed, which is responsive to glutathione (GSH). This polymer self-assemble into nanoparticles (denoted as Pt-In NP) that can disintegrate in cancer cells due to the GSH responsiveness, releasing In to inhibit the COX-2 expression, hence overcoming the chemoresistance and amplifying cisplatin-induced pyroptosis. In a pancreatic cancer mouse model, Pt-In NP significantly inhibit tumor growth and elicit both innate and adaptive immune responses. Moreover, when combined with anti-programmed death ligand (α-PD-L1) treatment, Pt-In NP demonstrate the ability to completely suppress metastatic tumors, transforming "cold tumors" into "hot tumors". Overall, the sustained release of Pt(IV) and In from Pt-In NP amplifies platinum-drug-induced pyroptosis to elicit long-term immune responses, hence presenting a generalizable strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Bingzheng Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yushu Wang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Tiejun Bing
- Immunology and Oncology center, ICE Bioscience, Beijing, 100176, China
| | - Yujing Tang
- SINOPEC (Beijing) Research Institute of Chemical Industry Co., Ltd, Beijing, 100013, China
| | - Jia Huang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, China
| | - Chaoyong Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
7
|
Zhao Z, Huang H, Ke S, Deng B, Wang YX, Xu N, Peng A, Han G, Liang E, He X, He Q, Ke PF, Huang XZ, He M. Triptolide inhibits the proinflammatory potential of myeloid-derived suppressor cells via reducing Arginase-1 in rheumatoid arthritis. Int Immunopharmacol 2024; 127:111345. [PMID: 38086266 DOI: 10.1016/j.intimp.2023.111345] [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: 11/25/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024]
Abstract
Triptolide (TPT) is widely used in the treatment of rheumatoid arthritis (RA). However, its regulatory mechanisms are not fully understood. This study demonstrated that Myeloid-derived suppressor cells (MDSCs) were expanded in both RA patients and arthritic mice. The frequency of MDSCs was correlated with RA disease severity and T helper 17 (Th17) responses. MDSCs from RA patients promoted the polarization of Th17 cells in vitro, which could be substantially attenuated by blocking arginase-1 (Arg-1). TPT inhibited the differentiation of MDSCs, particularly the monocytic MDSCs (M-MDSCs) subsets, as well as the expression of Arg-1 in a dose dependent manner. Alongside, TPT treatment reduced the potential of MDSCs to promote the polarization of IL-17+ T cell in vitro. Consistently, TPT immunotherapy alleviated adjuvant-induced arthritis (AIA) in a mice model, and reduced the frequency of MDSCs, M-MDSCs and IL-17+ T cells simultaneously. The presented data suggest a pathogenic role of MDSCs in RA and may function as a novel and effective therapeutic target for TPT in RA.
Collapse
Affiliation(s)
- Ziling Zhao
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huijie Huang
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sikai Ke
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bishun Deng
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun-Xiu Wang
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ning Xu
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Anping Peng
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guang Han
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Enyu Liang
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong He
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospitals of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qinglian He
- Department of Pathology, the Second Affiliated Hospitals of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pei-Feng Ke
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xian-Zhang Huang
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospitals of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Min He
- Department of Laboratory Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospitals of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|